1
|
Simats A, Sager HB, Liesz A. Heart-brain axis in health and disease: role of innate and adaptive immunity. Cardiovasc Res 2025; 120:2325-2335. [PMID: 39180327 DOI: 10.1093/cvr/cvae185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/24/2024] [Accepted: 06/13/2024] [Indexed: 08/26/2024] Open
Abstract
The importance of the brain-heart interaction has been increasingly recognized as a critical physiological axis that is altered in disease. In this review, we explore the intricate relationship between the central nervous system and cardiovascular health, focusing particularly on immunological mechanisms that influence the course of both neurological and cardiovascular diseases. While previous studies have established a key role of the autonomic nervous system (ANS) in linking brain and the heart, more recent studies have expanded our understanding of the multifaceted inter-organ interactions. As such, circulating mediators include immune cells of the adaptive and innate immune system and their secreted immunogenic factors have come into the focus as mediators along this bidirectional communication. Hence, in this review we briefly discuss the contribution of the ANS and then focus on innate and adaptive immune mechanisms along the heart-to-brain and brain-to-heart axes, illustrating how cardiovascular diseases affect cognitive functions and how brain pathologies lead to cardiac complications.
Collapse
Affiliation(s)
- Alba Simats
- Department of Neuroscience and Experimental Therapeutics, Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), c/Rosselló 161, 08036 Barcelona, Spain
| | - Hendrik B Sager
- DZHK (German Center for Cardiovascular Research), Partner site Munich Heart Alliance, Munich, Germany
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany
| | - Arthur Liesz
- Institute for Stroke and Dementia Research (ISD), University Medical Center Munich, Feodor-Lynen-Straße 17, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Feodor-Lynen-Str. 17, 81377 Munich, Germany
| |
Collapse
|
2
|
Yue X, Cui J, Huang S, Liu W, Qi J, He K, Li T. An interpretable radiomics-based machine learning model for predicting reverse left ventricular remodeling in STEMI patients using late gadolinium enhancement of myocardial scar. Eur Radiol 2025:10.1007/s00330-025-11419-1. [PMID: 40180637 DOI: 10.1007/s00330-025-11419-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 11/11/2024] [Accepted: 12/23/2024] [Indexed: 04/05/2025]
Abstract
OBJECTIVES To evaluate the added value of the late gadolinium enhancement (LGE)-scar radiomics features in predicting reverse left ventricular remodeling (r-LVR) in ST-segment elevation myocardial infarction (STEMI) patients using machine learning (ML). MATERIALS AND METHODS This retrospective study included 105 STEMI patients who underwent CMR within 7 days and 5 months post-percutaneous coronary intervention (PCI) on 1.5-T or 3.0-T MRI scanners (January 2014-2023). Radiomics features from LGE scar images and routine CMR markers were analyzed using a LightGBM model enhanced by Shapley Additive exPlanations (SHAP) for interpretability. Patients were divided into training (80) and test (25) sets. Three predictive models were developed: traditional CMR, LGE-scar radiomics, and a combined model integrating both. Model performance was assessed using ROC curves and AUC analysis. RESULTS In the training set, the traditional CMR model achieved an AUC of 0.745 (95% CI: 0.62-0.86), the LGE-scar radiomics model had an AUC of 0.712 (95% CI: 0.58-0.83), and the combined model showed the highest AUC of 0.754 (95% CI: 0.63-0.86). In the test set, the traditional CMR model's AUC decreased to 0.656 (95% CI: 0.42-0.88), while the LGE-scar radiomics model improved to 0.818 (95% CI: 0.59-1.00). The combined model achieved the highest AUC of 0.890 (95% CI: 0.75-1.00). SHAP analysis highlighted significant predictors such as infarct percentage of LV mass and wavelet-transformed texture features. CONCLUSION Integrating LGE scar radiomics features with traditional CMR parameters in a LightGBM model enhances predictive accuracy for r-LVR in STEMI patients, potentially improving patient stratification and treatment personalization. KEY POINTS Question Predicting r-LVR in STEMI patients remains challenging due to limitations in current imaging approaches. Findings Integrating LGE-scar radiomics and cardiac magnetic resonance markers in the LightGBM model significantly improves prediction accuracy for r-LVR. Clinical relevance This interpretable ML model enhances r-LVR prediction, supporting patient stratification and optimizing treatment strategies to improve patient outcomes.
Collapse
Affiliation(s)
- Xiuzheng Yue
- Medical Big Data Research Center, Medical Innovation Research Division of PLA General Hospital, Beijing, China
- Chinese PLA Medical School, Chinese PLA General Hospital, Beijing, China
| | - Jianing Cui
- Department of Radiology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | | | - Wenjia Liu
- Department of Radiology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jing Qi
- Medical Big Data Research Center, Medical Innovation Research Division of PLA General Hospital, Beijing, China
- Chinese PLA Medical School, Chinese PLA General Hospital, Beijing, China
| | - Kunlun He
- Medical Big Data Research Center, Medical Innovation Research Division of PLA General Hospital, Beijing, China.
| | - Tao Li
- Department of Radiology, The First Medical Center, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
3
|
Bahrami P, Aromolaran KA, Aromolaran AS. Mechanistic Relevance of Ventricular Arrhythmias in Heart Failure with Preserved Ejection Fraction. Int J Mol Sci 2024; 25:13423. [PMID: 39769189 PMCID: PMC11677834 DOI: 10.3390/ijms252413423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/05/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is increasing at an alarming rate worldwide, with limited effective therapeutic interventions in patients. Sudden cardiac death (SCD) and ventricular arrhythmias present substantial risks for the prognosis of these patients. Obesity is a risk factor for HFpEF and life-threatening arrhythmias. Obesity and its associated metabolic dysregulation, leading to metabolic syndrome, are an epidemic that poses a significant public health problem. More than one-third of the world population is overweight or obese, leading to an enhanced risk of incidence and mortality due to cardiovascular disease (CVD). Obesity predisposes patients to atrial fibrillation and ventricular and supraventricular arrhythmias-conditions that are caused by dysfunction in the electrical activity of the heart. To date, current therapeutic options for the cardiomyopathy of obesity are limited, suggesting that there is considerable room for the development of therapeutic interventions with novel mechanisms of action that will help normalize sinus rhythms in obese patients. Emerging candidates for modulation by obesity are cardiac ion channels and Ca-handling proteins. However, the underlying molecular mechanisms of the impact of obesity on these channels and Ca-handling proteins remain incompletely understood. Obesity is marked by the accumulation of adipose tissue, which is associated with a variety of adverse adaptations, including dyslipidemia (or abnormal systemic levels of free fatty acids), increased secretion of proinflammatory cytokines, fibrosis, hyperglycemia, and insulin resistance, which cause electrical remodeling and, thus, predispose patients to arrhythmias. Furthermore, adipose tissue is also associated with the accumulation of subcutaneous and visceral fat, which is marked by distinct signaling mechanisms. Thus, there may also be functional differences in the effects of the regional distribution of fat deposits on ion channel/Ca-handling protein expression. Evaluating alterations in their functional expression in obesity will lead to progress in the knowledge of the mechanisms responsible for obesity-related arrhythmias. These advances are likely to reveal new targets for pharmacological modulation. Understanding how obesity and related mechanisms lead to cardiac electrical remodeling is likely to have a significant medical and economic impact. Nevertheless, substantial knowledge gaps remain regarding HFpEF treatment, requiring further investigations to identify potential therapeutic targets. The objective of this study is to review cardiac ion channel/Ca-handling protein remodeling in the predisposition to metabolic HFpEF and arrhythmias. This review further highlights interleukin-6 (IL-6) as a potential target, cardiac bridging integrator 1 (cBIN1) as a promising gene therapy agent, and leukotriene B4 (LTB4) as an underappreciated pathway in future HFpEF management.
Collapse
Affiliation(s)
- Pegah Bahrami
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, 95 S 2000 E, Salt Lake City, UT 84112, USA; (P.B.); (K.A.A.)
| | - Kelly A. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, 95 S 2000 E, Salt Lake City, UT 84112, USA; (P.B.); (K.A.A.)
| | - Ademuyiwa S. Aromolaran
- Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), University of Utah School of Medicine, 95 S 2000 E, Salt Lake City, UT 84112, USA; (P.B.); (K.A.A.)
- Department of Surgery, Division of Cardiothoracic Surgery, Nutrition & Integrative Physiology, Biochemistry & Molecular Medicine Program, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| |
Collapse
|
4
|
McCabe JJ, Walsh C, Gorey S, Arnold M, DeMarchis GM, Harris K, Hervella P, Iglesias-Rey R, Jern C, Katan M, Li L, Miyamoto N, Montaner J, Purroy F, Rothwell PM, Stanne TM, Sudlow C, Ueno Y, Vicente-Pascual M, Whiteley W, Woodward M, Kelly PJ. Interleukin-6, C-Reactive Protein, and Recurrence After Stroke: A Time-Course Analysis of Individual-Participant Data. Stroke 2024; 55:2825-2834. [PMID: 39479747 DOI: 10.1161/strokeaha.124.047820] [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: 05/28/2024] [Revised: 09/12/2024] [Accepted: 09/24/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Inflammation promotes atherogenesis. Randomized controlled trials of anti-inflammatory therapies for prevention after stroke have not yet demonstrated clear benefit. IL-6 (interleukin-6) and hsCRP (high-sensitivity C-reactive protein) are independently associated with major adverse cardiovascular events poststroke and may guide patient selection in future randomized controlled trials. Optimal timing of hsCRP/IL-6 measurement poststroke is unknown, as early blood levels may be confounded by the inflammatory response to brain infarction. METHODS Using individual-participant data from a systematic review, we performed a time-course analysis to investigate the association between hsCRP/IL-6 and recurrent events stratified by timing of sampling. The prespecified coprimary end points after sample measurement were: (1) recurrent major adverse cardiovascular events (first major coronary event, recurrent stroke, or vascular death) and (2) recurrent stroke (ischemic, hemorrhagic, or unspecified). The poststroke dynamics of IL-6/hsCRP were analyzed by plotting their median (interquartile interval) concentrations within each tenth of the sampling timeframe. Acute/postacute phases were defined for each biomarker according to the shape of this relationship. RESULTS There were data for 9798 patients from 11 studies (19 891 person-years follow-up, 10 observational cohorts, and 1 randomized trial). Each marker was measured once. IL-6 was markedly elevated <24 hours poststroke compared with postacute levels (≥24 hours; 11.6 versus 3.02 pg/mL; P<0.001). HsCRP was elevated for 10 days. IL-6 was associated with recurrent major adverse cardiovascular events in the postacute phase (≥24 hours; risk ratio, 1.30 [CI, 1.19-1.41], per unit logeIL-6), but not in the acute phase (<24 hours; risk ratio, 1.10 [CI, 0.98-1.25]; Pinteraction=0.03). After adjustment for risk factors/medication, the association remained for postacute IL-6 when analyzed per logeunit (risk ratio, 1.16 [CI, 1.05-1.66]) and per quarter increase (risk ratio, 1.55 [CI, 1.19-2.02]; Q4 versus Q1), but not if measured acutely. Similar findings were observed for recurrent stroke. There was no evidence of time-dependent interaction with hsCRP. CONCLUSIONS Timing of sample measurement after stroke modifies the association with recurrent major adverse cardiovascular events for IL-6 but not hsCRP. These data inform future randomized controlled trial designs incorporating biomarker-based selection of patients for anti-inflammatory therapies.
Collapse
Affiliation(s)
- John J McCabe
- Health Research Board Stroke Clinical Trials Network Ireland, Dublin (J.J.M., C.W., S.G., P.J.K.)
- School of Medicine, University College Dublin, Ireland (J.J.M., S.G., P.J.K.)
- Stroke Service, Mater Misericordiae University Hospital, Dublin, Ireland (J.J.M., S.G., P.J.K.)
| | - Cathal Walsh
- Health Research Board Stroke Clinical Trials Network Ireland, Dublin (J.J.M., C.W., S.G., P.J.K.)
- Department of Biostatistics, Trinity College Dublin, Ireland (C.W.)
| | - Sarah Gorey
- Health Research Board Stroke Clinical Trials Network Ireland, Dublin (J.J.M., C.W., S.G., P.J.K.)
- School of Medicine, University College Dublin, Ireland (J.J.M., S.G., P.J.K.)
- Stroke Service, Mater Misericordiae University Hospital, Dublin, Ireland (J.J.M., S.G., P.J.K.)
| | - Markus Arnold
- Neurology Department, University Hospital of Zurich, Switzerland (M.A., M.K.)
| | | | - Katie Harris
- The George Institute for Global Health, University of New South Wales, Sydney, Australia (K.H., M.W.)
| | - Pablo Hervella
- Neuroimaging and Biotechnology Laboratory, Health Research Institute of Santiago de Compostela, Spain (P.H., R.I.-R.)
| | - Ramon Iglesias-Rey
- Neuroimaging and Biotechnology Laboratory, Health Research Institute of Santiago de Compostela, Spain (P.H., R.I.-R.)
| | - Christina Jern
- Laboratory Medicine Department, Institute of Biomedicine, the Sahlgrenska Academy, University of Gothenburg, Sweden (C.J., T.M.S.)
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Gothenburg, Sweden (C.J.)
| | - Mira Katan
- Neurology Department, University Hospital of Zurich, Switzerland (M.A., M.K.)
- Neurology Department, University Hospital Basel, Switzerland (G.M.D., M.K.)
| | - Linxin Li
- Wolfson Centre for the Prevention of Stroke and Dementia (L.L., P.M.R.), University of Oxford, United Kingdom
| | - Nobukazu Miyamoto
- Neurology Department, Juntendo University School of Medicine, Tokyo, Japan (N.M., Y.U.)
| | - Joan Montaner
- Neurology Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain (J.M.)
- Institute de Biomedicine of Seville, IBiS/Hospital Universitario Virgen del Rocío/CSIC/University of Seville, Spain (J.M.)
- Virgen Macarena Hospital, Neurology, Sevilla, Spain (J.M.)
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research, Universitat Autònoma de Barcelona, Spain (J.M.)
| | - Francisco Purroy
- Department of Neurology, Hospital Universitari Arnau de Vilanova, Lleida, Spain (F.P., M.V.-P.)
- Department of Clinical Neurosciences, University of Lleida, Spain (F.P., M.V.-P.)
| | - Peter M Rothwell
- Wolfson Centre for the Prevention of Stroke and Dementia (L.L., P.M.R.), University of Oxford, United Kingdom
| | - Tara M Stanne
- Laboratory Medicine Department, Institute of Biomedicine, the Sahlgrenska Academy, University of Gothenburg, Sweden (C.J., T.M.S.)
| | - Catherine Sudlow
- Centre for Medical Informatics, Usher Institute of Population Health Sciences and Informatics (C.S.), University of Edinburgh, United Kingdom
- Centre for Clinical Brain Sciences (C.S., W.W.), University of Edinburgh, United Kingdom
| | - Yuji Ueno
- Neurology Department, Juntendo University School of Medicine, Tokyo, Japan (N.M., Y.U.)
| | - Mikel Vicente-Pascual
- Department of Neurology, Hospital Universitari Arnau de Vilanova, Lleida, Spain (F.P., M.V.-P.)
- Department of Clinical Neurosciences, University of Lleida, Spain (F.P., M.V.-P.)
| | - William Whiteley
- Nuffield Department of Population Health (W.W.), University of Oxford, United Kingdom
- Centre for Clinical Brain Sciences (C.S., W.W.), University of Edinburgh, United Kingdom
| | - Mark Woodward
- The George Institute for Global Health, University of New South Wales, Sydney, Australia (K.H., M.W.)
- The George Institute for Global Health, School of Public Health, Imperial College London, United Kingdom (M.W.)
| | - Peter J Kelly
- Health Research Board Stroke Clinical Trials Network Ireland, Dublin (J.J.M., C.W., S.G., P.J.K.)
- School of Medicine, University College Dublin, Ireland (J.J.M., S.G., P.J.K.)
- Stroke Service, Mater Misericordiae University Hospital, Dublin, Ireland (J.J.M., S.G., P.J.K.)
| |
Collapse
|
5
|
Lukovic D, Gyöngyösi M, Pavo IJ, Mester-Tonczar J, Einzinger P, Zlabinger K, Kastner N, Spannbauer A, Traxler D, Pavo N, Goliasch G, Pils D, Jakab A, Szankai Z, Michel-Behnke I, Zhang L, Devaux Y, Graf S, Beitzke D, Winkler J. Increased [ 18F]FDG uptake in the infarcted myocardial area displayed by combined PET/CMR correlates with snRNA-seq-detected inflammatory cell invasion. Basic Res Cardiol 2024; 119:807-829. [PMID: 38922408 PMCID: PMC11461641 DOI: 10.1007/s00395-024-01064-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024]
Abstract
Combined [18F]FDG PET-cardiac MRI imaging (PET/CMR) is a useful tool to assess myocardial viability and cardiac function in patients with acute myocardial infarction (AMI). Here, we evaluated the prognostic value of PET/CMR in a porcine closed-chest reperfused AMI (rAMI) model. Late gadolinium enhancement by PET/CMR imaging displayed tracer uptake defect at the infarction site by 3 days after the rAMI in the majority of the animals (group Match, n = 28). Increased [18F]FDG uptake at the infarcted area (metabolism/contractility mismatch) with reduced tracer uptake in the remote viable myocardium (group Mismatch, n = 12) 3 days after rAMI was observed in the animals with larger infarct size and worse left ventricular ejection fraction (LVEF) (34 ± 8.7 vs 42.0 ± 5.2%), with lower LVEF also at the 1-month follow-up (35.8 ± 9.5 vs 43.0 ± 6.3%). Transcriptome analyses by bulk and single-nuclei RNA sequencing of the infarcted myocardium and border zones (n = 3 of each group, and 3 sham-operated controls) revealed a strong inflammatory response with infiltration of monocytes and macrophages in the infarcted and border areas in Mismatch animals. Our data indicate a high prognostic relevance of combined PET/MRI in the subacute phase of rAMI for subsequent impairment of heart function and underline the adverse effects of an excessive activation of the innate immune system in the initial phase after rAMI.
Collapse
Affiliation(s)
- Dominika Lukovic
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Mariann Gyöngyösi
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria.
| | - Imre J Pavo
- Division of Pediatric Cardiology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Julia Mester-Tonczar
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Patrick Einzinger
- Institute of Information Systems Engineering, Research Unit of Information and Software Engineering, Vienna University of Technology, 1040, Vienna, Austria
| | - Katrin Zlabinger
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Nina Kastner
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Andreas Spannbauer
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Denise Traxler
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Noemi Pavo
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Georg Goliasch
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Dietmar Pils
- Division of General Surgery, Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Andras Jakab
- Center for MR-Research, University Children's Hospital Zurich, Zurich, Switzerland
| | | | - Ina Michel-Behnke
- Division of Pediatric Cardiology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Lu Zhang
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Senta Graf
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Dietrich Beitzke
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Johannes Winkler
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
6
|
Cederström S, Jernberg T, Samnegård A, Johansson F, Silveira A, Tornvall P, Lundman P. Inflammatory biomarkers and long-term outcome in young patients three months after a first myocardial infarction. Cytokine 2024; 182:156696. [PMID: 39059290 DOI: 10.1016/j.cyto.2024.156696] [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: 04/29/2024] [Revised: 06/18/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Studies on predictive value of circulating inflammatory biomarkers after myocardial infarction (MI) have often been limited by blood sampling only in an acute setting and short follow-up time. We aimed to compare the long-term predictive value of nine inflammatory biomarkers, known to be involved in atherosclerosis, in young patients investigated three months after a first-time MI. METHODS Nine biomarkers (high-sensitivity C-reactive protein, interleukin (IL)-6, IL-18, monocyte chemoattractant protein-1, matrix metalloproteinase (MMP)-1, MMP-3, MMP-9, serum amyloid A and tumor necrosis factor-alfa) were sampled in 382 young (<60 years) patients and in age and sex-matched controls, three months after a first-time MI between 1996 and 2000. Swedish national patient registers were used to determine cardiovascular (CV) outcomes during 20 years of follow-up. RESULTS In cases, random forest models identified IL-6 as the most important predictor of the primary composite endpoint of death, heart failure (HF) or MI hospitalization, and the separate endpoints death and HF hospitalization. IL-18 was the most important predictor of MI hospitalization. In a Cox regression, the highest tertile of IL-6 was associated with the composite endpoint (HR (95% CI) 1.91 (1.31-2.79)), death (2.38 (1.42-3.98)) and HF hospitalization (2.70 (1.32-5.50)), when adjusting for age, sex and CV risk factors. The highest tertile of IL-18 was associated with MI hospitalization (2.31 (1.08-4.91)) when severity of coronary atherosclerosis was added to the same type of model. CONCLUSIONS When nine inflammatory markers involved in atherosclerosis were analyzed three months after the acute event in young MI patients, IL-6 and IL-18 were the most important biomarkers to predict long-term CV outcomes during 20 years of follow-up.
Collapse
Affiliation(s)
- Sofia Cederström
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Sweden.
| | - Tomas Jernberg
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Sweden
| | - Ann Samnegård
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Sweden
| | - Fredrik Johansson
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Sweden
| | - Angela Silveira
- Department of Medicine Solna K2, Karolinska Institutet and Karolinska University Hospital Solna
| | - Per Tornvall
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Pia Lundman
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Sweden
| |
Collapse
|
7
|
崔 佳, 刘 文, 闫 非, 赵 亚, 陈 伟, 罗 春, 张 兴, 李 涛. [Predictive value of cardiac magnetic resonance imaging for adverse left ventricular remodeling after acute ST-segment elevation myocardial infarction]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:553-562. [PMID: 38597447 PMCID: PMC11006702 DOI: 10.12122/j.issn.1673-4254.2024.03.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Indexed: 04/11/2024]
Abstract
OBJECTIVE To assess the value of cardiac magnetic resonance (CMR) imaging for predicting adverse left ventricular remodeling in patients with ST-segment elevation myocardial infarction (STEMI). METHODS We retrospectively analyzed the clinical data and serial CMR (cine and LGE sequences) images of 86 STEMI patients within 1 week and 5 months after percutaneous coronary intervention (PCI), including 25 patients with adverse LV remodeling and 61 without adverse LV remodeling, defined as an increase of left ventricular end-systolic volume (LVESV) over 15% at the second CMR compared to the initial CMR. The CMR images were analyzed for LV volume, infarct characteristics, and global and infarct zone myocardial function. The independent predictors of adverse LV remodeling following STEMI were analyzed using univariate and multivariate Logistic regression methods. RESULTS The initial CMR showed no significant differences in LV volume or LV ejection fraction (LVEF) between the two groups, but the infarct mass and microvascular obstructive (MVO) mass were significantly greater in adverse LV remodeling group (P < 0.05). Myocardial injury and cardiac function of the patients recovered over time in both groups. At the second CMR, the patients with adverse LV remodeling showed a significantly lower LVEF, a larger left ventricular end-systolic volume index (LVESVI) and a greater extent of infarct mass (P < 0.001) with lower global peak strains and strain rates in the radial, circumferential, and longitudinal directions (P < 0.05), infarct zone peak strains in the 3 directions, and infarct zone peak radial and circumferential strain rates (P < 0.05). The independent predictors for adverse LV remodeling following STEMI included the extent of infarct mass (AUC=0.793, 95% CI: 0.693-0.873; cut-off value: 30.67%), radial diastolic peak strain rate (AUC=0.645, 95% CI: 0.534-0.745; cut-off value: 0.58%), and RAAS inhibitor (AUC= 0.699, 95% CI: 0.590-0.793). CONCLUSION The extent of infarct mass, peak radial diastolic strain rate, and RAAS inhibitor are independent predictors of adverse LV remodeling following STEMI.
Collapse
Affiliation(s)
- 佳宁 崔
- 中国人民解放军总医院第一医学中心放射诊断科,北京 100853Department of Radiology, First Medical Center, PLA General Hospital, Beijing 100853, China
- 首都医科大学附属北京积水潭医院放射科,北京 100035Department of Radiology, Beijing Jishuitan Hospital, Capital Medical University, Beijing 100035, China
| | - 文佳 刘
- 中国人民解放军总医院第一医学中心放射诊断科,北京 100853Department of Radiology, First Medical Center, PLA General Hospital, Beijing 100853, China
| | - 非 闫
- 中国人民解放军总医院第一医学中心放射诊断科,北京 100853Department of Radiology, First Medical Center, PLA General Hospital, Beijing 100853, China
| | - 亚男 赵
- 中国人民解放军总医院第一医学中心放射诊断科,北京 100853Department of Radiology, First Medical Center, PLA General Hospital, Beijing 100853, China
| | - 伟杰 陈
- 中国人民解放军联勤保障部队第九八五医院放射科,山西 太原 030001Department of Radiology, 985th Hospital of Joint Logistics Support Force, Taiyuan 030001, China
| | - 春材 罗
- 中国人民解放军总医院第一医学中心放射诊断科,北京 100853Department of Radiology, First Medical Center, PLA General Hospital, Beijing 100853, China
| | - 兴华 张
- 中国人民解放军总医院第一医学中心放射诊断科,北京 100853Department of Radiology, First Medical Center, PLA General Hospital, Beijing 100853, China
| | - 涛 李
- 中国人民解放军总医院第一医学中心放射诊断科,北京 100853Department of Radiology, First Medical Center, PLA General Hospital, Beijing 100853, China
| |
Collapse
|
8
|
Hu Y, Li Q, Wang Y. Serum ACSL4 levels in patients with ST-segment elevation myocardial infarction (STEMI) and its association with one-year major adverse cardiovascular events (MACE): A prospective cohort study. Medicine (Baltimore) 2024; 103:e36870. [PMID: 38215103 PMCID: PMC10783377 DOI: 10.1097/md.0000000000036870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 12/15/2023] [Indexed: 01/14/2024] Open
Abstract
In the present prospective cohort research, we aimed to explore the serum levels of Acyl-CoA synthetase long-chain family member 4 (ACSL4) in patients with ST-segment elevation myocardial infarction (STEMI) and its association with 1-year major adverse cardiovascular events (MACE). This prospective cohort study recruited 507 patients who underwent percutaneous coronary intervention for the treatment of STEMI at our hospital during August 2019 to July 2022. The serum ACSL4, tumor necrosis factor-α, interleukin (IL)-6, IL-1β, and C-reactive protein levels were measured by enzyme-linked immunosorbent assay. Demographic and clinical statistics were also collected. In addition, all patients were followed up for 1 year, and patients with MACE were defined as poor prognosis group. All data used SPSS 26.0 to statistical analyses. The poor prognosis group had significantly higher age and low-density leptin cholesterol (LDLC) levels compared to the favorable prognosis group (P < .05). STEMI patients exhibited significantly elevated serum levels of ACSL4, tumor necrosis factor-α, IL-6, IL-1β, and C-reactive protein (P < .05). Serum ACSL4 and IL-1β levels in the poor prognosis group were remarkably enhanced compared to the favorable prognosis group. Curvilinear regression analysis demonstrated that ACSL4 was associated with LDLC and IL-1β. Moreover, ACSL4 (B = 0.138, 95% CI 1.108-1.189, P < .001), LDLC (B = 2.317, 95% CI 5.253-19.603, P < .001), and IL-1β (B = 0.061, 95%CI 1.008-1.122, P = .025) levels were the risk factors for STEMI patients with 1-year MACE. This study showed that the serum ACSL4 levels was remarkably elevated in STEMI patients. This study might provide new targets and a comprehensive approach to cardiovascular protection in STEMI patients.
Collapse
Affiliation(s)
- Yun Hu
- Emergency Department, Wuhan Dongxihu District People’s Hospital, Wuhan, China
| | - Qingye Li
- Emergency Department, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Yinglin Wang
- Emergency Department, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
9
|
Cui J, Zhao Y, Qian G, Yue X, Luo C, Li T. Cardiac magnetic resonance for the early prediction of reverse left ventricular remodeling in patients with ST-segment elevation myocardial infarction. Eur Radiol 2023; 33:8501-8512. [PMID: 37458756 DOI: 10.1007/s00330-023-09907-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 03/16/2023] [Accepted: 03/30/2023] [Indexed: 11/26/2023]
Abstract
OBJECTIVES To evaluate the changes in cardiac magnetic resonance (CMR) characteristics and investigate the predictors of reverse left ventricular remodeling (r-LVR) in ST-segment elevation myocardial infarction (STEMI) patients. MATERIALS AND METHODS Eighty-six STEMI patients (median 56 years) were retrospectively studied. The patients were divided into r-LVR and without r-LVR groups. CMR analysis included LV volume, infarct characteristics, and global and regional myocardial function. The strain and displacement were assessed by CMR-feature tracking. The predictors of r-LVR were analyzed by the logistic regression method. RESULTS There were 37 patients in the r-LVR group and 49 patients in the without r-LVR group. At initial CMR, there was no difference in LV volume and global cardiac function between the two groups. However, the infarct zone radial and longitudinal displacements were higher in the r-LVR group (p < 0.05, respectively). At the second CMR, the r-LVR group showed higher LVEF, lower LV volume, and total enhanced mass (all p < 0.05). The infarct zone radial and circumferential strains and radial displacement were higher in the r-LVR group (all p < 0.05). The r-LVR group had better recovery of myocardial injury and function. Of note, microvascular obstruction (MVO) mass (odds ratio: 0.779 (0.613-0.989), p = 0.041) and infarct zone peak longitudinal displacement (PLD) (odds ratio: 1.448 (1.044-2.008), p = 0.026) were independent predictors of r-LVR. CONCLUSIONS At initial CMR, there were no differences in global cardiac function between the two groups, but infarct zone displacements were higher in the r-LVR group. The r-LVR group had better recovery of cardiac function. In addition, MVO mass and infarct zone PLD were independent predictors of r-LVR. CLINICAL RELEVANCE STATEMENT Our study assessed changes in cardiac structure, function, and tissue characteristics after STEMI by CMR, investigated the best predictors of r-LVR in STEMI patients, and laid the foundation for the development of new parameter-guided treatment strategies for STEMI patients. KEY POINTS • At initial CMR, the reverse left ventricular remodeling (r-LVR) group had less myocardial damage and higher infarct zone displacement, but there were no differences in global function between the two groups. • Both groups showed recovery of myocardial injury and cardiac function over time, but the r-LVR group had less enhanced mass and better cardiac function compared to the without r-LVR group at the second CMR. • Microvascular obstruction mass and infarct zone peak longitudinal displacement by cardiac magnetic resonance feature-tracking were significant predictors of r-LVR in STEMI patients.
Collapse
Affiliation(s)
- Jianing Cui
- Department of Radiology, the First Medical center, PLA General Hospital, Beijing, China
| | - Yanan Zhao
- Department of Radiology, the First Medical center, PLA General Hospital, Beijing, China
| | - Geng Qian
- Department of Cardiology, The Six Medical Center, PLA General Hospital, Beijing, China
| | | | - Chuncai Luo
- Department of Radiology, the First Medical center, PLA General Hospital, Beijing, China
| | - Tao Li
- Department of Radiology, the First Medical center, PLA General Hospital, Beijing, China.
| |
Collapse
|
10
|
Mitsis A, Avraamides P, Lakoumentas J, Kyriakou M, Sokratous S, Karmioti G, Drakomathioulakis M, Theodoropoulos KC, Nasoufidou A, Evangeliou A, Vassilikos V, Fragakis N, Ziakas A, Tzikas S, Kassimis G. Role of inflammation following an acute myocardial infarction: design of INFINITY. Biomark Med 2023; 17:971-981. [PMID: 38235565 DOI: 10.2217/bmm-2023-0491] [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: 08/03/2023] [Accepted: 12/07/2023] [Indexed: 01/19/2024] Open
Abstract
After a myocardial infarction, the inflammatory response is connected to major adverse outcomes such as ischemia-reperfusion injury, adverse cardiac remodeling, infarct size and poor prognosis. INFlammatIoN amI sTudY (INFINITY) is a multicenter, prospective, observational, cohort study designed to investigate the prognostic role of the cytokines IL-6, IL-10, IL-18 and IL-17 and the adipokines leptin, apelin and chemerin in patients with acute coronary syndrome. The study will test if these inflammatory biomarkers reflect different clinical manifestations of coronary artery disease and have a prognostic role in a 6-month follow-up period. This study represents an opportunity to investigate further the prognostic role of a selected combination of proinflammatory and anti-inflammatory biomarkers in the prognosis and risk stratification of acute coronary syndrome patients.
Collapse
Affiliation(s)
- Andreas Mitsis
- Cardiology Department, Nicosia General Hospital, Nicosia, 2029, Cyprus
| | | | - John Lakoumentas
- Department of Medical Physics, School of Medicine, University of Patras, Patras, 26504, Greece
| | - Michaela Kyriakou
- Cardiology Department, Nicosia General Hospital, Nicosia, 2029, Cyprus
| | | | - Georgia Karmioti
- Cardiology Department, Nicosia General Hospital, Nicosia, 2029, Cyprus
| | | | - Konstantinos C Theodoropoulos
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, 54636, Greece
| | - Athina Nasoufidou
- Second Department of Cardiology, Aristotle University of Thessaloniki, Thessaloniki, 54642, Greece
| | - Alexandros Evangeliou
- Third Department of Cardiology, Aristotle University of Thessaloniki, Thessaloniki, 54642, Greece
| | - Vassilios Vassilikos
- Third Department of Cardiology, Aristotle University of Thessaloniki, Thessaloniki, 54642, Greece
| | - Nikolaos Fragakis
- Second Department of Cardiology, Aristotle University of Thessaloniki, Thessaloniki, 54642, Greece
| | - Antonios Ziakas
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, 54636, Greece
| | - Stergios Tzikas
- Third Department of Cardiology, Aristotle University of Thessaloniki, Thessaloniki, 54642, Greece
| | - George Kassimis
- Second Department of Cardiology, Aristotle University of Thessaloniki, Thessaloniki, 54642, Greece
| |
Collapse
|
11
|
Benedikt M, Mangge H, Aziz F, Curcic P, Pailer S, Herrmann M, Kolesnik E, Tripolt NJ, Pferschy PN, Wallner M, Zirlik A, Sourij H, von Lewinski D. Impact of the SGLT2-inhibitor empagliflozin on inflammatory biomarkers after acute myocardial infarction - a post-hoc analysis of the EMMY trial. Cardiovasc Diabetol 2023; 22:166. [PMID: 37407956 PMCID: PMC10324245 DOI: 10.1186/s12933-023-01904-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023] Open
Abstract
BACKGROUND SGTL2-inhibitors are a cornerstone in the treatment of heart failure, but data on patients with acute myocardial infarction (AMI) is limited. The EMMY trial was the first to show a significant reduction in NTproBNP levels as well as improved cardiac structure and function in post-AMI patients treated with Empagliflozin compared to placebo. However, data on the potential impact of SGLT2-inhibitors on inflammatory biomarkers after AMI are scarce. MATERIALS AND METHODS The EMMY trial is an investigator-initiated, multicentre, double-blind, placebo-controlled trial, which enrolled patients after AMI, receiving either 10 mg Empagliflozin once daily or placebo over a period of 26 weeks on top of standard guideline-recommended therapy starting within 72 h after percutaneous coronary intervention. In this post-hoc subgroup analysis of the EMMY trial, we investigated inflammatory biomarkers of 374 patients. The endpoints investigated were the mean change in inflammatory biomarkers such as high-sensitive c-reactive protein (hsCRP), interleukin-6 (IL-6), neutrophils, leukocytes, neutrophile/lymphocyte ratio (NLR) and platelet to lymphocyte ratio (PLR) from baseline to 26 weeks. RESULTS Baseline median (interquartile ranges) IL-6 was 17.9 pg/mL (9.0-38.7), hsCRP 18.9 mg/L (11.2-37.1), neutrophil count 7.9 x G/L (6.2-10.1), leukocyte count 10.8 x G/L (9.1-12.8) and neutrophile/lymphocyte ratio (NLR) of 0.74 (0.67-0.80). At week 26, a significant mean reduction in inflammatory biomarkers was observed, being 35.1 ± 3.2% (p < 0.001) for IL-6, 57.4 ± 0.7% (p < 0.001) for hsCRP, 26.1 ± 0.7% (p < 0.001) for neutrophils, 20.5 ± 0.6% (p < 0.001) for leukocytes, 10.22 ± 0.50% (p < 0.001) for NLR, and - 2.53 ± 0.92% for PLR (p = 0.006) with no significant difference between Empagliflozin and placebo treatment. CONCLUSION Trajectories of inflammatory biomarkers showed a pronounced decline after AMI, but Empagliflozin treatment did not impact this decline indicating no central role in blunted systemic inflammation mediating beneficial effects.
Collapse
Affiliation(s)
- Martin Benedikt
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, Auenbruggerplatz 15, Graz, 8036, Austria
| | - Harald Mangge
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Faisal Aziz
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, Auenbruggerplatz 15, Graz, 8036, Austria
- Interdisciplinary Metabolic Medicine Trials Unit, Medical University of Graz, Graz, Austria
| | - Pero Curcic
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Sabine Pailer
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Markus Herrmann
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Ewald Kolesnik
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, Auenbruggerplatz 15, Graz, 8036, Austria
| | - Norbert J Tripolt
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, Auenbruggerplatz 15, Graz, 8036, Austria
- Interdisciplinary Metabolic Medicine Trials Unit, Medical University of Graz, Graz, Austria
| | - Peter N Pferschy
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, Auenbruggerplatz 15, Graz, 8036, Austria
- Interdisciplinary Metabolic Medicine Trials Unit, Medical University of Graz, Graz, Austria
| | - Markus Wallner
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, Auenbruggerplatz 15, Graz, 8036, Austria
| | - Andreas Zirlik
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, Auenbruggerplatz 15, Graz, 8036, Austria
| | - Harald Sourij
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, Auenbruggerplatz 15, Graz, 8036, Austria.
- Interdisciplinary Metabolic Medicine Trials Unit, Medical University of Graz, Graz, Austria.
| | - Dirk von Lewinski
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, Auenbruggerplatz 15, Graz, 8036, Austria.
| |
Collapse
|
12
|
Wu O, Yuan C, Leng J, Zhang X, Liu W, Yang F, Zhang H, Li J, Khederzadeh S, Jiang Z, Fang H, Liu X, Lu X, Xia J. Colorable role of interleukin (IL)-6 in obesity hypertension: A hint from a Chinese adult case-control study. Cytokine 2023; 168:156226. [PMID: 37235887 DOI: 10.1016/j.cyto.2023.156226] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 04/10/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023]
Abstract
BACKGROUND Obesity and hypertension are major risk factors for cardiovascular diseases that affect millions of people worldwide. Both conditions are associated with chronic low-grade inflammation, which is mediated by cytokines such as interleukin-6 (IL-6). IL-6 is a multifunctional cytokine that can have pro-inflammatory or anti-inflammatory effects depending on the context. The exact role of IL-6 in obesity-associated hypertension is unclear. OBJECTIVE To investigate how IL-6 affects blood pressure, inflammation, and metabolic function in obesity-hypertension using a Chinese adult case-control study. METHODS A total of 153 participants were sorted into four subgroups according to their body mass index (BMI) and blood pressure (BP): normal healthy group (NH), just obesity group (JO), just-hypertension group (JH), and obesity-hypertension group (OH). Serum IL-6 concentrations were measured by Enzyme-linked Immunosorbent Assay (ELISA) and their correlations with anthropometric and laboratory parameters and their differences across the subgroups were examined. Multiple linear regression analysis was performed to identify the predictors of serum IL-6 concentrations in each group. RESULTS Serum IL-6 concentrations were higher in NH group than in JO group and correlated positively with diastolic blood pressure in NH and JO groups, but not in JH and OH groups. Serum IL-6 concentrations also correlated with albumin in NH group, alkaline phosphatase in JO group, serum creatinine and fasting blood glucose in JH group. The influencing factors of serum IL-6 concentrations varied among the four groups, with gender, diastolic blood pressure and albumin being significant predictors in NH group, alkaline phosphatase in JO group, age and serum creatinine in JH group, and none in OH group. CONCLUSIONS These results suggest that IL-6 may play diverse effects in the pathogenesis of obesity- hypertension, depending on the presence or absence of obesity and hypertension. Further studies are needed to elucidate the underlying mechanisms of IL-6 signaling and function in these diseases.
Collapse
Affiliation(s)
- Ou Wu
- Shulan International Medical College, Zhejiang Shuren University, Zhejiang, China.
| | - Chengda Yuan
- Department of Dermatology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou 310007, China
| | - Jianhang Leng
- Department of Central Laboratory/Medical examination center of Hangzhou, The Frist People's Hospital of Hangzhou, 310003, Zhejiang, China
| | - Xingyu Zhang
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Wei Liu
- JFIntelligent Healthcare Technology Co. Ltd, Building No.5-7, No.699 Tianxiang Avenue, Hi-Tech Zone, Nanchang, Jiangxi Province, China
| | - Fenfang Yang
- Department of Central Laboratory/Medical examination center of Hangzhou, The Frist People's Hospital of Hangzhou, 310003, Zhejiang, China
| | - Hu Zhang
- Department of Thoracic Surgery, Sir Run Run Shaw Hospital Affiliated with Medical College of Zhejiang University, Zhejiang, China
| | - Jiajia Li
- Department of Central Laboratory, the First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Saber Khederzadeh
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Zhizhi Jiang
- ZhaNongKou Street Community Health Service Center, Hangzhou, Zhejiang, China
| | - Hangyan Fang
- Hangzhou Linping District Center for Disease Prevention and Control, zhejiang, China
| | - Xiaodong Liu
- Hangzhou center for disease control and prevention, Zhejiang, China
| | - Xi Lu
- Hangzhou Vocational and Technical College, Zhejiang, China.
| | - Jiangwei Xia
- Department of Neurology, Xuanwu Hospital, National Center for Neurological Disorders, Capital Medical University, Beijing, China; Beijing Municipal Geriatric Medical Research Center, Beijing, China.
| |
Collapse
|
13
|
Hume RD, Deshmukh T, Doan T, Shim WJ, Kanagalingam S, Tallapragada V, Rashid F, Marcuello M, Blessing D, Selvakumar D, Raguram K, Pathan F, Graham D, Ounzain S, Kizana E, Harvey RP, Palpant NJ, Chong JJ. PDGF-AB Reduces Myofibroblast Differentiation Without Increasing Proliferation After Myocardial Infarction. JACC Basic Transl Sci 2023. [DOI: 10.1016/j.jacbts.2022.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
|
14
|
Alieva AM, Butenko AV, Teplova NV, Reznik EV, Valiev RK, Skripnichenko EА, Sozykin AV, Nikitin IG. The role of interleukin-6 in the development of cardiovascular diseases: A review. CONSILIUM MEDICUM 2023. [DOI: 10.26442/20751753.2022.12.201948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Currently, the search and study of new biological markers that can provide early diagnosis of cardiovascular diseases, serve as a laboratory tool for assessing the effectiveness of treatment, or be used as prognostic markers and risk stratification criteria is ongoing. Our literature review indicates the potentially important diagnostic and prognostic value of assessing members of the interleukin-6 family. It is expected that further scientific and clinical studies will demonstrate the possibility of using members of the interleukin-6 family as an additional laboratory tool for the diagnosis, risk stratification and prediction of cardiovascular events in cardiac patients. It is necessary to evaluate in detail the possibilities of blockade of these interleukin-6 molecules in patients with cardiovascular diseases in vitro and in vivo.
Collapse
|
15
|
Klotho improves cardiac fibrosis, inflammatory cytokines, ferroptosis, and oxidative stress in mice with myocardial infarction. J Physiol Biochem 2023:10.1007/s13105-023-00945-5. [PMID: 36701072 DOI: 10.1007/s13105-023-00945-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 01/16/2023] [Indexed: 01/27/2023]
Abstract
The anti-aging protein Klotho has been associated with cardiovascular health protection. Nevertheless, the protective mechanism remains unknown. The present study is aimed at exploring the effect of Klotho on cardiac remodeling and its potential mechanism in mice with myocardial infarction (MI). We used left anterior coronary artery descending ligation to develop an MI model for in vivo analyses. In contrast, H9C2 cells and cardiac fibroblasts were used to establish the oxygen-glucose deprivation (OGD) model in in vitro analyses. In vivo and in vitro models were treated with Klotho. Compound C, an AMPK signaling inhibitor, was used to determine whether Klotho's effects are mediated through the AMPK/mTOR signaling pathway. Echocardiography, Masson trichrome staining, immunofluorescence, immunohistochemistry, real-time polymerase chain reaction (RT-PCR), and western blot were used to detect the related indicators. The findings of the in vivo model indicate that Klotho treatment improved the mice's cardiac function, reduced cardiac fibrosis, and attenuated myocardial inflammatory factors, ferroptosis, and oxidative stress. The results of the in vitro model were in line with the findings of in vivo modeling. An AMPK inhibitor, Compound C, reversed all these effects. In conclusion, Klotho potentially improves cardiac remodeling in MI mice by regulating AMPK/mTOR signaling, demonstrating Klotho as an effective MI therapeutic agent.
Collapse
|
16
|
Modulation of the miR-122/Sirt-6/ACE2 axis on experimentally-induced myocardial infarction. Chem Biol Interact 2023; 369:110276. [PMID: 36414029 DOI: 10.1016/j.cbi.2022.110276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 11/03/2022] [Accepted: 11/18/2022] [Indexed: 11/20/2022]
Abstract
Myocardial infarction (MI) is a progressive myocardial necrosis that can lead to a number of life-threatening complications. MiRNAs have a crucial role in the pathogenesis of many cardiovascular diseases. Remarkably, miR-122 targets the sirtuin-6 (Sirt-6) gene, which is an essential regulator of cardiovascular function and is considered a partial angiotensin converting enzyme 2 (ACE2) activator. Modulation of this axis is supposed to contribute to MI pathogenesis. The current study aims to investigate the cardioprotective effects of xanthenone through targeting the miR-122/Sirt-6/ACE2 axis on experimentally-induced MI in rats. Xanthenone was administered for 14 days and isoprenaline was injected in the last 2 days of the experiment. Xanthenone treatment resulted in a significant downregulation of miR-122, which further upregulated Sirt-6 and thus activated the adenosine monophosphate-activated protein kinase (AMPK). AMPK increases ACE2 levels and results in a decrease in the level of its substrate angiotensin II resulting in the normalization of the inflammatory cytokines and the cardiac biomarkers. Finally, by targeting the miR-122/Sirt-6/AMPK/ACE2 axis, xanthenone has the potential to be a promising cardioprotective agent against MI.
Collapse
|
17
|
Chiorescu RM, Mocan M, Inceu AI, Buda AP, Blendea D, Vlaicu SI. Vulnerable Atherosclerotic Plaque: Is There a Molecular Signature? Int J Mol Sci 2022; 23:13638. [PMID: 36362423 PMCID: PMC9656166 DOI: 10.3390/ijms232113638] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 08/18/2023] Open
Abstract
Atherosclerosis and its clinical manifestations, coronary and cerebral artery diseases, are the most common cause of death worldwide. The main pathophysiological mechanism for these complications is the rupture of vulnerable atherosclerotic plaques and subsequent thrombosis. Pathological studies of the vulnerable lesions showed that more frequently, plaques rich in lipids and with a high level of inflammation, responsible for mild or moderate stenosis, are more prone to rupture, leading to acute events. Identifying the vulnerable plaques helps to stratify patients at risk of developing acute vascular events. Traditional imaging methods based on plaque appearance and size are not reliable in prediction the risk of rupture. Intravascular imaging is a novel technique able to identify vulnerable lesions, but it is invasive and an operator-dependent technique. This review aims to summarize the current data from literature regarding the main biomarkers involved in the attempt to diagnose vulnerable atherosclerotic lesions. These biomarkers could be the base for risk stratification and development of the new therapeutic drugs in the treatment of patients with vulnerable atherosclerotic plaques.
Collapse
Affiliation(s)
- Roxana Mihaela Chiorescu
- Internal Medicine Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Internal Medicine, Emergency Clinical County Hospital, 400006 Cluj-Napoca, Romania
| | - Mihaela Mocan
- Internal Medicine Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Internal Medicine, Emergency Clinical County Hospital, 400006 Cluj-Napoca, Romania
| | - Andreea Ioana Inceu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine, 400349 Cluj-Napoca, Romania
- Department of Cardiology, Nicolae Stăncioiu Heart Institute, 400001 Cluj-Napoca, Romania
| | - Andreea Paula Buda
- Department of Cardiology, Nicolae Stăncioiu Heart Institute, 400001 Cluj-Napoca, Romania
| | - Dan Blendea
- Department of Cardiology, Nicolae Stăncioiu Heart Institute, 400001 Cluj-Napoca, Romania
- Department of Cardiology, Iuliu Hațieganu University of Medicine and Pharmacy, 400437 Cluj-Napoca, Romania
| | - Sonia Irina Vlaicu
- Internal Medicine Department, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Internal Medicine, Emergency Clinical County Hospital, 400006 Cluj-Napoca, Romania
| |
Collapse
|
18
|
Thomas ND, Montgomery SC, Behers B, Reyes E, Ledermann T, Grzywacz JG. Work-family enrichment: A potential buffer of inflammation among black adults? Brain Behav Immun Health 2022; 25:100517. [PMID: 36164462 PMCID: PMC9508348 DOI: 10.1016/j.bbih.2022.100517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 09/15/2022] [Accepted: 09/18/2022] [Indexed: 12/02/2022] Open
Abstract
Background Inflammation plays a known role in the development of cardiovascular disease (CVD), the leading cause of death in the United States and a condition that disproportionately affects Blacks. Although social stressors are frequently studied, the role of positive experiences in inflammation and its potential for CVD remains understudied. To address this gap, we examined the relationship between work family enrichment and inflammation in a population-based sample of working adults. Methods Participants were 447 working adults from Refresher Cohort of the National Study of Midlife Development in the United States (MIDUS) and the oversample of Blacks from the Milwaukee, WI. Serum concentration of pro-inflammatory biomarkers (IL-6/sIL-6r; CPR; Fibrinogen) were obtained via blood draw. Family-to-work enrichment (FtoWE) and work-to-family enrichment (WtoFE) were each assessed with four established survey questions. Results Blacks had higher concentrations of IL-6, CRP and Fibrinogen, and lower levels of sIL-6r than whites. A significant inverse relationship was observed between WtoFE and systemic inflammation as well as WtoFE and serum IL-6 concentration. Conclusions Individuals who perceived a stronger enhancing effect from work onto family showed lower levels of systemic inflammation and decreased concentrations of the pro-inflammatory cytokine IL-6; highlighting the potential work-family enrichment or other positive experiences may have in buffering the negative cardiovascular effects of inflammation. However, variation between racial groups remain undetermined.
Collapse
Affiliation(s)
| | | | | | - Eduardo Reyes
- Florida State University College of Medicine, FL, USA
| | - Thomas Ledermann
- Florida State University College of Health and Human Sciences, FL, USA
| | | |
Collapse
|
19
|
Duddu S, Agrawal M, Chakrabarti R, Ghosh A, Chakravorty N, Tiwari A, Chandra Shukla P. Meta-analysis reveals inhibition of the inflammatory cytokine IL-6 affords limited protection post-myocardial ischemia/infarction. Heliyon 2022; 8:e10435. [PMID: 36090222 PMCID: PMC9449900 DOI: 10.1016/j.heliyon.2022.e10435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 05/02/2022] [Accepted: 08/18/2022] [Indexed: 12/09/2022] Open
Abstract
Background Proinflammatory cytokine cascades play crucial roles in the onset and progression of myocardial ischemia and infarction. Clinically, elevated serum levels of pro-inflammatory cytokine interleukin-6 is a poor prognostic indicator for future cardiac events and cardiac morbidity. Despite several reports, there is no clear evidence of cardiac benefits of inhibiting IL-6 in pre-clinical and clinical settings. Objective To analyze the available data systematically and perform a meta-analysis to show the evidence of effects of IL-6 inhibition on cardiac remodeling and mortality in ischemic animal models. Methods We used PICO framework and the quality of the studies was assessed using SYRCLE's risk of bias tool. Studies with interventions i.e., genetic deletion or pharmacological inhibition of IL-6/IL-6R were included for the meta-analysis. Systematic review was synthesized by including pre-clinical as well as randomized clinical trials involving myocardial infarction patients treated with IL-6 inhibitors. The effect size of the pooled data was determined using standard mean difference and 95% confidence intervals. Results A total of 12 pre-clinical studies were included in the review for analysis. Most of the studies showed an unclear risk of bias as the selection and reporting criteria were poorly described. We observed high heterogeneity in the included studies due to the varying duration of myocardial infarction and the dosage of IL-6 antibodies used in the studies. Overall inhibition of IL-6 significantly increased area at risk [p = 0.001, SMD = 0.49 (95% CI: -0.36, 1.35)] and significantly reduced ejection fraction [p = 0.001, SMD = -0.19 (95% CI: -1.39, 1.01)] and end-diastolic diameter [p = 0.02, SMD = -0.25 (95% CI: -0.87, 0.36)] of left ventricle post-MI, but no effects on infarct size [p < 0.01, SMD = 0.00; 95% CI: -1.34, 0.58). In randomized clinical trials, the overall effect on C-reactive protein remains significantly unchanged on CRP levels (SMD = -0.38; 95% CI: -1.94, 0.55) post-treatment with IL-6R inhibitor tocilizumab. The meta-regression demonstrates a significant positive correlation (p = 0.058) between the increase in ischemic area and duration of ischemia post-surgery in the absence of IL-6. This meta-analysis indicates mixed effect of IL-6 inhibition on cardiac remodeling post-MI, particularly in protecting the myocardium viability from damaging acute inflammation but not significant on cardiac function of ischemic animal models. Conclusion Despite the well-established pro-inflammatory nature of IL-6 in myocardial ischemia, our meta-analysis reports a limited contribution of IL-6 in the cardiac remodeling of hearts in animal models of myocardial ischemia. Moreover, genetically deleted IL-6 murine models produced contrasting results. Additional pre-clinical studies exploring the pharmacological inhibition of IL-6R are required to determine the beneficial effects of IL-6 inhibitors in regulating cardiac remodeling. The findings from IL-6R inhibition have better clinical relevance compared to genetically inhibited IL-6.
Collapse
|
20
|
Timpau AS, Miftode RS, Leca D, Timpau R, Miftode IL, Petris AO, Costache II, Mitu O, Nicolae A, Oancea A, Jigoranu A, Tuchilus CG, Miftode EG. A Real Pandora's Box in Pandemic Times: A Narrative Review on the Acute Cardiac Injury Due to COVID-19. LIFE (BASEL, SWITZERLAND) 2022; 12:life12071085. [PMID: 35888173 PMCID: PMC9318707 DOI: 10.3390/life12071085] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 01/08/2023]
Abstract
The intricate relationship between severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the cardiovascular system is an extensively studied pandemic topic, as there is an ever-increasing amount of evidence that reports a high prevalence of acute cardiac injury in the context of viral infection. In patients with Coronavirus disease 2019, COVID-19, a significant increase in serum levels of cardiac troponin or other various biomarkers was observed, suggesting acute cardiac injury, thus predicting both a severe course of the disease and a poor outcome. Pathogenesis of acute cardiac injury is not yet completely elucidated, though several mechanisms are allegedly involved, such as a direct cardiomyocyte injury, oxygen supply-demand inequity caused by hypoxia, several active myocardial depressant factors during sepsis, and endothelial dysfunction due to the hyperinflammatory status. Moreover, the increased levels of plasma cytokines and catecholamines and a significantly enhanced prothrombotic environment may lead to the destabilization and rupture of atheroma plaques, subsequently triggering an acute coronary syndrome. In the present review, we focus on describing the epidemiology, pathogenesis, and role of biomarkers in the diagnosis and prognosis of patients with acute cardiac injury in the setting of the COVID-19 pandemic. We also explore some novel therapeutic strategies involving immunomodulatory therapy, as well as their role in preventing a severe form of the disease, with both the short-term outcome and the long-term cardiovascular sequelae being equally important in patients with SARS-CoV-2 induced acute cardiac injury.
Collapse
Affiliation(s)
- Amalia-Stefana Timpau
- Department of Infectious Diseases (Internal Medicine II), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (A.-S.T.); (D.L.); (I.-L.M.); (E.-G.M.)
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (A.O.P.); (O.M.); (A.N.); (A.O.); (A.J.)
| | - Radu-Stefan Miftode
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (A.O.P.); (O.M.); (A.N.); (A.O.); (A.J.)
- Correspondence: (R.-S.M.); (I.I.C.)
| | - Daniela Leca
- Department of Infectious Diseases (Internal Medicine II), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (A.-S.T.); (D.L.); (I.-L.M.); (E.-G.M.)
| | - Razvan Timpau
- Department of Radiology and Medical Imaging, St. Spiridon Emergency Hospital, 700115 Iasi, Romania;
| | - Ionela-Larisa Miftode
- Department of Infectious Diseases (Internal Medicine II), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (A.-S.T.); (D.L.); (I.-L.M.); (E.-G.M.)
| | - Antoniu Octavian Petris
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (A.O.P.); (O.M.); (A.N.); (A.O.); (A.J.)
| | - Irina Iuliana Costache
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (A.O.P.); (O.M.); (A.N.); (A.O.); (A.J.)
- Correspondence: (R.-S.M.); (I.I.C.)
| | - Ovidiu Mitu
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (A.O.P.); (O.M.); (A.N.); (A.O.); (A.J.)
| | - Ana Nicolae
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (A.O.P.); (O.M.); (A.N.); (A.O.); (A.J.)
| | - Alexandru Oancea
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (A.O.P.); (O.M.); (A.N.); (A.O.); (A.J.)
| | - Alexandru Jigoranu
- Department of Internal Medicine I (Cardiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (A.O.P.); (O.M.); (A.N.); (A.O.); (A.J.)
| | - Cristina Gabriela Tuchilus
- Department of Preventive Medicine and Interdisciplinarity (Microbiology), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania;
| | - Egidia-Gabriela Miftode
- Department of Infectious Diseases (Internal Medicine II), Faculty of Medicine, University of Medicine and Pharmacy “Gr. T. Popa”, 700115 Iasi, Romania; (A.-S.T.); (D.L.); (I.-L.M.); (E.-G.M.)
| |
Collapse
|
21
|
Mitsis A, Kadoglou NPE, Lambadiari V, Alexiou S, Theodoropoulos KC, Avraamides P, Kassimis G. Prognostic role of inflammatory cytokines and novel adipokines in acute myocardial infarction: An updated and comprehensive review. Cytokine 2022; 153:155848. [PMID: 35301174 DOI: 10.1016/j.cyto.2022.155848] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/23/2022] [Accepted: 02/28/2022] [Indexed: 12/19/2022]
Abstract
Acute myocardial infarction (AMI) is one of the major causes of morbidity and mortality worldwide. The inflammation response during and after AMI is common and seems to play a key role in the peri-AMI period, related with ischaemia-reperfusion injury, adverse cardiac remodelling, infarct size and poor prognosis. In this article, we provide an updated and comprehensive overview of the most important cytokines and adipokines involved in the complex pathophysiology mechanisms in AMI, summarizing their prognostic role post-AMI. Data so far support that elevated levels of the major proinflammatory cytokines TNFα, IL-6 and IL-1 and the adipokines adiponectin, visfatin and resistin, are linked to high mortality and morbidity. In contrary, there is evidence that anti-inflammatory cytokines and adipokines as IL-10, omentin-1 and ghrelin can suppress the AMI-induced inflammatory response and are correlated with better prognosis. Mixed data make unclear the role of the novel adipokines leptin and apelin. After all, imbalance of pro-inflammatory and anti-inflammatory cytokines may result in worst AMI prognosis. The incorporation of these inflammation biomarkers in established prognostic models could further improve their prognostic power improving overall the management of AMI patients.
Collapse
Affiliation(s)
- Andreas Mitsis
- Cardiology Department, Nicosia General Hospital, Cyprus.
| | | | - Vaia Lambadiari
- Second Department of Internal Medicine, Research Institute and Diabetes Centre, Athens University Medical School, Attikon University General Hospital, Athens, Greece
| | - Sophia Alexiou
- Second Cardiology Department, "Hippokration" Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | | | - George Kassimis
- Second Cardiology Department, "Hippokration" Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
22
|
Bochaton T, Leboube S, Paccalet A, Crola Da Silva C, Buisson M, Mewton N, Amaz C, Varillon Y, Bonnefoy-Cudraz E, Rioufol G, Cho TH, Ovize M, Bidaux G, Nighoghossian N, Mechtouff L. Impact of Age on Systemic Inflammatory Profile of Patients With ST-Segment-Elevation Myocardial Infarction and Acute Ischemic Stroke. Stroke 2022; 53:2249-2259. [PMID: 35354295 DOI: 10.1161/strokeaha.121.036806] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Aging is associated with a chronic low-grade inflammatory state. This condition may affect the acute inflammatory response involved in ST-segment-elevation myocardial infarction (STEMI) or acute ischemic stroke (AIS). We sought to compare the profile of a set of circulating inflammatory markers between young and older patients admitted for STEMI or AIS. METHODS HIBISCUS-STEMI (Cohort of Patients to Identify Biological and Imaging Markers of Cardiovascular Outcomes in ST Elevation Myocardial Infarction) and HIBISCUS-STROKE (Cohort of Patients to Identify Biological and Imaging Markers of Cardiovascular Outcomes in Stroke) are 2 cohort studies that enrolled patients with STEMI treated with primary percutaneous coronary intervention in the cardiac intensive care unit of Lyon and patients with AIS treated with mechanical thrombectomy in the Lyon Stroke Center, respectively from 2016 to 2019. Patients were classified as older if they were ≥65 years and as young if they were <65 years. In both cohorts, CRP (C-reactive protein), IL (interleukin)-6, IL-8, IL-10, MCP (monocyte chemoattractant protein), sTNF-RI (soluble tumor necrosis factor receptor I), sST2 (soluble form suppression of tumorigenicity 2), and VCAM-1 (vascular cellular adhesion molecule-1) were measured on serum collected at 5 time points using enzyme-linked immunosorbent assay. A multiple logistic regression model was performed to detect an association between area under the curve of circulating inflammatory markers within the first 48 hours and older age. RESULTS A total of 260 patients with STEMI and 164 patients with AIS were included. Of them, there were 76 (29%) and 105 (64%) older patients with STEMI and AIS, respectively. Following multivariable analysis, a high area under the curve of IL-6 and sTNF-RI, a low lymphocyte count, and a high neutrophil-lymphocyte ratio at 24 hours were associated with older age in patients with STEMI and AIS. CONCLUSIONS Older patients had higher IL-6 and sTFN-RI levels within the first 48 hours associated with a lower lymphocyte count and a higher neutrophil-lymphocyte ratio at 24 hours in both cohorts.
Collapse
Affiliation(s)
- Thomas Bochaton
- Cardiac Intensive Care Unit (T.B., E.B.-C.).,Hospices Civils de Lyon, F-69500 Bron, France. INSERM U1060, CarMeN laboratory, Université de Lyon, Groupement Hospitalier Est, Bâtiment B13, F-69500 Bron, France (T.B., S.L., A.P., C.C.D.S., N.M., E.B.-C., T.-H.C., M.O., G.B., N.N., L.M.)
| | - Simon Leboube
- Hospices Civils de Lyon, F-69500 Bron, France. INSERM U1060, CarMeN laboratory, Université de Lyon, Groupement Hospitalier Est, Bâtiment B13, F-69500 Bron, France (T.B., S.L., A.P., C.C.D.S., N.M., E.B.-C., T.-H.C., M.O., G.B., N.N., L.M.)
| | - Alexandre Paccalet
- Hospices Civils de Lyon, F-69500 Bron, France. INSERM U1060, CarMeN laboratory, Université de Lyon, Groupement Hospitalier Est, Bâtiment B13, F-69500 Bron, France (T.B., S.L., A.P., C.C.D.S., N.M., E.B.-C., T.-H.C., M.O., G.B., N.N., L.M.)
| | - Claire Crola Da Silva
- Hospices Civils de Lyon, F-69500 Bron, France. INSERM U1060, CarMeN laboratory, Université de Lyon, Groupement Hospitalier Est, Bâtiment B13, F-69500 Bron, France (T.B., S.L., A.P., C.C.D.S., N.M., E.B.-C., T.-H.C., M.O., G.B., N.N., L.M.)
| | - Marielle Buisson
- Clinical Investigation Center, INSERM 1407 (M.B., N.M., C.A., Y.V., M.O.)
| | - Nathan Mewton
- Clinical Investigation Center, INSERM 1407 (M.B., N.M., C.A., Y.V., M.O.).,Hospices Civils de Lyon, F-69500 Bron, France. INSERM U1060, CarMeN laboratory, Université de Lyon, Groupement Hospitalier Est, Bâtiment B13, F-69500 Bron, France (T.B., S.L., A.P., C.C.D.S., N.M., E.B.-C., T.-H.C., M.O., G.B., N.N., L.M.)
| | - Camille Amaz
- Clinical Investigation Center, INSERM 1407 (M.B., N.M., C.A., Y.V., M.O.)
| | - Yvonne Varillon
- Clinical Investigation Center, INSERM 1407 (M.B., N.M., C.A., Y.V., M.O.)
| | - Eric Bonnefoy-Cudraz
- Cardiac Intensive Care Unit (T.B., E.B.-C.).,Hospices Civils de Lyon, F-69500 Bron, France. INSERM U1060, CarMeN laboratory, Université de Lyon, Groupement Hospitalier Est, Bâtiment B13, F-69500 Bron, France (T.B., S.L., A.P., C.C.D.S., N.M., E.B.-C., T.-H.C., M.O., G.B., N.N., L.M.)
| | - Gilles Rioufol
- Hospices Civils de Lyon, F-69500 Bron, France. INSERM U1060, CarMeN laboratory, Université de Lyon, Groupement Hospitalier Est, Bâtiment B13, F-69500 Bron, France (T.B., S.L., A.P., C.C.D.S., N.M., E.B.-C., T.-H.C., M.O., G.B., N.N., L.M.)
| | - Tae-Hee Cho
- Louis Pradel Hospital, Stroke Center, Hôpital Pierre Wertheimer (T.-H.C., N.N., L.M.).,Hospices Civils de Lyon, F-69500 Bron, France. INSERM U1060, CarMeN laboratory, Université de Lyon, Groupement Hospitalier Est, Bâtiment B13, F-69500 Bron, France (T.B., S.L., A.P., C.C.D.S., N.M., E.B.-C., T.-H.C., M.O., G.B., N.N., L.M.)
| | - Michel Ovize
- Clinical Investigation Center, INSERM 1407 (M.B., N.M., C.A., Y.V., M.O.).,Hospices Civils de Lyon, F-69500 Bron, France. INSERM U1060, CarMeN laboratory, Université de Lyon, Groupement Hospitalier Est, Bâtiment B13, F-69500 Bron, France (T.B., S.L., A.P., C.C.D.S., N.M., E.B.-C., T.-H.C., M.O., G.B., N.N., L.M.)
| | - Gabriel Bidaux
- Department of Interventional Cardiology, Cardiovascular Hospital and Claude-Bernard University, F-69500 Bron, France (G.R.)
| | - Norbert Nighoghossian
- Louis Pradel Hospital, Stroke Center, Hôpital Pierre Wertheimer (T.-H.C., N.N., L.M.).,Hospices Civils de Lyon, F-69500 Bron, France. INSERM U1060, CarMeN laboratory, Université de Lyon, Groupement Hospitalier Est, Bâtiment B13, F-69500 Bron, France (T.B., S.L., A.P., C.C.D.S., N.M., E.B.-C., T.-H.C., M.O., G.B., N.N., L.M.)
| | - Laura Mechtouff
- Louis Pradel Hospital, Stroke Center, Hôpital Pierre Wertheimer (T.-H.C., N.N., L.M.).,Hospices Civils de Lyon, F-69500 Bron, France. INSERM U1060, CarMeN laboratory, Université de Lyon, Groupement Hospitalier Est, Bâtiment B13, F-69500 Bron, France (T.B., S.L., A.P., C.C.D.S., N.M., E.B.-C., T.-H.C., M.O., G.B., N.N., L.M.)
| |
Collapse
|
23
|
Feng Y, Ye D, Wang Z, Pan H, Lu X, Wang M, Xu Y, Yu J, Zhang J, Zhao M, Xu S, Pan W, Yin Z, Ye J, Wan J. The Role of Interleukin-6 Family Members in Cardiovascular Diseases. Front Cardiovasc Med 2022; 9:818890. [PMID: 35402550 PMCID: PMC8983865 DOI: 10.3389/fcvm.2022.818890] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 02/11/2022] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease is one of the main causes of human mortality. Cytokines play crucial roles in the development of cardiovascular disease. Interleukin (IL)-6 family members are a series of cytokines, including IL-6, IL-11, IL-30, IL-31, OSM, LIF, CNTF, CT-1, CT-2, and CLC, that regulate multiple biological effects. Experimental and clinical evidence shows that IL-6 family members are closely related to cardiovascular diseases such as atherosclerosis, hypertension, aortic dissection, cardiac fibrosis, and cardiomyopathy. This review mainly discusses the role of IL-6 family members in cardiovascular disease for the sake of identifying possible intervention targets for cardiovascular disease prevention and treatment.
Collapse
Affiliation(s)
- Yongqi Feng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Di Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zhen Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Heng Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xiyi Lu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Junping Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shuwan Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wei Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zheng Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
- Cardiovascular Research Institute, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
24
|
Alkouri A, Cybularz M, Mierke J, Nowack T, Biedermann J, Ulbrich S, Fischer J, Heidrich FM, Jellinghaus S, Speiser U, Linke A, Pfluecke C. The predictive role of early CRP values for one-year mortality in the first two days after acute myocardial infarction. Biomarkers 2022; 27:293-298. [PMID: 35138209 DOI: 10.1080/1354750x.2022.2040591] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND An excessive inflammatory reaction after acute myocardial infarction (AMI) is known to be harmful. New anti-inflammatory therapies are required. PURPOSE This study assessed the predictive role of early CRP in patients with STEMI. METHODS 1003 patients with STEMI were analyzed. 180 patients with proven infection were excluded. CRP after 12 h, 24 h, and 48 h after pain onset were evaluated. RESULTS Of 823 patients, 103 (12.5%) died within one year after AMI. The deceased patients showed higher CRP, even after already 12 h (6 vs. 13 mg/l, p < 0.001), 24 h (13 vs. 25 mg/l, p < 0.001) and after 48 h (40 vs. 92 mg/l, p < 0.001). A CRP of ≥8 mg/l, 12 h after AMI, was found in 45% and was independently associated with long-term mortality (OR: 2.7, p = 0.03), after 24 h: CRP ≥18 mg/l in 44% (OR: 2.5, p = 0.03), after 48 h: CRP ≥53 mg/l in 44% (OR 1.9, p = 0.03). Early CRP values correlated strongly with the later maximum value of CRP (p < 0.001). CONCLUSIONS Already early CRP values are accurate for risk-prediction following AMI. By identifying patients who are beginning to develop an excessive inflammatory response, it may be possible to identify those who benefit from anti-inflammatory therapies.
Collapse
Affiliation(s)
- Ahmad Alkouri
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden.,Department for Internal Medicine and Cardiology, Städtisches Klinikum Görlitz, Germany
| | - Maria Cybularz
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden
| | - Johannes Mierke
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden
| | - Thomas Nowack
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden
| | - Jonathan Biedermann
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden
| | - Stefan Ulbrich
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden
| | - Julia Fischer
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden
| | - Felix M Heidrich
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden
| | - Stefanie Jellinghaus
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden
| | - Uwe Speiser
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden
| | - Axel Linke
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden
| | - Christian Pfluecke
- Department for Internal Medicine and Cardiology, Herzzentrum Dresden, University Clinic, Technische Universität Dresden.,Department for Internal Medicine and Cardiology, Städtisches Klinikum Görlitz, Germany
| |
Collapse
|
25
|
Ibanez B. Targeting inflammation to improve long-term outcome in ST-segment elevation myocardial infarction survivors. EUROPEAN HEART JOURNAL. ACUTE CARDIOVASCULAR CARE 2022; 11:124-126. [PMID: 35136996 DOI: 10.1093/ehjacc/zuac002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 01/11/2022] [Indexed: 06/14/2023]
Affiliation(s)
- Borja Ibanez
- Clinical Research Department, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernandez Almagro, 3. 28029. Madrid, Spain
- Cardiology Department, IIS-Fundación Jiménez Díaz University Hospital, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Spain
| |
Collapse
|
26
|
Tiller C, Reindl M, Holzknecht M, Lechner I, Schwaiger J, Brenner C, Mayr A, Klug G, Bauer A, Metzler B, Reinstadler SJ. Association of plasma interleukin-6 with infarct size, reperfusion injury, and adverse remodelling after ST-elevation myocardial infarction. EUROPEAN HEART JOURNAL. ACUTE CARDIOVASCULAR CARE 2022; 11:113-123. [PMID: 34849677 DOI: 10.1093/ehjacc/zuab110] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/19/2021] [Accepted: 11/03/2021] [Indexed: 11/13/2022]
Abstract
AIMS Little is known about the clinical relevance of interleukin (IL)-6 in patients with acute ST-elevation myocardial infarction (STEMI). This study examined the possible associations of plasma IL-6 concentrations with infarct size (IS), reperfusion injury and adverse left ventricular remodelling (LVR), in STEMI patients treated with primary percutaneous coronary intervention (PCI). METHODS AND RESULTS We prospectively included 170 consecutive STEMI patients (median age 57 years, 14% women) treated with primary PCI between 2017 and 2019. Blood samples for biomarker analyses including IL-6 were collected on Day 2. Left ventricular ejection fraction (LVEF), IS, and reperfusion injury [microvascular obstruction (MVO) and intramyocardial haemorrhage (IMH)] were determined using cardiac magnetic resonance (CMR) imaging on Day 4. Left ventricular remodelling was defined as ≥10% increase in left ventricular end-diastolic volume from baseline to 4 months CMR follow-up. Patients with IL-6 concentrations ≥median (17 ng/L) showed a significantly lower LVEF (43% vs. 52%, P < 0.001), larger IS (22% vs. 13%, P < 0.001), larger MVO (1.9% vs. 0.0%, P < 0.001), and more frequent IMH (52% vs. 18%, P < 0.001). Left ventricular remodelling was more common in patients with IL-6 ≥ median (24% vs. 9%, P = 0.005). In both linear and binary multivariable regression analyses, IL-6 remained independently associated with lower LVEF [odds ratio (OR): 0.10, 95% confidence interval (CI) 0.02-0.42, P = 0.002], larger IS (OR: 5.29, 95% CI 1.52-18.40, P = 0.009), larger MVO (OR: 5.20, 95% CI 1.30-20.85, P = 0.020), with presence of IMH (OR: 3.73, 95% CI 1.27-10.99, P = 0.017), and adverse LVR (OR: 2.72, 95% 1.06-6.98, P = 0.038). CONCLUSIONS High concentrations of circulating plasma IL-6 on Day 2 after STEMI were independently associated with worse myocardial function, larger infarct extent, more severe reperfusion injury, and a higher likelihood for LVR, suggesting IL-6 as a useful biomarker of more serious outcome and potential therapeutic target. CLINICAL TRIAL REGISTRATION https://clinicaltrials.gov/ct2/show/NCT04113356;NCT04113356.
Collapse
Affiliation(s)
- Christina Tiller
- Departement of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck A-6020, Austria
| | - Martin Reindl
- Departement of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck A-6020, Austria
| | - Magdalena Holzknecht
- Departement of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck A-6020, Austria
| | - Ivan Lechner
- Departement of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck A-6020, Austria
| | - Johannes Schwaiger
- Department of Internal Medicine, Academic Teaching Hospital Hall in Tirol, Milserstrasse 10, Hall in Tirol A-6060, Austria
| | - Christoph Brenner
- Departement of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck A-6020, Austria
| | - Agnes Mayr
- Department of Radiology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck A-6020, Austria
| | - Gert Klug
- Departement of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck A-6020, Austria
| | - Axel Bauer
- Departement of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck A-6020, Austria
| | - Bernhard Metzler
- Departement of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck A-6020, Austria
| | - Sebastian J Reinstadler
- Departement of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstrasse 35, Innsbruck A-6020, Austria
| |
Collapse
|
27
|
Letson HL, Biros E, Morris JL, Dobson GP. ALM Fluid Therapy Shifts Sympathetic Hyperactivity to Parasympathetic Dominance in the Rat Model of Non-Compressible Hemorrhagic Shock. Shock 2022; 57:264-273. [PMID: 34798632 DOI: 10.1097/shk.0000000000001886] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
ABSTRACT Excessive sympathetic outflow following trauma can lead to cardiac dysfunction, inflammation, coagulopathy, and poor outcomes. We previously reported that buprenorphine analgesia decreased survival after hemorrhagic trauma. Our aim is to examine the underlying mechanisms of mortality in a non-compressible hemorrhage rat model resuscitated with saline or adenosine, lidocaine, magnesium (ALM). Anesthetized adult male Sprague-Dawley rats were randomly assigned to Saline control group or ALM therapy group (both n = 10). Hemorrhage was induced by 50% liver resection. After 15 min, 0.7 mL/kg 3% NaCl ± ALM intravenous bolus was administered, and after 60 min, 0.9% NaCl ± ALM was infused for 4 h (0.5 mL/kg/h) with 72 h monitoring. Animals received 6-12-hourly buprenorphine for analgesia. Hemodynamics, heart rate variability, echocardiography, and adiponectin were measured. Cardiac tissue was analyzed for adrenergic/cholinergic receptor expression, inflammation, and histopathology. Four ALM animals and one Saline control survived to 72 h. Mortality was associated with up to 97% decreases in adrenergic (β-1, α-1A) and cholinergic (M2) receptor expression, cardiac inflammation, myocyte Ca2+ loading, and histopathology, indicating heart ischemia/failure. ALM survivors had higher cardiac output and stroke volume, a 30-fold increase in parasympathetic/sympathetic receptor expression ratio, and higher circulating adiponectin compared to Saline controls. Paradoxically, Saline cardiac adiponectin hormone levels were higher than ALM, with no change in receptor expression, indicating intra-cardiac synthesis. Mortality appears to be a "systems failure" associated with CNS dysregulation of cardiac function. Survival involves an increased parasympathetic dominance to support cardiac pump function with reduced myocardial inflammation. Increased cardiac α-1A adrenergic receptor in ALM survivors may be significant, as this receptor is highly protective during heart dysfunction/failure.
Collapse
Affiliation(s)
- Hayley L Letson
- Heart and Trauma Research Laboratory, College of Medicine and Dentistry, James Cook University, Queensland, Australia
| | | | | | | |
Collapse
|
28
|
Scărlătescu AI, Micheu MM, Popa-Fotea N, Pascal AM, Mihail AM, Petre I, Deaconu S, Vîjîiac A, Dorobanțu M. IL-6, IL-1RA and Resistin as Predictors of Left Ventricular Remodelling and Major Adverse Cardiac Events in Patients with Acute ST Elevation Myocardial Infarction. Diagnostics (Basel) 2022; 12:266. [PMID: 35204357 PMCID: PMC8871243 DOI: 10.3390/diagnostics12020266] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/12/2022] [Accepted: 01/17/2022] [Indexed: 12/14/2022] Open
Abstract
Despite continuous advances in diagnostic and therapeutic methods, acute myocardial infarction (AMI) remains a leading cause of morbidity and mortality worldwide. Considering the role of inflammation in AMI etiopathogenesis, we aimed to explore the role of a group of three inflammatory cytokines (IL-1RA, IL-6 and resistin) as an independent prognostic factor for LVR assessed by 3D echocardiography and MACE in patients with STEMI. We enrolled 41 patients with STEMI who underwent primary PCI. We assessed the occurrence of LVR (defined as an increase of over 20% in end-diastolic left ventricular volume at 6 months compared with baseline values) and MACE. Using the enzyme-linked immunosorbent assays (ELISA) method, we measured plasmatic levels of IL-6, IL-1RA and resistin (within 48 h after AMI and at 6 months). Out of 41 STEMI patients, 20.5% presented signs of LVR at follow up, and in 24.4%, MACE occurred. In univariate logistic regression analysis, baseline levels of IL-6 (OR = 1.042, p = 0.004), IL-1RA (OR = 1.004, p = 0.05) and resistin (OR = 1.7, p = 0.007) were all significantly associated with LVR. ROC analysis showed that the three cytokines as a group (AUC 0.946, p = 0.000) have a better predictive value for LVR than any individual cytokine. The group of cytokines also proved to have a better predictive value for MACE together than separately (AUC = 0.875, p = 0.000 for ROC regression model). IL-6, IL-1RA and resistin plasma levels at baseline have a good predictive value both as independent variables and also as a group for the development of adverse LVR and MACE at 6 months follow up after STEMI.
Collapse
Affiliation(s)
- Alina Ioana Scărlătescu
- Department of Cardiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.I.S.); (N.P.-F.); (I.P.); (A.V.); (M.D.)
- Emergency Clinical Hospital Bucharest, Department of Cardiology, 014461 Bucharest, Romania; (A.M.P.); (A.M.M.); (S.D.)
| | - Miruna Mihaela Micheu
- Emergency Clinical Hospital Bucharest, Department of Cardiology, 014461 Bucharest, Romania; (A.M.P.); (A.M.M.); (S.D.)
| | - Nicoleta Popa-Fotea
- Department of Cardiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.I.S.); (N.P.-F.); (I.P.); (A.V.); (M.D.)
- Emergency Clinical Hospital Bucharest, Department of Cardiology, 014461 Bucharest, Romania; (A.M.P.); (A.M.M.); (S.D.)
| | - Ana Maria Pascal
- Emergency Clinical Hospital Bucharest, Department of Cardiology, 014461 Bucharest, Romania; (A.M.P.); (A.M.M.); (S.D.)
| | - Ana Maria Mihail
- Emergency Clinical Hospital Bucharest, Department of Cardiology, 014461 Bucharest, Romania; (A.M.P.); (A.M.M.); (S.D.)
| | - Ioana Petre
- Department of Cardiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.I.S.); (N.P.-F.); (I.P.); (A.V.); (M.D.)
- Emergency Clinical Hospital Bucharest, Department of Cardiology, 014461 Bucharest, Romania; (A.M.P.); (A.M.M.); (S.D.)
| | - Silvia Deaconu
- Emergency Clinical Hospital Bucharest, Department of Cardiology, 014461 Bucharest, Romania; (A.M.P.); (A.M.M.); (S.D.)
| | - Aura Vîjîiac
- Department of Cardiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.I.S.); (N.P.-F.); (I.P.); (A.V.); (M.D.)
- Emergency Clinical Hospital Bucharest, Department of Cardiology, 014461 Bucharest, Romania; (A.M.P.); (A.M.M.); (S.D.)
| | - Maria Dorobanțu
- Department of Cardiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.I.S.); (N.P.-F.); (I.P.); (A.V.); (M.D.)
- Emergency Clinical Hospital Bucharest, Department of Cardiology, 014461 Bucharest, Romania; (A.M.P.); (A.M.M.); (S.D.)
| |
Collapse
|
29
|
Tøllefsen IM, Shetelig C, Seljeflot I, Eritsland J, Hoffmann P, Andersen GØ. High levels of interleukin-6 are associated with final infarct size and adverse clinical events in patients with STEMI. Open Heart 2021; 8:openhrt-2021-001869. [PMID: 34933964 PMCID: PMC8693166 DOI: 10.1136/openhrt-2021-001869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/22/2021] [Indexed: 11/29/2022] Open
Abstract
Objective Inflammation has emerged as a new treatment target in patients with coronary artery disease and inflammation seems to play an important role in ischaemia/reperfusion injury that follows ST-elevation myocardial infarction (STEMI). We aimed to explore the role of acute and sustained interleukin 6 (IL-6) signalling, including soluble IL-6 receptor (IL-6R), with regard to infarct size, adverse remodelling and future cardiovascular events in patients with STEMI. Methods We included 269 patients with first-time STEMI, symptom duration <6 hours and treated with percutaneous coronary intervention. Blood sampling and cardiac MRI were performed in the acute phase and after 4 months. Clinical events and all-cause mortality were registered during 12-month and 70-month follow-up, respectively. Results IL-6 levels above median at all sampling points were significantly associated with increased infarct size and reduced left ventricular ejection fraction (LVEF). IL-6 levels in the highest quartile were at all sampling points associated with an increased risk of having an adverse clinical event during the first 12 months and with long-term all-cause mortality. IL-6R was not associated with infarct size, LVEF, myocardial salvage or long-term all-cause mortality. Conclusion Acute and sustained elevation of IL-6 measured 4 months after STEMI were associated with larger infarct size, reduced LVEF and adverse clinical events including all-cause mortality. The results add important information to the sustained role of inflammation in patients with STEMI and IL-6 as a potential target for long-term intervention. Trial registration number NCT00922675.
Collapse
Affiliation(s)
| | - Christian Shetelig
- Department of Cardiology, Oslo University Hospital Ulleval, Oslo, Norway
| | - Ingebjørg Seljeflot
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo Faculty of Medicine, Oslo, Norway
| | - Jan Eritsland
- Department of Cardiology, Oslo University Hospital Ulleval, Oslo, Norway
| | - Pavel Hoffmann
- Section of Interventional Cardiology, Oslo universitetssykehus Ulleval, Oslo, Norway
| | | |
Collapse
|
30
|
Ou D, Ni D, Li R, Jiang X, Chen X, Li H. Galectin‑1 alleviates myocardial ischemia‑reperfusion injury by reducing the inflammation and apoptosis of cardiomyocytes. Exp Ther Med 2021; 23:143. [PMID: 35069824 PMCID: PMC8756402 DOI: 10.3892/etm.2021.11066] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 09/16/2021] [Indexed: 11/06/2022] Open
Affiliation(s)
- Dengke Ou
- Department of Cardiovascular Medicine, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, P.R. China
| | - Dan Ni
- Department of Nuclear Medicine, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, P.R. China
| | - Rong Li
- Department of Interventional Therapy, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, P.R. China
| | - Xiaobo Jiang
- Department of Cardiovascular Medicine, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, P.R. China
| | - Xiaoxiao Chen
- Department of Cardiovascular Medicine, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, P.R. China
| | - Hongfei Li
- Department of Cardiovascular Medicine, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, P.R. China
| |
Collapse
|
31
|
Wang K, Li Z, Sun Y, Liu X, Ma W, Ding Y, Hong J, Qian L, Xu D. Dapagliflozin Improves Cardiac Function, Remodeling, Myocardial Apoptosis, and Inflammatory Cytokines in Mice with Myocardial Infarction. J Cardiovasc Transl Res 2021; 15:786-796. [PMID: 34855147 DOI: 10.1007/s12265-021-10192-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 11/22/2021] [Indexed: 11/28/2022]
Abstract
Dapagliflozin (DAPA) exerts cardioprotective effects in non-diabetic patients. Nonetheless, the protective mechanism remains unknown. This study aims to evaluate the performance of DAPA on cardiac function and remodeling as well as its potential mechanism in mice with myocardial infarction (MI). Here, a MI mice model was established. One week after MI, mice were treated with saline or DAPA (1.5 mg/kg/day) for 4 weeks. At the end of this study, echocardiography was performed to assess cardiac structure and function. Myocardial apoptosis was analyzed by Western blot and immunofluorescence. Inflammatory cytokines and cardiac fibrosis were analyzed by real-time PCR and Masson's trichrome stain, respectively. Results showed that DAPA improved cardiac structure and function, attenuated cardiac fibrosis, and inhibited inflammatory cytokines and myocardial apoptosis. Moreover, the inhibition of PI3K/AKT/mTOR pathway might be related to the cardioprotective role of DAPA. These findings reveal that dapagliflozin is a potential therapeutic agent for MI patients without diabetes.
Collapse
Affiliation(s)
- Kai Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Zhongming Li
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Yan Sun
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Xianling Liu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Wenjie Ma
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Yinzhang Ding
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Jian Hong
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Lijun Qian
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Di Xu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
32
|
Martins-Marques T. Connecting different heart diseases through intercellular communication. Biol Open 2021; 10:bio058777. [PMID: 34494646 PMCID: PMC8443862 DOI: 10.1242/bio.058777] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 07/12/2021] [Indexed: 12/22/2022] Open
Abstract
Well-orchestrated intercellular communication networks are pivotal to maintaining cardiac homeostasis and to ensuring adaptative responses and repair after injury. Intracardiac communication is sustained by cell-cell crosstalk, directly via gap junctions (GJ) and tunneling nanotubes (TNT), indirectly through the exchange of soluble factors and extracellular vesicles (EV), and by cell-extracellular matrix (ECM) interactions. GJ-mediated communication between cardiomyocytes and with other cardiac cell types enables electrical impulse propagation, required to sustain synchronized heart beating. In addition, TNT-mediated organelle transfer has been associated with cardioprotection, whilst communication via EV plays diverse pathophysiological roles, being implicated in angiogenesis, inflammation and fibrosis. Connecting various cell populations, the ECM plays important functions not only in maintaining the heart structure, but also acting as a signal transducer for intercellular crosstalk. Although with distinct etiologies and clinical manifestations, intercellular communication derailment has been implicated in several cardiac disorders, including myocardial infarction and hypertrophy, highlighting the importance of a comprehensive and integrated view of complex cell communication networks. In this review, I intend to provide a critical perspective about the main mechanisms contributing to regulate cellular crosstalk in the heart, which may be considered in the development of future therapeutic strategies, using cell-based therapies as a paradigmatic example. This Review has an associated Future Leader to Watch interview with the author.
Collapse
Affiliation(s)
- Tania Martins-Marques
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, 3000-548 Coimbra, Portugal
- Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), 3004-504 Coimbra, Portugal
- Clinical Academic Centre of Coimbra (CACC), 3004-561 Coimbra, Portugal
| |
Collapse
|
33
|
Erdoğan M, Kaya Kalem A, Öztürk S, Erdöl MA, Kayaaslan B, Özbebek YE, Güner R. Interleukin-6 level is an independent predictor of right ventricular systolic dysfunction in patients hospitalized with COVID-19. Anatol J Cardiol 2021; 25:555-564. [PMID: 34369883 PMCID: PMC8357443 DOI: 10.5152/anatoljcardiol.2021.24946] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVE Cytokine storm with elevated levels of multiple proinflammatory cytokines and inflammatory system activation underlie the pathogenesis of coronavirus disease 2019 (COVID-19). In this study, we aimed to investigate whether increased interleukin (IL)-6 levels can predict right ventricular (RV) systolic impairment in patients hospitalized with COVID-19. METHODS This prospective, observational study included 100 consecutive patients hospitalized with mild and moderate COVID-19. All the patients underwent chest computerized tomography, detailed laboratory tests including IL-6, and two dimensional (2D) transthoracic echocardiography (TTE) with assessment of 2D conventional and Doppler echocardiography parameters and RV systolic functions. RESULTS After the elimination of six patients with exclusion criteria, the remaining patients were classified into two groups, namely normal RV systolic functions (n=60) and impaired RV systolic functions (n=34). IL-6 levels were significantly higher in patients with impaired RV systolic functions than in those with normal RV systolic functions (20.3, 4.6, p<0.001, respectively). Tricuspid annular plane systolic excursion and RV derived tissue Doppler imaging (TDI) S' measurements were similar between the two groups. RV fractional area change was significantly lower, and RV TDI derived index of myocardial performance was significantly higher in patients with impaired RV systolic functions. In multivariate analysis, IL-6 levels independently predicted deterioration in RV systolic function at a significant level (odds ratio: 1.12, 95% confidence interval: 1.04-1.20, p=0.003). CONCLUSION IL-6 is an independent predictor of RV systolic impairment in patients hospitalized with mild and moderate COVID-19 suggesting a possible pathogenetic mechanism. IL-6 levels can be used to predict RV systolic impairment in patients suffering from this infection.
Collapse
Affiliation(s)
- Mehmet Erdoğan
- Department of Cardiology, Ankara Yıldırım Beyazıt University; Ankara-Turkey
- Department of Cardiology, Ministry of Health, Ankara City Hospital; Ankara-Turkey
| | - Ayşe Kaya Kalem
- Department of Infectious Disease and Clinical Microbiology, Faculty of Medicine, Ankara Yıldırım Beyazıt University; Ankara-Turkey
| | - Selçuk Öztürk
- Department of Cardiology, Faculty of Medicine, Yozgat Bozok University; Yozgat-Turkey
| | - Mehmet Akif Erdöl
- Department of Cardiology, Ministry of Health, Ankara City Hospital; Ankara-Turkey
| | - Bircan Kayaaslan
- Department of Infectious Disease and Clinical Microbiology, Faculty of Medicine, Ankara Yıldırım Beyazıt University; Ankara-Turkey
| | - Yunus Emre Özbebek
- Department of Cardiology, Ankara Yıldırım Beyazıt University; Ankara-Turkey
| | - Rahmet Güner
- Department of Infectious Disease and Clinical Microbiology, Faculty of Medicine, Ankara Yıldırım Beyazıt University; Ankara-Turkey
| |
Collapse
|
34
|
Opincariu D, Rodean I, Rat N, Hodas R, Benedek I, Benedek T. Systemic Vulnerability, as Expressed by I-CAM and MMP-9 at Presentation, Predicts One Year Outcomes in Patients with Acute Myocardial Infarction-Insights from the VIP Clinical Study. J Clin Med 2021; 10:jcm10153435. [PMID: 34362217 PMCID: PMC8347806 DOI: 10.3390/jcm10153435] [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: 06/28/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/18/2022] Open
Abstract
(1) Background: The prediction of recurrent events after acute myocardial infarction (AMI) does not sufficiently integrate systemic inflammation, coronary morphology or ventricular function in prediction algorithms. We aimed to evaluate the accuracy of inflammatory biomarkers, in association with angiographical and echocardiographic parameters, in predicting 1-year MACE after revascularized AMI. (2) Methods: This is an extension of a biomarker sub-study of the VIP trial (NCT03606330), in which 225 AMI patients underwent analysis of systemic vulnerability and were followed for 1 year. Hs-CRP, MMP-9, IL-6, I-CAM, V-CAM and E-selectin were determined at 1 h after revascularization. The primary end-point was the 1-year MACE rate. (3) Results: The MACE rate was 24.8% (n = 56). There were no significant differences between groups in regard to IL-6, V-CAM and E-selectin. The following inflammatory markers were significantly higher in MACE patients: hs-CRP (11.1 ± 13.8 vs. 5.1 ± 4.4 mg/L, p = 0.03), I-CAM (452 ± 283 vs. 220.5 ± 104.6, p = 0.0003) and MMP-9 (2255 ± 1226 vs. 1099 ± 706.1 ng/mL p = 0.0001). The most powerful predictor for MACE was MMP-9 of >1155 ng/mL (AUC-0.786, p < 0.001) even after adjustments for diabetes, LVEF, acute phase complications and other inflammatory biomarkers. For STEMI, the most powerful predictors for MACE included I-CAM > 239.7 ng/mL, V-CAM > 877.9 ng/mL and MMP-9 > 1393 ng/mL. (4) Conclusions: High levels of I-CAM and MMP-9 were the most powerful predictors for recurrent events after AMI for the overall study population. For STEMI subjects, the most important predictors included increased levels of I-CAM, V-CAM and MMP-9, while none of the analyzed parameters had proven to be predictive. Inflammatory biomarkers assayed during the acute phase of AMI presented a more powerful predictive capacity for MACE than the LVEF.
Collapse
Affiliation(s)
- Diana Opincariu
- Department of Cardiology, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade”, 540142 Târgu Mureș, Romania; (I.R.); (N.R.); (I.B.); (T.B.)
- Cardiomed Medical Center, 22 December 1989 Street, No. 76, 540124 Târgu Mureș, Romania
- Correspondence: or (D.O.); or (R.H.); Tel.: +40-756-787-587 (D.O.); +40-742-385-600 (R.H.)
| | - Ioana Rodean
- Department of Cardiology, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade”, 540142 Târgu Mureș, Romania; (I.R.); (N.R.); (I.B.); (T.B.)
| | - Nora Rat
- Department of Cardiology, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade”, 540142 Târgu Mureș, Romania; (I.R.); (N.R.); (I.B.); (T.B.)
- Cardiomed Medical Center, 22 December 1989 Street, No. 76, 540124 Târgu Mureș, Romania
| | - Roxana Hodas
- Department of Cardiology, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade”, 540142 Târgu Mureș, Romania; (I.R.); (N.R.); (I.B.); (T.B.)
- Correspondence: or (D.O.); or (R.H.); Tel.: +40-756-787-587 (D.O.); +40-742-385-600 (R.H.)
| | - Imre Benedek
- Department of Cardiology, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade”, 540142 Târgu Mureș, Romania; (I.R.); (N.R.); (I.B.); (T.B.)
- Cardiomed Medical Center, 22 December 1989 Street, No. 76, 540124 Târgu Mureș, Romania
| | - Theodora Benedek
- Department of Cardiology, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade”, 540142 Târgu Mureș, Romania; (I.R.); (N.R.); (I.B.); (T.B.)
- Cardiomed Medical Center, 22 December 1989 Street, No. 76, 540124 Târgu Mureș, Romania
| |
Collapse
|
35
|
Wang J, Liu W, Chen H, Liu C, Wang M, Chen H, Zhou H, Liu Z, Zhang S, Yu Z, Duan S, Deng Q, Sun J, Jiang H, Yu L. Novel Insights Into the Interaction Between the Autonomic Nervous System and Inflammation on Coronary Physiology: A Quantitative Flow Ratio Study. Front Cardiovasc Med 2021; 8:700943. [PMID: 34386531 PMCID: PMC8354298 DOI: 10.3389/fcvm.2021.700943] [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: 04/27/2021] [Accepted: 07/02/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Heart rate variability (HRV) was proposed as a noninvasive biomarker to stratify the risk of cardiovascular disease. However, it remains to be determined if HRV can be used as a surrogate for coronary artery physiology as analyzed by quantitative flow ratio (QFR) in patients with new-onset unstable angina pectoris (UAP). Methods: A total of 129 consecutive patients with new-onset UAP who underwent 24-h long-range 12-channel electrocardiography from June 2020 to December 2020 were included in this study. HRV, coronary angiography, and QFR information was retrieved from patient medical records, the severity of coronary lesions was evaluated using the Gensini score (GS), and total atherosclerotic burden was assessed using the three-vessel contrast QFR (3V-cQFR) calculated as the sum of cQFR in three vessels. Results: Multivariate logistic analysis showed that low-frequency power (LF) and high-sensitivity C-reactive protein (hs-CRP) were directly correlated with functional ischemia of target vessel, which were inversely correlated with total atherosclerotic burden as assessed by 3V-cQFR. Moreover, incorporation of the increase in LF into the existing model that uses clinical risk factors, GS, and hs-CRP significantly increased the discriminatory ability for evaluating coronary artery physiology of target vessel. Conclusions: LF and hs-CRP are independently associated with functional ischemia in patients with new-onset UAP. The relative increase of LF and hs-CRP could add value to the use of classical cardiovascular risk factors to predict the functional severity of coronary artery stenosis. Our results suggest a potential association between the autonomic nervous system, inflammation, and coronary artery physiology.
Collapse
Affiliation(s)
- Jun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Centre of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wei Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Centre of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Huaqiang Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Centre of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Chengzhe Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Centre of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Meng Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Centre of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Hu Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Centre of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Huixin Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Centre of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zhihao Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Centre of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Song Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Centre of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zhongyang Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Centre of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shoupeng Duan
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Centre of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Qiang Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Centre of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Ji Sun
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Centre of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Centre of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Lilei Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiac Autonomic Nervous System Research Centre of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
36
|
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: 69] [Impact Index Per Article: 17.3] [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
|
37
|
Abstract
IL (interleukin)-6 is a pivotal cytokine of innate immunity, which enacts a broad set of physiological functions traditionally associated with host defense, immune cell regulation, proliferation, and differentiation. Following recognition of innate immune pathways leading from the NLRP3 (NOD-, LRR-, and pyrin domain-containing protein 3) inflammasome to IL-1 to IL-6 and on to the hepatically derived clinical biomarker CRP (C-reactive protein), an expanding literature has led to understanding of the proatherogenic role for IL-6 in cardiovascular disease and thus the potential for IL-6 inhibition as a novel method for vascular protection. In this review, we provide an overview of the mechanisms by which IL-6 signaling occurs and how that impacts upon pharmacological inhibition; describe murine models of IL-6 and atherogenesis; summarize human epidemiological data outlining the utility of IL-6 as a biomarker of vascular risk; outline genetic data suggesting a causal role for IL-6 in systemic atherothrombosis and aneurysm formation; and then detail the potential role of IL-6 inhibition in stable coronary disease, acute coronary syndromes, heart failure, and the atherothrombotic complications associated with chronic kidney disease and end-stage renal failure. Finally, we review anti-inflammatory and antithrombotic findings for ziltivekimab, a novel IL-6 ligand inhibitor being developed specifically for use in atherosclerotic disease and poised to be tested formally in a large-scale cardiovascular outcomes trial focused on individuals with chronic kidney disease and elevated levels of CRP, a population at high residual atherothrombotic risk, high residual inflammatory risk, and considerable unmet clinical need.
Collapse
MESH Headings
- Aneurysm/etiology
- Animals
- Antibodies, Monoclonal, Humanized/therapeutic use
- Atherosclerosis/etiology
- Atherosclerosis/metabolism
- C-Reactive Protein/metabolism
- Cardiovascular Diseases/metabolism
- Cardiovascular Diseases/therapy
- Cell Differentiation
- Cell Proliferation
- Disease Models, Animal
- Humans
- Immunity, Cellular
- Immunity, Innate
- Inflammasomes
- Inflammation/complications
- Interleukin-1beta/antagonists & inhibitors
- Interleukin-1beta/metabolism
- Interleukin-6/antagonists & inhibitors
- Interleukin-6/genetics
- Interleukin-6/immunology
- Interleukin-6/metabolism
- Mice
- Myocardial Ischemia/therapy
- NLR Family, Pyrin Domain-Containing 3 Protein
- Receptors, Interleukin-6/antagonists & inhibitors
- Receptors, Interleukin-6/metabolism
- Renal Dialysis
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/therapy
- Thrombosis/etiology
Collapse
Affiliation(s)
- Paul M Ridker
- Department of Medicine, Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Manas Rane
- Department of Medicine, Center for Cardiovascular Disease Prevention, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
38
|
Hofbauer TM, Mangold A, Ondracek AS, Panzenböck A, Scherz T, Müller J, Distelmaier K, Seidl V, Kastl S, Müller-Nurasyid M, Peters A, Strauch K, Winker R, Wohlschläger-Krenn E, Nistler S, Lang IM. Deoxyribonuclease 1 Q222R single nucleotide polymorphism and long-term mortality after acute myocardial infarction. Basic Res Cardiol 2021; 116:29. [PMID: 33891165 PMCID: PMC8064981 DOI: 10.1007/s00395-021-00864-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 03/18/2021] [Indexed: 12/23/2022]
Abstract
Upon activation, neutrophils release neutrophil extracellular traps (NETs), which contribute to circulating DNA burden and thrombosis, including ST-segment elevation myocardial infarction (STEMI). Deoxyribonuclease (DNase) 1 degrades circulating DNA and NETs. Lower DNase activity correlates with NET burden and infarct size. The DNase 1 Q222R single nucleotide polymorphism (SNP), impairing DNase 1 function, is linked with myocardial infarction. We assessed whether the Q222R SNP is connected to increased NET burden in STEMI and influences long-term outcomes. We enrolled 711 STEMI patients undergoing primary percutaneous coronary intervention (pPCI), and 1422 controls. Genotyping was performed for DNase 1 Q222R SNP. DNase activity, double-stranded (ds)DNA and citrullinated histone H3 were determined in culprit site and peripheral plasma during pPCI. The association of the Q222R variant on cardiovascular and all-cause mortality was assessed by multivariable Cox regression adjusted for cardiovascular risk factors. Homozygous Q222R DNase 1 variant was present in 64 (9.0%) STEMI patients, at the same frequency as in controls. Patients homozygous for Q222R displayed less DNase activity and increased circulating DNA burden. In overall patients, median survival was 60 months. Homozygous Q222R variant was independently associated with cardiovascular and all-cause mortality after STEMI. dsDNA/DNase ratio independently predicted cardiovascular and all-cause mortality. These findings highlight that the Q222R DNase 1 SNP is associated with increased NET burden and decreased compensatory DNase activity, and may serve as an independent risk factor for poor outcome after STEMI.
Collapse
Affiliation(s)
- Thomas M Hofbauer
- Department of Cardiology, Division of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Andreas Mangold
- Department of Cardiology, Division of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Anna S Ondracek
- Department of Cardiology, Division of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Adelheid Panzenböck
- Department of Cardiology, Division of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Thomas Scherz
- Department of Cardiology, Division of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Julian Müller
- Department of Cardiology, Division of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Klaus Distelmaier
- Department of Cardiology, Division of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Veronika Seidl
- Department of Cardiology, Division of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Stefan Kastl
- Department of Cardiology, Division of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,IBE (Institute for Medical Information Processing, Biometry, and Epidemiology), Faculty of Medicine, LMU Munich, Munich, Germany.,Department of Internal Medicine I (Cardiology), Hospital of the Ludwig-Maximilians-University (LMU) Munich, Munich, Germany.,Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Chair of Genetic Epidemiology, Faculty of Medicine, IBE, LMU Munich, Munich, Germany.,Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Robert Winker
- Health and Prevention Center, Sanatorium Hera, Löblichgasse 14, 1090, Vienna, Austria
| | | | - Sonja Nistler
- Health and Prevention Center, Sanatorium Hera, Löblichgasse 14, 1090, Vienna, Austria
| | - Irene M Lang
- Department of Cardiology, Division of Cardiology, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| |
Collapse
|
39
|
Libby P. Targeting Inflammatory Pathways in Cardiovascular Disease: The Inflammasome, Interleukin-1, Interleukin-6 and Beyond. Cells 2021; 10:951. [PMID: 33924019 PMCID: PMC8073599 DOI: 10.3390/cells10040951] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 02/08/2023] Open
Abstract
Recent clinical trials have now firmly established that inflammation participates causally in human atherosclerosis. These observations point the way toward novel treatments that add to established therapies to help stem the growing global epidemic of cardiovascular disease. Fortunately, we now have a number of actionable targets whose clinical exploration will help achieve the goal of optimizing beneficial effects while avoiding undue interference with host defenses or other unwanted actions. This review aims to furnish the foundation for this quest by critical evaluation of the current state of anti-inflammatory interventions within close reach of clinical application, with a primary focus on innate immunity. In particular, this paper highlights the pathway from the inflammasome, through interleukin (IL)-1 to IL-6 supported by a promising body of pre-clinical, clinical, and human genetic data. This paper also considers the use of biomarkers to guide allocation of anti-inflammatory therapies as a step toward realizing the promise of precision medicine. The validation of decades of experimental work and association studies in humans by recent clinical investigations provides a strong impetus for further efforts to target inflammation in atherosclerosis to address the considerable risk that remains despite current therapies.
Collapse
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
40
|
Zegeye MM, Andersson JSO, Wennberg P, Repsilber D, Sirsjö A, Ljungberg LU. IL-6 as a Mediator of the Association Between Traditional Risk Factors and Future Myocardial Infarction: A Nested Case-Control Study. Arterioscler Thromb Vasc Biol 2021; 41:1570-1579. [PMID: 33657883 DOI: 10.1161/atvbaha.120.315793] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Mulugeta M Zegeye
- Cardiovascular Research Centre, School of Medical Sciences, Örebro University, Sweden (M.M.Z., D.R., A.S., L.U.L.)
| | - Jonas S O Andersson
- Skellefteå Research Unit, Department of Public Health and Clinical Medicine (J.S.O.A.), Umeå University, Sweden
| | - Patrik Wennberg
- Department of Public Health and Clinical Medicine, Family Medicine (P.W.), Umeå University, Sweden
| | - Dirk Repsilber
- Cardiovascular Research Centre, School of Medical Sciences, Örebro University, Sweden (M.M.Z., D.R., A.S., L.U.L.)
| | - Allan Sirsjö
- Cardiovascular Research Centre, School of Medical Sciences, Örebro University, Sweden (M.M.Z., D.R., A.S., L.U.L.)
| | - Liza U Ljungberg
- Cardiovascular Research Centre, School of Medical Sciences, Örebro University, Sweden (M.M.Z., D.R., A.S., L.U.L.)
| |
Collapse
|
41
|
Pearce L, Davidson SM, Yellon DM. Does remote ischaemic conditioning reduce inflammation? A focus on innate immunity and cytokine response. Basic Res Cardiol 2021; 116:12. [PMID: 33629195 PMCID: PMC7904035 DOI: 10.1007/s00395-021-00852-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 02/04/2021] [Indexed: 02/07/2023]
Abstract
The benefits of remote ischaemic conditioning (RIC) have been difficult to translate to humans, when considering traditional outcome measures, such as mortality and heart failure. This paper reviews the recent literature of the anti-inflammatory effects of RIC, with a particular focus on the innate immune response and cytokine inhibition. Given the current COVID-19 pandemic, the inflammatory hypothesis of cardiac protection is an attractive target on which to re-purpose such novel therapies. A PubMed/MEDLINE™ search was performed on July 13th 2020, for the key terms RIC, cytokines, the innate immune system and inflammation. Data suggest that RIC attenuates inflammation in animals by immune conditioning, cytokine inhibition, cell survival and the release of anti-inflammatory exosomes. It is proposed that RIC inhibits cytokine release via a reduction in nuclear factor kappa beta (NF-κB)-mediated NLRP3 inflammasome production. In vivo, RIC attenuates pro-inflammatory cytokine release in myocardial/cerebral infarction and LPS models of endotoxaemia. In the latter group, cytokine inhibition is associated with a profound survival benefit. Further clinical trials should establish whether the benefits of RIC in inflammation can be observed in humans. Moreover, we must consider whether uncomplicated MI and elective surgery are the most suitable clinical conditions in which to test this hypothesis.
Collapse
Affiliation(s)
- Lucie Pearce
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK.
| |
Collapse
|
42
|
Dal Fabbro J, Candreva A, Rossi VA, Shahin M, Yousif N, Lüscher TF, Duru F, Denegri A. Clinical and electrocardiographic features of patients with myocardial infarction with non-obstructive coronary artery disease (MINOCA). J Cardiovasc Med (Hagerstown) 2021; 22:104-109. [PMID: 32706560 DOI: 10.2459/jcm.0000000000001027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AIMS Myocardial infarction with non-obstructive coronary artery disease (MINOCA) is often an underdiagnosed and undertreated condition. This study aimed to evaluate clinical and ECG characteristics of MINOCA in a large cohort of patients admitted for acute coronary syndrome. METHODS All coronary angiograms performed at the University Heart Center in Zurich (Switzerland) between 2012 and 2016 were investigated. MINOCA was defined according to European Society of Cardiology guidelines and patients were divided into two groups, based on the presence or absence of coronary sclerosis at angiogram[nonobstructive coronary artery disease (noCAD) and normal coronary arteries (NCA)]), after exclusion of myocarditis and Takotsubo syndrome. RESULTS Out of 13 669 angiographic studies, 3695 were diagnosed with acute coronary syndrome; of these, 244 patients presented MINOCA (6.6%). Patients with noCAD were more likely to be older (67.9 vs. 59.2 years, P < 0.001) with higher prevalence of traditional cardiovascular risk factors (hypertension 64.1 vs. 41.2%, P = 0.002; diabetes 19.7 vs. 10.8%, P = 0.036; hypercholesterolemia 36.6 vs. 23.5%, P = 0.037). On surface ECG, anterior ST- segment elevation was more frequent in NCA patients (13.7 vs. 5.0%, P = 0.016). Secondary prevention therapy was significantly more prescribed in noCAD compared with NCA patients (acetylsalicylic acid 68.3 vs. 21.6%, P less than 0.001; statins 76.1 vs. 22.5%, P less than 0.001; angiotensin-converting enzyme inhibitor-AT1 blockers 51.4 vs. 31.3%, P = 0.006). One-year mortality was very low (0.4% for noCAD patients). CONCLUSION noCAD patients were older, with higher prevalence of cardiovascular risk factors and more frequently discharged with secondary prevention therapy. NCA patients presented more frequently anterior ST- segment elevation. Further diagnostic tests should be highly recommended to determine the underlying mechanism of MINOCA.
Collapse
Affiliation(s)
- Jan Dal Fabbro
- Department of Cardiology, University Heart Center, University Hospital Zurich
| | - Alessandro Candreva
- Department of Cardiology, University Heart Center, University Hospital Zurich
| | - Valentina A Rossi
- Department of Cardiology, University Heart Center, University Hospital Zurich
| | - Mohammady Shahin
- Department of Cardiology, University Heart Center, University Hospital Zurich
| | - Nooraldaem Yousif
- Department of Cardiology, University Heart Center, University Hospital Zurich
| | - Thomas F Lüscher
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Zurich, Switzerland.,Royal Brompton and Harefield Hospitals Trust and Imperial College, London, UK
| | - Firat Duru
- Department of Cardiology, University Heart Center, University Hospital Zurich.,Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Andrea Denegri
- Department of Cardiology, University Heart Center, University Hospital Zurich.,Division of Cardiology, Policlinico di Modena, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
43
|
Niculet E, Chioncel V, Elisei AM, Miulescu M, Buzia OD, Nwabudike LC, Craescu M, Draganescu M, Bujoreanu F, Marinescu E, Arbune M, Radaschin DS, Bobeica C, Nechita A, Tatu AL. Multifactorial expression of IL-6 with update on COVID-19 and the therapeutic strategies of its blockade (Review). Exp Ther Med 2021; 21:263. [PMID: 33603870 PMCID: PMC7851683 DOI: 10.3892/etm.2021.9693] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 11/20/2020] [Indexed: 02/07/2023] Open
Abstract
Interleukin 6 (IL-6), a cytokine produced by various cells of the human body (macrophages, lymphocytes, astrocytes, ischemic myocytes, endothelial cells) has both pro-inflammatory and anti-inflammatory properties, being a key component in regulating various physiologic and pathological processes. The structure of this molecule and the receptor system it possesses are important due to the different activities that IL-6 can exert; through trans-signaling pro-inflammatory activities are mediated, while through classic signaling, IL-6 is responsible for anti-inflammatory and regenerative activities. IL-6 signaling is involved in coronary artery disease and the global COVID-19 pandemic. This proatherogenic cytokine reaches elevated serum levels in the cytokine storm generated by SARS-CoV-2, and is also associated with smoking or obesity-classic cardiovascular risk factors which promote inflammatory states. IL-6 levels are proportionally correlated with dyslipidemia, hypertension and glucose dysregulation, and they are associated with poor outcomes in patients with unstable angina or acute myocardial infarction. IL-6 targeting for treatment development (not only) in cardiovascular disease and COVID-19 is still a matter of ongoing research, although tocilizumab has proven to be effective in reducing the proatherogenic effects of IL-6 and is suggested to improve COVID-19 patient survival.
Collapse
Affiliation(s)
- Elena Niculet
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, 'Dunărea de Jos' University, 800010 Galati, Romania
| | - Valentin Chioncel
- Cardio-Thoracic Department, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Clinical Cardiology Department, 'Bagdasar Arseni' Emergency Hospital, 041915 Bucharest, Romania
| | - Alina M Elisei
- Department of Pharmaceutical Sciences, Faculty of Medicine and Pharmacy, 'Dunărea de Jos' University, 800010 Galati, Romania.,Research Center in The Field of Medical and Pharmaceutical Sciences, ReFORM-UDJ, 'Dunărea de Jos' University, 800010 Galati, Romania
| | - Magdalena Miulescu
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, 'Dunărea de Jos' University, 800010 Galati, Romania
| | - Olimpia D Buzia
- Department of Pharmaceutical Sciences, Faculty of Medicine and Pharmacy, 'Dunărea de Jos' University, 800010 Galati, Romania.,Research Center in The Field of Medical and Pharmaceutical Sciences, ReFORM-UDJ, 'Dunărea de Jos' University, 800010 Galati, Romania
| | - Lawrence C Nwabudike
- Department of Diabetic Foot Care, 'Prof. N. Paulescu' National Institute of Diabetes, 011233 Bucharest, Romania
| | - Mihaela Craescu
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, 'Dunărea de Jos' University, 800010 Galati, Romania
| | - Miruna Draganescu
- Clinical Department, Faculty of Medicine and Pharmacy, 'Dunărea de Jos' University, 800010 Galati, Romania
| | - Florin Bujoreanu
- Department of Dermatology, 'Sf. Cuvioasa Parascheva' Clinical Hospital of Infectious Diseases, 800179 Galati, Romania
| | - Elisabeta Marinescu
- Department of Pharmaceutical Sciences, Faculty of Medicine and Pharmacy, 'Dunărea de Jos' University, 800010 Galati, Romania
| | - Manuela Arbune
- Centre of Medical-Pharmaceutical Research, 'Dunărea de Jos' University, 800010 Galati, Romania
| | - Diana Sabina Radaschin
- Research Center in The Field of Medical and Pharmaceutical Sciences, ReFORM-UDJ, 'Dunărea de Jos' University, 800010 Galati, Romania
| | - Carmen Bobeica
- Department of Dermato-Venereology, Doctoral School, University of Medicine and Pharmacy 'Gr. T. Popa', 700115 Iași, Romania
| | - Aurel Nechita
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, 'Dunărea de Jos' University, 800010 Galati, Romania.,Department of Pediatrics, 'Sf. Ioan' Clinical Hospital for Children, 800487 Galati, Romania
| | - Alin L Tatu
- Research Center in The Field of Medical and Pharmaceutical Sciences, ReFORM-UDJ, 'Dunărea de Jos' University, 800010 Galati, Romania.,Clinical Department, Faculty of Medicine and Pharmacy, 'Dunărea de Jos' University, 800010 Galati, Romania.,Department of Dermatology, 'Sf. Cuvioasa Parascheva' Clinical Hospital of Infectious Diseases, 800179 Galati, Romania
| |
Collapse
|
44
|
Song J, Frieler RA, Whitesall SE, Chung Y, Vigil TM, Muir LA, Ma J, Brombacher F, Goonewardena SN, Lumeng CN, Goldstein DR, Mortensen RM. Myeloid interleukin-4 receptor α is essential in postmyocardial infarction healing by regulating inflammation and fibrotic remodeling. Am J Physiol Heart Circ Physiol 2021; 320:H323-H337. [PMID: 33164548 PMCID: PMC7847075 DOI: 10.1152/ajpheart.00251.2020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 10/23/2020] [Accepted: 10/29/2020] [Indexed: 02/06/2023]
Abstract
Interleukin-4 receptor α (IL4Rα) signaling plays an important role in cardiac remodeling during myocardial infarction (MI). However, the target cell type(s) of IL4Rα signaling during this remodeling remains unclear. Here, we investigated the contribution of endogenous myeloid-specific IL4Rα signaling in cardiac remodeling post-MI. We established a murine myeloid-specific IL4Rα knockout (MyIL4RαKO) model with LysM promoter-driven Cre recombination. Macrophages from MyIL4RαKO mice showed significant downregulation of alternatively activated macrophage markers but an upregulation of classical activated macrophage markers both in vitro and in vivo, indicating the successful inactivation of IL4Rα signaling in macrophages. To examine the role of myeloid IL4Rα during MI, we subjected MyIL4RαKO and littermate floxed control (FC) mice to MI. We found that cardiac function was significantly impaired as a result of myeloid-specific IL4Rα deficiency. This deficiency resulted in a dysregulated inflammatory response consisting of decreased production of anti-inflammatory cytokines. Myeloid IL4Rα deficiency also led to reduced collagen 1 deposition and an imbalance of matrix metalloproteinases (MMPs)/tissue inhibitors of metalloproteinases (TIMPs), with upregulated MMPs and downregulated TIMPs, which resulted in insufficient fibrotic remodeling. In conclusion, this study identifies that myeloid-specific IL4Rα signaling regulates inflammation and fibrotic remodeling during MI. Therefore, myeloid-specific activation of IL4Rα signaling could offer protective benefits after MI.NEW & NOTEWORTHY This study showed, for the first time, the role of endogenous IL4Rα signaling in myeloid cells during cardiac remodeling and the underlying mechanisms. We identified myeloid cells are the critical target cell types of IL4Rα signaling during cardiac remodeling post-MI. Deficiency of myeloid IL4Rα signaling causes deteriorated cardiac function post-MI, due to dysregulated inflammation and insufficient fibrotic remodeling. This study sheds light on the potential of activating myeloid-specific IL4Rα signaling to modify remodeling post-MI. This brings hope to patients with MI and diminishes side effects by cell type-specific instead of whole body treatment.
Collapse
Affiliation(s)
- Jianrui Song
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan
| | - Ryan A Frieler
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Steven E Whitesall
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Yutein Chung
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Thomas M Vigil
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Lindsey A Muir
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan
| | - Jun Ma
- Department of Thoracic Surgery, Shanxi Province People's Hospital, Taiyuan, People's Republic of China
| | - Frank Brombacher
- International Center for Genetic Engineering and Biotechnology, University of Cape Town, Cape Town, South Africa
| | - Sascha N Goonewardena
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Carey N Lumeng
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan
| | - Daniel R Goldstein
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Institute of Gerontology, University of Michigan, Ann Arbor, Michigan
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan
| | - Richard M Mortensen
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
45
|
Benotmane I, Perrin P, Vargas GG, Bassand X, Keller N, Lavaux T, Ohana M, Bedo D, Baldacini C, Sagnard M, Bozman DF, Chiesa MD, Cognard N, Olagne J, Delagreverie H, Marx D, Heibel F, Braun L, Moulin B, Fafi-Kremer S, Caillard S. Biomarkers of Cytokine Release Syndrome Predict Disease Severity and Mortality From COVID-19 in Kidney Transplant Recipients. Transplantation 2021; 105:158-169. [PMID: 33009284 DOI: 10.1097/tp.0000000000003480] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Data on coronavirus disease 2019 (COVID-19) in immunocompromised kidney transplant recipients (KTR) remain scanty. Although markers of inflammation, cardiac injury, and coagulopathy have been previously associated with mortality in the general population of patients with COVID-19, their prognostic impact amongst KTR with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection has not been specifically investigated. METHODS We conducted a cohort study of 49 KTR who presented with COVID-19. Clinical and laboratory risk factors for severe disease and mortality were prospectively collected and analyzed with respect to outcomes. The study participants were divided into 3 groups: (1) mild disease manageable in an outpatient setting (n = 8), (2) nonsevere disease requiring hospitalization (n = 21), and (3) severe disease (n = 20). RESULTS Gastrointestinal manifestations were common at diagnosis. The 30-day mortality rate in hospitalized patients was 19.5%. Early elevations of C-reactive protein (>100 mg/L) and interleukin-6 (>65 ng/L) followed by increases in high-sensitivity troponin I (>30 ng/L) and D-dimer (>960 ng/mL) were significantly associated with severe disease and mortality. Viral load did not have prognostic significance in our sample, suggesting that outcomes were chiefly driven by a cytokine release syndrome (CRS). CONCLUSIONS Regular monitoring of CRS biomarkers in KTR with COVID-19 is paramount to improve clinical outcomes.
Collapse
Affiliation(s)
- Ilies Benotmane
- Department of Nephrology and Transplantation, University Hospital, Strasbourg, France
- Department of Virology, Strasbourg University Hospital, Strasbourg, France
- INSERM U1109, LabEx TRANSPLANTEX, Strasbourg, France
| | - Peggy Perrin
- Department of Nephrology and Transplantation, University Hospital, Strasbourg, France
- INSERM U1109, LabEx TRANSPLANTEX, Strasbourg, France
| | | | - Xavier Bassand
- Department of Nephrology and Transplantation, University Hospital, Strasbourg, France
| | - Nicolas Keller
- Department of Nephrology and Dialysis, University Hospital, Strasbourg, France
| | - Thomas Lavaux
- Department of Biochemistry and Molecular Biology, University Hospital, Strasbourg, France
| | - Mickael Ohana
- Department of Radiology, University Hospital, Strasbourg, France
| | - Dimitri Bedo
- Department of Nephrology and Transplantation, University Hospital, Strasbourg, France
| | - Clément Baldacini
- Department of Nephrology and Transplantation, University Hospital, Strasbourg, France
| | - Mylene Sagnard
- Department of Nephrology and Transplantation, University Hospital, Strasbourg, France
| | - Dogan-Firat Bozman
- Department of Nephrology and Transplantation, University Hospital, Strasbourg, France
| | - Margaux Della Chiesa
- Department of Nephrology and Transplantation, University Hospital, Strasbourg, France
| | - Noëlle Cognard
- Department of Nephrology and Transplantation, University Hospital, Strasbourg, France
| | - Jérôme Olagne
- Department of Nephrology and Transplantation, University Hospital, Strasbourg, France
| | | | - David Marx
- Department of Nephrology and Transplantation, University Hospital, Strasbourg, France
| | - Françoise Heibel
- Department of Nephrology and Transplantation, University Hospital, Strasbourg, France
| | - Laura Braun
- Department of Nephrology and Transplantation, University Hospital, Strasbourg, France
| | - Bruno Moulin
- Department of Nephrology and Transplantation, University Hospital, Strasbourg, France
- INSERM U1109, LabEx TRANSPLANTEX, Strasbourg, France
| | - Samira Fafi-Kremer
- Department of Nephrology and Transplantation, University Hospital, Strasbourg, France
- INSERM U1109, LabEx TRANSPLANTEX, Strasbourg, France
| | - Sophie Caillard
- Department of Nephrology and Transplantation, University Hospital, Strasbourg, France
- INSERM U1109, LabEx TRANSPLANTEX, Strasbourg, France
| |
Collapse
|
46
|
Fernández-Mestre M, Salazar-Alcalá E, Matos-González G, Márquez I. Polimorfismo de genes de citocinas: ¿factores de riesgo cardiovascular en la población venezolana? ARCHIVOS DE CARDIOLOGIA DE MEXICO 2020; 91:281-288. [PMID: 33075041 PMCID: PMC8351660 DOI: 10.24875/acm.200003301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/20/2020] [Indexed: 11/17/2022] Open
Abstract
Objective To examine whether the polymorphisms of the IL6, TNFA and IL10 genes represent a risk marker for acute myocardial infarction (AMI) and to analyze their correlation with risk factors, age of occurrence and type of AMI. Method Association study that included 310 unrelated Venezuelan individuals, grouped in 190 patients with AMI and 120 controls with or without cardiovascular risk factors. The IL6-174 G/C (rs1800795), TNFA -308 G/A (rs1800629), and IL10-1082 A/G (rs1800896), -819 C/T (rs1800871) and -592 C/A (rs1800872) polymorphisms were determined using the polymerase chain reaction technique with sequence-specific primers. Results Comparison of genotypic and allelic frequencies, using adjusted logistic regression analysis for risk factors, showed a significantly increased frequency of the genotype combination G/G-A/A of TNFA-308 G/A (odds ratio [OR]: 3.8; p = 0.00007), GG/-C/C of IL6-174 G/C (OR: 2.3; p = 0.009), A/G-G/G of IL10-1082 A/G (OR: 3.8; p = 0.00001) and the GCC haplotype of IL10 (OR: 3.71; p = 0.0053) in infarcted patients compared to controls. Interactions between the IL10-1082 A/G and TNFA-308 G/A polymorphisms and hypertension were observed. Conclusions The association of the variants of the TNFA, IL6 and IL10 genes with AMI suggest that the imbalance in the production of cytokines promotes an exacerbated inflammatory process, supporting the fundamental role of inflammation in all stages of the atherosclerotic process.
Collapse
Affiliation(s)
- Mercedes Fernández-Mestre
- Sección Inmunogenética, Laboratorio de Fisiopatología, Centro de Medicina Experimental Miguel Layrisse, Instituto Venezolano de Investigaciones Científicas
| | - Eva Salazar-Alcalá
- Sección Inmunogenética, Laboratorio de Fisiopatología, Centro de Medicina Experimental Miguel Layrisse, Instituto Venezolano de Investigaciones Científicas
| | - Gelly Matos-González
- Unidad de Cardiología, Hospital General del Este Dr. Domingo Luciani. Caracas, Venezuela
| | - Ingrid Márquez
- Unidad de Cardiología, Hospital General del Este Dr. Domingo Luciani. Caracas, Venezuela
| |
Collapse
|
47
|
Li H, Cen K, Sun W, Feng B. Predictive Value of Blood Interleukin-6 Level in Patients with Acute Coronary Syndrome: A Meta-analysis. Immunol Invest 2020; 50:964-976. [PMID: 32811241 DOI: 10.1080/08820139.2020.1795876] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Conflicting results have been reported on the association between blood level of interleukin-6 and adverse outcomes in patients with acute coronary syndrome (ACS). The current meta-analysis aimed to evaluate the predictive utility of elevated blood interleukin-6 level in patients with ACS. METHODS A systematically literature search was performed using PubMed and Embase databases up to December 31, 2019. Observational studies or post hoc analysis of randomized controlled trials investigating the values of blood interleukin-6 level for predicting major adverse cardiovascular events (MACE including death, re-infarction, revascularization, angina, heart failure, malignant arrhythmia, or stroke), all-cause mortality or cardiovascular mortality in ACS patients were eligible. The predictive values were summarized by pooling the multivariable-adjusted risk ratio (RR) and 95% confidence intervals (CI) for the highest versus lowest category of interleukin-6 level. RESULTS Thirteen studies enrolling 30,289 patients with ACS were included. When comparing the highest with lowest category of interleukin-6 level, the pooled RR was 1.29 (95% CI 1.12-1.48) for MACE, 1.50 (95% CI 1.35-1.67) for all-cause mortality, and 1.55 (95% CI 1.06-2.28) for cardiovascular mortality, respectively. Moreover, the predictive values of interleukin-6 level on MACE were consistently found in different study designs, subtypes of patients, sample sizes, follow-up duration, and cutoff value of interleukin-6 elevation subgroups. CONCLUSION Increased blood level of interleukin-6may be independently associated with higher risk of MACE, cardiovascular and all-cause mortality in patients with ACS. Measurement of blood interleukin-6 level has potential to improve risk stratification of ACS.
Collapse
Affiliation(s)
- Hengdong Li
- Department of Cardiology, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China.,Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | - Kaidong Cen
- Department of Cardiology, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Weifeng Sun
- Department of Cardiology, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Beili Feng
- Department of Cardiology, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China.,Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| |
Collapse
|
48
|
Ljuca F, Hadžiefendić B, Jahić E, Tihić N, Lukić S. Pentraxin 3 might be better prognostic serum marker than IL-6, IL-10, and high-sensitivity C-reactive protein for major adverse cardiovascular events in patients with ST-elevation myocardial infarction after bare-metal stent implantation. Saudi Med J 2020; 40:1202-1208. [PMID: 31828271 PMCID: PMC6969622 DOI: 10.15537/smj.2019.12.24737] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Objectives: To assess the prognostic value of pentraxin 3 (PTX3) in patients with ST-elevation myocardial infarction (STEMI) after bare-metal stent (BMS) implantation. Methods: In this prospective study, PTX3, interleukin (IL-6), IL-10, high-sensitivity c-reactive protein (hsCRP), and cardiac troponin I (cTnI) plasma values were determined before and 24hours after BMS implantation in 97 consecutively enrolled patients with STEMI who were admitted to University Clinical Center Tuzla, Tuzla, Bosnia and Herzegovina between February 2016 and February 2017. Patients were followed for 24 months to assess major adverse cardiovascular events (MACEs). Results: At 24 hours after percutaneous coronary intervention (PCI), plasma values of PTX3, IL-6, hsCRP, and cTnI were significantly increased; and IL-10 levels were significantly decreased compared with the values determined before PCI. Patients with MACEs had significantly higher plasma PTX3 levels at 24 hours after BMS-PCI than in patients without MACEs. Patients with PTX3 plasma values ≥5042 ng/ml had a significantly higher risk of MACEs than patients with PTX3 levels <5.042 ng/mL. Pentraxin 3 levels exhibited strong and significant correlations with IL-6 and IL-10 levels. Pentraxin 3, cTnI, and IL-6, but not hsCRP levels have showed independent association with MACEs, according to the multivariate Cox regression analysis. Conclusion: Pentraxin 3 might be better serum prognostic marker than IL-6, IL-10 or high sensitivity CRP for MACEs after BMS-PCI. It might help to make better risk stratification of those patients who are undergoing BMS-PCI.
Collapse
Affiliation(s)
- Farid Ljuca
- Department of Physiology, School of Medicine, University of Tuzla, Tuzla, Bosnia and Herzegovina. E-mail.
| | | | | | | | | |
Collapse
|
49
|
Reindl M, Tiller C, Holzknecht M, Lechner I, Eisner D, Riepl L, Pamminger M, Henninger B, Mayr A, Schwaiger JP, Klug G, Bauer A, Metzler B, Reinstadler SJ. Global longitudinal strain by feature tracking for optimized prediction of adverse remodeling after ST-elevation myocardial infarction. Clin Res Cardiol 2020; 110:61-71. [PMID: 32296969 DOI: 10.1007/s00392-020-01649-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 04/08/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND The role of left ventricular (LV) myocardial strain by cardiac magnetic resonance feature tracking (CMR-FT) for the prediction of adverse remodeling following ST-elevation myocardial infarction (STEMI), as well as its prognostic validity compared to LV ejection fraction (LVEF) and CMR infarct severity parameters, is unclear. This study aimed to evaluate the independent and incremental value of LV strain by CMR-FT for the prediction of adverse LV remodeling post-STEMI. METHODS STEMI patients treated with primary percutaneous coronary intervention were enrolled in this prospective observational study. CMR core laboratory analysis was performed to assess LVEF, infarct pathology and LV myocardial strain. The primary endpoint was adverse remodeling, defined as ≥ 20% increase in LV end-diastolic volume from baseline to 4 months. RESULTS From the 232 patients included, 38 (16.4%) reached the primary endpoint. Global longitudinal strain (GLS), global radial strain, and global circumferential strain were all predictive of adverse remodeling (p < 0.01 for all), but only GLS was an independent predictor of adverse remodeling (odds ratio: 1.36[1.03-1.78]; p = 0.028) after adjustment for strain parameters, LVEF and CMR markers of infarct severity. A GLS > - 14% was associated with a fourfold increase in the risk for LV remodeling (odds ratio: 4.16[1.56-11.13]; p = 0.005). Addition of GLS to a baseline model comprising LVEF, infarct size and microvascular obstruction resulted in net reclassification improvement of 0.26 ([0.13-0.38]; p < 0.001) and integrated discrimination improvement of 0.02 ([0.01-0.03]; p = 0.006). CONCLUSIONS In STEMI survivors, determination of GLS using CMR-FT provides important prognostic information for the development of adverse remodeling that is incremental to LVEF and CMR markers of infarct severity. CLINICAL TRIAL REGISTRATION NCT04113356.
Collapse
Affiliation(s)
- Martin Reindl
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Christina Tiller
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Magdalena Holzknecht
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Ivan Lechner
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Dorothea Eisner
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Laura Riepl
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Mathias Pamminger
- University Clinic of Radiology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Benjamin Henninger
- University Clinic of Radiology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Agnes Mayr
- University Clinic of Radiology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Johannes P Schwaiger
- Department of Internal Medicine, Academic Teaching Hospital Hall in Tirol, Innsbruck, Austria
| | - Gert Klug
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Axel Bauer
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Bernhard Metzler
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Sebastian J Reinstadler
- University Clinic of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Anichstrasse 35, 6020, Innsbruck, Austria.
| |
Collapse
|
50
|
Pawluk H, Woźniak A, Grześk G, Kołodziejska R, Kozakiewicz M, Kopkowska E, Grzechowiak E, Kozera G. The Role of Selected Pro-Inflammatory Cytokines in Pathogenesis of Ischemic Stroke. Clin Interv Aging 2020; 15:469-484. [PMID: 32273689 PMCID: PMC7110925 DOI: 10.2147/cia.s233909] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/13/2020] [Indexed: 12/29/2022] Open
Abstract
Stroke is currently one of the most common causes of death and disability in the world, and its pathophysiology is a complex process, involving the oxidative stress and inflammatory reaction. Unfortunately, no biochemical factors useful in the diagnostics and treatment of stroke have been clearly established to date. Therefore, researchers are increasingly interested in the inflammatory response triggered by cerebral ischemia and its role in the development of cerebral infarction. This article gives an overview of the available literature data concerning the role of pro-inflammatory cytokines in acute stroke. Detailed analysis of their role in cerebral circulation disturbances can also suggest certain immune response regulatory mechanisms aimed to reduce damage to the nervous tissue in the course of stroke.
Collapse
Affiliation(s)
- Hanna Pawluk
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Alina Woźniak
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Grzegorz Grześk
- 2nd Department of Cardiology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Renata Kołodziejska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Mariusz Kozakiewicz
- Department of Geriatrics, Division of Biochemistry and Biogerontology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Ewa Kopkowska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Elżbieta Grzechowiak
- Department of Neurology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Grzegorz Kozera
- Department of Neurology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
- Medical Stimulation Center, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|