1
|
Sun W, Chen Z, Luo Y. Association Between Systemic Immune-Inflammation Index and Outcomes of Acute Myocardial Infarction: A Systemic Review and Meta-Analysis. Surg Infect (Larchmt) 2025; 26:183-194. [PMID: 39699344 DOI: 10.1089/sur.2024.172] [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] [Indexed: 12/20/2024] Open
Abstract
Objective: To assess the link between systemic immune-inflammation index (SII) and risk of major adverse cardiovascular events (MACE), contrast-induced nephropathy (CIN), and overall mortality in patients with acute myocardial infarction (AMI). Patients and Methods: Electronic search of PubMed, EMBASE, Web of Science, and Scopus databases was done for observational studies with the data on the association of SII and outcomes, such as MACE, and CIN in adult (≥18 y) patients with AMI. A random-effects model was used, and the pooled effect sizes were expressed as relative risk (RR) with corresponding 95% confidence intervals (CI). Subgroup analysis was conducted on the basis of the type of AMI (ST elevation myocardial infarction and non-ST elevation myocardial infarction), sample size (≥500 and <500), and study design. GRADE assessment was used to evaluate the certainty of the evidence. Results: The analysis included 23 studies. Most studies were conducted in China (n = 13), followed by Turkey (n = 10). Majority of the studies (n = 20) had a retrospective cohort design. Patients with high SII had increased risk of MACE (RR 2.95, 95% CI: 1.25, 6.99; n = 5, I2 = 97.5%), overall mortality (RR 2.59, 95% CI: 1.64, 4.07; n = 6, I2 = 58.0%), and CIN (RR 4.58, 95% CI: 3.44, 6.10; n = 4, I2 = 0.0%), compared with patients with lower SII. Egger's test detected publication bias for MACE (p = 0.047) and overall mortality (p = 0.012) but not for CIN. These associations remained valid in subgroup analysis. Conclusion: Findings suggest that higher SII in patients with AMI is associated with increased risks of MACE, CIN, and overall mortality. This underscores SII's potential as a prognostic marker in AMI.
Collapse
Affiliation(s)
- Wen Sun
- EICU, Changxing People's Hospital of Zhejiang, Huzhou City, China
| | - Zheye Chen
- Department of Emergency, Changxing People's Hospital of Zhejiang, Huzhou City, China
| | - Yi Luo
- EICU, Changxing People's Hospital of Zhejiang, Huzhou City, China
| |
Collapse
|
2
|
Bećirović E, Bećirović M, Ljuca K, Babić M, Bećirović A, Ljuca N, Babić Jušić Z, Abdić A, Buljubašić L, Begagić E. The Inflammatory Burden in Heart Failure: A Cohort Study on Potential Biomarkers in Heart Failure With Reduced and Mildly Reduced Ejection Fraction. Cureus 2025; 17:e80159. [PMID: 40190877 PMCID: PMC11972061 DOI: 10.7759/cureus.80159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2025] [Indexed: 04/09/2025] Open
Abstract
Background Heart failure (HF) is characterized by impaired cardiac function. Based on left ventricular ejection fraction (LVEF), it is classified into HF with reduced ejection fraction (HFrEF), mildly reduced ejection fraction (HFmrEF), and preserved ejection fraction (HFpEF). Each phenotype has distinct pathophysiological mechanisms and clinical features. Recent findings indicate that systemic inflammation is a significant factor in the progression of heart failure. Inflammatory biomarkers, including neutrophil-to-lymphocyte ratio (NLR), monocyte-to-lymphocyte ratio (MLR), and lymphocyte-to-monocyte ratio (LMR), may serve as valuable tools for evaluating the inflammatory response in heart failure. Materials and methods This prospective observational study, which included 171 HF patients, was conducted from February 2022 to January 2023 at the Intensive Care Unit, University Clinical Centre Tuzla. Based on LVEF, patients were categorized into HFrEF, HFmrEF, and a control group (HFpEF). The study aimed to assess the prognostic value of NLR, MLR, and LMR in predicting major adverse cardiovascular events (MACE) and mortality over a 12-month follow-up period. Results NLR and MLR were significantly higher, while LMR was lower in both HFrEF and HFmrEF compared to controls, indicating a strong inflammatory response, particularly in HFrEF. NLR demonstrated a strong ability to distinguish between HF phenotypes. HFmrEF's markedly higher high-sensitivity troponin I (hsTroponin I) level suggested higher cardiac stress. MACE rates were similar across groups; mortality was significantly higher in HFrEF. Conclusion Inflammatory biomarkers NLR, MLR, LMR, and hsTroponin I could be crucial in assessing heart failure, particularly in patients with HFrEF and HFmrEF.
Collapse
Affiliation(s)
- Emir Bećirović
- Internal Medicine Clinic, University Clinical Centre Tuzla, Tuzla, BIH
| | - Minela Bećirović
- Internal Medicine Clinic, University Clinical Centre Tuzla, Tuzla, BIH
| | - Kenana Ljuca
- Gynecology and Obstetrics, University Clinical Centre Ljubljana, Ljubljana, SVN
| | - Mirza Babić
- Department of Internal Medicine, Cantonal Hospital Bihać, Bihać, BIH
| | - Amir Bećirović
- Internal Medicine Clinic, University Clinical Centre Tuzla, Tuzla, BIH
| | - Nadina Ljuca
- School of Medicine, University of Tuzla, Tuzla, BIH
| | | | - Admir Abdić
- Department of Surgery, Cantonal Hospital Bihać, Bihać, BIH
| | | | - Emir Begagić
- Department of General Medicine, School of Medicine, University of Zenica, Zenica, BIH
| |
Collapse
|
3
|
Xie S, Zhu X, Han F, Wang S, Cui K, Xue J, Xi X, Shi C, Li S, Wang F, Tian J. Discussion on the comparison of Raman spectroscopy and cardiovascular disease-related imaging techniques and the future applications of Raman technology: a systematic review. Lasers Med Sci 2025; 40:116. [PMID: 39988624 PMCID: PMC11847755 DOI: 10.1007/s10103-025-04315-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Accepted: 01/23/2025] [Indexed: 02/25/2025]
Abstract
Cardiovascular disease (CVD) is a major cause of unnatural death worldwide, so timely diagnosis of CVD is crucial for improving patient outcomes. Although the traditional diagnostic tools can locate plaque and observe inner wall of blood vessel structure, they commonly have radioactivity and cannot detect the chemical composition of the plaque accurately. Recently emerging Raman techniques can detect the plaque composition precisely, and have the advantages of being fast, high-resolution and marker-free. This makes Raman have great potential for detecting blood samples, understanding disease conditions, and real-time monitoring. This review summarizes the origin and state-of-art of Raman techniques, including the following aspects: (a) the principle and technical classification of Raman techniques; (b) the applicability of Raman techniques and its comparison with traditional diagnostic tools at different diagnosis targets; (c) the applicability of Raman spectroscopy in advanced CVD. Lastly, we highlight the possible future applications of Raman techniques in CVD diagnosis.
Collapse
Affiliation(s)
- Songcai Xie
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaotong Zhu
- Wuhan National Laboratory for Optoelectronics, Hua zhong Univeresity of Science and Technology, Wuhan, China
| | - Feiyuan Han
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shengyuan Wang
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Kexin Cui
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jing Xue
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangwen Xi
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chengyu Shi
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuo Li
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fan Wang
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China.
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Jinwei Tian
- Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, China.
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
4
|
Zgutka K, Tkacz M, Tomasiak P, Piotrowska K, Ustianowski P, Pawlik A, Tarnowski M. Gestational Diabetes Mellitus-Induced Inflammation in the Placenta via IL-1β and Toll-like Receptor Pathways. Int J Mol Sci 2024; 25:11409. [PMID: 39518962 PMCID: PMC11546908 DOI: 10.3390/ijms252111409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Gestational diabetes mellitus is characterised by an insufficient insulin response to hyperglycaemia and the development of insulin resistance. This state has adverse effects on the health outcomes of the mother and child. Existing hyperglycaemia triggers a state of inflammation that involves several tissues, including the placenta. In this study, we analysed the putative pathomechanism of GDM, with special emphasis on the role of chronic, sterile, pro-inflammatory pathways. The expression and regulation of the elements of IL-1β and Toll-like receptor (TLR) pathways in GDM maternal blood plasma, healthy placental explants and a choriocarcinoma cell line (BeWo cell line) stimulated with pro-inflammatory factors was evaluated. Our results indicate elevated expression of the IL-1β and TLR pathways in GDM patients. After stimulation with IL-1β or LPS, the placental explants and BeWo cell line showed increased production of pro-inflammatory IL-6, TNFa and IL-1β together with increased expression of the elements of the signalling pathways. The application of selected inhibitors of NF-ĸB, MAPK and recombinant interleukin 1 receptor antagonist (IL1RA) proved the key involvement of the IL-1β pathway and TLRs in the pathogenesis of GDM. Our results show the possible existence of loops of autocrine stimulation and a possible inflammatory pathomechanism in placentas affected by GDM.
Collapse
Affiliation(s)
- Katarzyna Zgutka
- Department of Physiology in Health Sciences, Faculty of Health Sciences, Pomeranian Medical University, 70-210 Szczecin, Poland
| | - Marta Tkacz
- Department of Physiology in Health Sciences, Faculty of Health Sciences, Pomeranian Medical University, 70-210 Szczecin, Poland
| | - Patrycja Tomasiak
- Institute of Physical Culture Sciences, University of Szczecin, 70-453 Szczecin, Poland
| | - Katarzyna Piotrowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Przemysław Ustianowski
- Department of Nursing, Faculty of Health Sciences, Pomeranian Medical University in Szczecin, 70-210 Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Maciej Tarnowski
- Department of Physiology in Health Sciences, Faculty of Health Sciences, Pomeranian Medical University, 70-210 Szczecin, Poland
| |
Collapse
|
5
|
Chen D, Lin Y, Ko P, Lin J, Huang C, Wang G, Chang KC. Effect of targeted temperature management on systemic inflammatory responses after out-of-hospital cardiac arrest: A prospective cohort study. Medicine (Baltimore) 2024; 103:e39780. [PMID: 39312301 PMCID: PMC11419506 DOI: 10.1097/md.0000000000039780] [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: 04/04/2024] [Accepted: 08/30/2024] [Indexed: 09/25/2024] Open
Abstract
BACKGROUND Interleukin (IL)-6 is a major inflammatory cytokine that predicts mortality after out-of-hospital cardiac arrest (OHCA). Targeted temperature management (TTM) is associated with improved all-cause mortality in patients with OHCA. However, the effect of TTM on IL-6 production remains unclear. This study investigated whether TTM has additional anti-inflammatory effects after OHCA. METHODS This prospective cohort study included a total of 141 hospitalized patients with OHCA who were treated between January 2015 and June 2023. The study was conducted in the intensive care unit of China Medical University Hospital, Taichung. Postcardiac arrest care included TTM or the control approach (no TTM). The primary outcomes included the 90-day mortality rate and neurologic outcomes after OHCA. Differences between the TTM and control groups were examined using Student t test, chi-square test, and Kaplan-Meier survival curve analysis. Multivariate analysis of variance model was used to examine interaction effects. RESULTS Plasma IL-6 and IL-6/soluble IL-6 receptor complex levels were measured at 6 and 24 hours after resuscitation. IL-6 and IL-6/soluble IL-6 receptor complex production was lower in the TTM group than in the control group (-50.0% vs +136.7%, P < .001; +26.3% vs +102.40%, P < .001, respectively). In addition, the 90-day mortality rate and poor neurologic outcomes were lower in the TTM group than in the control group (36.8% vs 63.0%, relative risk 0.39, 95% confidence interval 0.24-0.64, P < .001; 65.5% vs 81.5%, relative risk 0.80, 95% confidence interval 0.66-0.98, P = .04). CONCLUSION TTM improves both the mortality rate and neurologic outcomes in patients resuscitated from OHCA, possibly by reducing IL-6-induced proinflammatory responses.
Collapse
Affiliation(s)
- Dalong Chen
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Yukai Lin
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Poyen Ko
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | - Jenjyh Lin
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chihyang Huang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| | - Gueijane Wang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- Pharmacy Department, Wizcare Medical Corporation Aggregate, Taichung, Taiwan
- School of Medicine, Weifang University of Science and Technology, Weifang, Shandong, China
| | - Kuan-Cheng Chang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
6
|
Bordoni B, Mapelli L, Toccafondi A, Di Salvo F, Cannadoro G, Gonella M, Escher AR, Morici N. Post-Myocardial Infarction Rehabilitation: The Absence in the Rehabilitation Process of the Diaphragm Muscle. Int J Gen Med 2024; 17:3201-3210. [PMID: 39070222 PMCID: PMC11277820 DOI: 10.2147/ijgm.s470878] [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: 05/27/2024] [Accepted: 06/25/2024] [Indexed: 07/30/2024] Open
Abstract
Myocardial infarction (MI) is one of the leading causes of death worldwide. There can be many reasons that cause MI, such as a sedentary lifestyle, a disordered diet, harmful habits such as smoking and alcoholism, concomitant congenital or acquired systemic pathologies. Patients who survive the acute event suffer a functional alteration of multiple body systems. The various cardiology associations recommend starting a rehabilitation process, pursuing the main objective of improving the patient's health status. A negative consequence that can be linked to MI is the dysfunction of the main breathing muscle, the diaphragm. The diaphragm is essential not only for respiratory mechanisms but also for adequate production of cardiac pressures. Post-MI patients present a reduction in the performance of the diaphragm muscle, and this condition can become a risk factor for further relapses or for the onset of heart failure. The article reviews the rehabilitation path for post-MI patients, to highlight the absence given to the diaphragm in the recovery of the patient's health status. The text reviews the post-MI diaphragmatic adaptation to highlight the importance of including targeted training for the diaphragm muscle in the rehabilitation process.
Collapse
Affiliation(s)
- Bruno Bordoni
- Dipartimento di Cardiologia, Fondazione Don Carlo Gnocchi IRCCS, Istituto di Ricovero e Cura, S Maria Nascente, Milano, 20100, Italy
| | - Luca Mapelli
- Dipartimento di Cardiologia, Fondazione Don Carlo Gnocchi IRCCS, Istituto di Ricovero e Cura, S Maria Nascente, Milano, 20100, Italy
| | - Anastasia Toccafondi
- Dipartimento di Cardiologia, Fondazione Don Carlo Gnocchi IRCCS, Istituto di Ricovero e Cura, S Maria Nascente, Milano, 20100, Italy
| | - Francesca Di Salvo
- Dipartimento di Cardiologia, Fondazione Don Carlo Gnocchi IRCCS, Istituto di Ricovero e Cura, S Maria Nascente, Milano, 20100, Italy
| | - Gianmarco Cannadoro
- Dipartimento di Cardiologia, Fondazione Don Carlo Gnocchi IRCCS, Istituto di Ricovero e Cura, S Maria Nascente, Milano, 20100, Italy
| | - Matteo Gonella
- Dipartimento di Cardiologia, Fondazione Don Carlo Gnocchi IRCCS, Istituto di Ricovero e Cura, S Maria Nascente, Milano, 20100, Italy
| | - Allan R Escher
- Department of Anesthesiology/Pain Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Nuccia Morici
- Dipartimento di Cardiologia, Fondazione Don Carlo Gnocchi IRCCS, Istituto di Ricovero e Cura, S Maria Nascente, Milano, 20100, Italy
| |
Collapse
|
7
|
Amaro-Prellezo E, Gómez-Ferrer M, Hakobyan L, Ontoria-Oviedo I, Peiró-Molina E, Tarazona S, Salguero P, Ruiz-Saurí A, Selva-Roldán M, Vives-Sanchez R, Sepúlveda P. Extracellular vesicles from dental pulp mesenchymal stem cells modulate macrophage phenotype during acute and chronic cardiac inflammation in athymic nude rats with myocardial infarction. Inflamm Regen 2024; 44:25. [PMID: 38807194 PMCID: PMC11134765 DOI: 10.1186/s41232-024-00340-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND/AIMS Extracellular vesicles (EVs) derived from dental pulp mesenchymal stem cells (DP-MSCs) are a promising therapeutic option for the treatment of myocardial ischemia. The aim of this study is to determine whether MSC-EVs could promote a pro-resolving environment in the heart by modulating macrophage populations. METHODS EVs derived from three independent biopsies of DP-MSCs (MSC-EVs) were isolated by tangential flow-filtration and size exclusion chromatography and were characterized by omics analyses. Biological processes associated with these molecules were analyzed using String and GeneCodis platforms. The immunomodulatory capacity of MSC-EVs to polarize macrophages towards a pro-resolving or M2-like phenotype was assessed by evaluating surface markers, cytokine production, and efferocytosis. The therapeutic potential of MSC-EVs was evaluated in an acute myocardial infarction (AMI) model in nude rats. Infarct size and the distribution of macrophage populations in the infarct area were evaluated 7 and 21 days after intramyocardial injection of MSC-EVs. RESULTS Lipidomic, proteomic, and miRNA-seq analysis of MSC-EVs revealed their association with biological processes involved in tissue regeneration and regulation of the immune system, among others. MSC-EVs promoted the differentiation of pro-inflammatory macrophages towards a pro-resolving phenotype, as evidenced by increased expression of M2 markers and decreased secretion of pro-inflammatory cytokines. Administration of MSC-EVs in rats with AMI limited the extent of the infarcted area at 7 and 21 days post-infarction. MSC-EV treatment also reduced the number of pro-inflammatory macrophages within the infarct area, promoting the resolution of inflammation. CONCLUSION EVs derived from DP-MSCs exhibited similar characteristics at the omics level irrespective of the biopsy from which they were derived. All MSC-EVs exerted effective pro-resolving responses in a rat model of AMI, indicating their potential as therapeutic agents for the treatment of inflammation associated with AMI.
Collapse
Affiliation(s)
- Elena Amaro-Prellezo
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital La Fe, Avda. Fernando Abril Martorell 106, Valencia, 46026, Spain
| | - Marta Gómez-Ferrer
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital La Fe, Avda. Fernando Abril Martorell 106, Valencia, 46026, Spain
| | - Lusine Hakobyan
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital La Fe, Avda. Fernando Abril Martorell 106, Valencia, 46026, Spain
- Department of Analytical Chemistry, Faculty of Chemistry, University of Valencia, Valencia, 46100, Spain
| | - Imelda Ontoria-Oviedo
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital La Fe, Avda. Fernando Abril Martorell 106, Valencia, 46026, Spain
| | - Esteban Peiró-Molina
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital La Fe, Avda. Fernando Abril Martorell 106, Valencia, 46026, Spain
- Hospital Universitari I Politècnic La Fe, Valencia, 46026, Spain
| | - Sonia Tarazona
- Department of Applied Statistics and Operations Research and Quality, Universitat Politècnica de València, Valencia, 46022, Spain
| | - Pedro Salguero
- Department of Applied Statistics and Operations Research and Quality, Universitat Politècnica de València, Valencia, 46022, Spain
| | - Amparo Ruiz-Saurí
- Department of Pathology, University of Valencia, Valencia, 46010, Spain
| | - Marta Selva-Roldán
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital La Fe, Avda. Fernando Abril Martorell 106, Valencia, 46026, Spain
| | - Rosa Vives-Sanchez
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital La Fe, Avda. Fernando Abril Martorell 106, Valencia, 46026, Spain
| | - Pilar Sepúlveda
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital La Fe, Avda. Fernando Abril Martorell 106, Valencia, 46026, Spain.
- Hospital Universitari I Politècnic La Fe, Valencia, 46026, Spain.
- Department of Pathology, University of Valencia, Valencia, 46010, Spain.
- Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), III Institute of Health, Madrid, Carlos, Spain.
| |
Collapse
|
8
|
Ozawa K, Packwood W, Muller MA, Qi Y, Xie A, Varlamov O, McCarty OJ, Chung D, López JA, Lindner JR. Removal of endothelial surface-associated von villebrand factor suppresses accelerate datherosclerosis after myocardial infarction. J Transl Med 2024; 22:412. [PMID: 38693516 PMCID: PMC11062912 DOI: 10.1186/s12967-024-05231-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/23/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUND Thromboinflammation involving platelet adhesion to endothelial surface-associated von Willebrand factor (VWF) has been implicated in the accelerated progression of non-culprit plaques after MI. The aim of this study was to use arterial endothelial molecular imaging to mechanistically evaluate endothelial-associated VWF as a therapeutic target for reducing remote plaque activation after myocardial infarction (MI). METHODS Hyperlipidemic mice deficient for the low-density lipoprotein receptor and Apobec-1 underwent closed-chest MI and were treated chronically with either: (i) recombinant ADAMTS13 which is responsible for proteolytic removal of VWF from the endothelial surface, (ii) N-acetylcysteine (NAC) which removes VWF by disulfide bond reduction, (iii) function-blocking anti-factor XI (FXI) antibody, or (iv) no therapy. Non-ischemic controls were also studied. At day 3 and 21, ultrasound molecular imaging was performed with probes targeted to endothelial-associated VWF A1-domain, platelet GPIbα, P-selectin and vascular cell adhesion molecule-1 (VCAM-1) at lesion-prone sites of the aorta. Histology was performed at day 21. RESULTS Aortic signal for P-selectin, VCAM-1, VWF, and platelet-GPIbα were all increased several-fold (p < 0.01) in post-MI mice versus sham-treated animals at day 3 and 21. Treatment with NAC and ADAMTS13 significantly attenuated the post-MI increase for all four molecular targets by > 50% (p < 0.05 vs. non-treated at day 3 and 21). On aortic root histology, mice undergoing MI versus controls had 2-4 fold greater plaque size and macrophage content (p < 0.05), approximately 20-fold greater platelet adhesion (p < 0.05), and increased staining for markers of platelet transforming growth factor-β1 signaling. Accelerated plaque growth and inflammatory activation was almost entirely prevented by ADAMTS13 and NAC. Inhibition of FXI had no significant effect on molecular imaging signal or plaque morphology. CONCLUSIONS Plaque inflammatory activation in remote arteries after MI is strongly influenced by VWF-mediated platelet adhesion to the endothelium. These findings support investigation into new secondary preventive therapies for reducing non-culprit artery events after MI.
Collapse
Affiliation(s)
- Koya Ozawa
- Sydney Medical School Nepean, Faculty of Medicine and Health, Department of Cardiology, The University of Sydney, Nepean Hospital, Sydney, NSW, Australia
| | - William Packwood
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Matthew A Muller
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Yue Qi
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, USA
| | - Aris Xie
- Cardiovascular Division and Robert M. Berne Cardiovascular Research Center, University of Virginia, Box 801394, 415 Lane Rd, Charlottesville, VA, 22908, USA
| | - Oleg Varlamov
- Oregon National Primate Research Center, Portland, OR, USA
| | - Owen J McCarty
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, USA
| | - Dominic Chung
- BloodWorks Research Institute, University of Washington, Seattle, WA, USA
| | - José A López
- BloodWorks Research Institute, University of Washington, Seattle, WA, USA
| | - Jonathan R Lindner
- Cardiovascular Division and Robert M. Berne Cardiovascular Research Center, University of Virginia, Box 801394, 415 Lane Rd, Charlottesville, VA, 22908, USA.
| |
Collapse
|
9
|
Barrère-Lemaire S, Vincent A, Jorgensen C, Piot C, Nargeot J, Djouad F. Mesenchymal stromal cells for improvement of cardiac function following acute myocardial infarction: a matter of timing. Physiol Rev 2024; 104:659-725. [PMID: 37589393 DOI: 10.1152/physrev.00009.2023] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/05/2023] [Accepted: 08/16/2023] [Indexed: 08/18/2023] Open
Abstract
Acute myocardial infarction (AMI) is the leading cause of cardiovascular death and remains the most common cause of heart failure. Reopening of the occluded artery, i.e., reperfusion, is the only way to save the myocardium. However, the expected benefits of reducing infarct size are disappointing due to the reperfusion paradox, which also induces specific cell death. These ischemia-reperfusion (I/R) lesions can account for up to 50% of final infarct size, a major determinant for both mortality and the risk of heart failure (morbidity). In this review, we provide a detailed description of the cell death and inflammation mechanisms as features of I/R injury and cardioprotective strategies such as ischemic postconditioning as well as their underlying mechanisms. Due to their biological properties, the use of mesenchymal stromal/stem cells (MSCs) has been considered a potential therapeutic approach in AMI. Despite promising results and evidence of safety in preclinical studies using MSCs, the effects reported in clinical trials are not conclusive and even inconsistent. These discrepancies were attributed to many parameters such as donor age, in vitro culture, and storage time as well as injection time window after AMI, which alter MSC therapeutic properties. In the context of AMI, future directions will be to generate MSCs with enhanced properties to limit cell death in myocardial tissue and thereby reduce infarct size and improve the healing phase to increase postinfarct myocardial performance.
Collapse
Affiliation(s)
- Stéphanie Barrère-Lemaire
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Anne Vincent
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Christian Jorgensen
- Institute of Regenerative Medicine and Biotherapies, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Centre Hospitalier Universitaire Montpellier, Montpellier, France
| | - Christophe Piot
- Département de Cardiologie Interventionnelle, Clinique du Millénaire, Montpellier, France
| | - Joël Nargeot
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- LabEx Ion Channel Science and Therapeutics, Université de Nice, Nice, France
| | - Farida Djouad
- Institute of Regenerative Medicine and Biotherapies, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Montpellier, France
- Centre Hospitalier Universitaire Montpellier, Montpellier, France
| |
Collapse
|
10
|
Li X, Yu C, Liu X, Chen Y, Wang Y, Liang H, Qiu S, Lei L, Xiu J. A Prediction Model Based on Systemic Immune-Inflammatory Index Combined with Other Predictors for Major Adverse Cardiovascular Events in Acute Myocardial Infarction Patients. J Inflamm Res 2024; 17:1211-1225. [PMID: 38410422 PMCID: PMC10895983 DOI: 10.2147/jir.s443153] [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: 10/04/2023] [Accepted: 02/06/2024] [Indexed: 02/28/2024] Open
Abstract
Objective To evaluate the prognostic value of the systemic immune-inflammatory index (SII) for predicting in-hospital major adverse cardiovascular events (MACEs) in patients with acute myocardial infarction (AMI) and establish a relevant nomogram. Methods This study included 954 AMI patients. We examined three inflammatory factors (SII, platelet to lymphocyte ratio (PLR) and neutrophil to lymphocyte ratio (NLR)) to see which one predicts in-hospital MACEs better. The predictors were subsequently screened using bidirectional stepwise regression method, and a MACE nomogram was constructed via logistic regression analysis. The predictive value of the model was evaluated using the area under the curve (AUC), sensitivity and specificity. In addition, the clinical utility of the nomogram was evaluated using decision curve analysis. We also compared the nomogram with the Global Registry of Acute Coronary Events (GRACE) scoring system. Results 334 (35.0%) patients had MACEs. The SII (AUC =0.684) had a greater predictive value for in-hospital MACEs in AMI patients than the PLR (AUC =0.597, P<0.001) or NLR (AUC=0.654, P=0.01). The area under the curve (AUC) of the SII-based multivariable model for predicting MACEs, which was based on the SII, Killip classification, left ventricular ejection fraction, age, urea nitrogen (BUN) concentration and electrocardiogram-based diagnosis, was 0.862 (95% CI: 0.833-0.891). Decision curve and calibration curve analysis revealed that SII-based multivariable model demonstrated a good fit and calibration and provided positive net benefits than the model without SII. The predictive value of the SII-based multivariable model was greater than that of the GRACE scoring system (P<0.001). Conclusion SII is a promising, reliable biomarker for identifying AMI patients at high risk of in-hospital MACEs, and SII-based multivariable model may serve as a quick and easy tool to identify these patients.
Collapse
Affiliation(s)
- Xiaobo Li
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
- Department of Cardiology, Xiangdong Hospital, Hunan Normal University, Liling, Hunan, People’s Republic of China
| | - Chen Yu
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Xuewei Liu
- The Tenth Affiliated Hospital of Southern Medical University (Dongguan People’s Hospital), Southern Medical University, Dongguan, Guangdong, People’s Republic of China
| | - Yejia Chen
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Yutian Wang
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Hongbin Liang
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - ShiFeng Qiu
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Li Lei
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Jiancheng Xiu
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| |
Collapse
|
11
|
Marriott E, Singanayagam A, El-Awaisi J. Inflammation as the nexus: exploring the link between acute myocardial infarction and chronic obstructive pulmonary disease. Front Cardiovasc Med 2024; 11:1362564. [PMID: 38450367 PMCID: PMC10915015 DOI: 10.3389/fcvm.2024.1362564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 02/01/2024] [Indexed: 03/08/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD), particularly following acute exacerbations (AE-COPD), significantly heightens the risks and mortality associated with acute myocardial infarction (AMI). The intersection of COPD and AMI is characterised by a considerable overlap in inflammatory mechanisms, which play a crucial role in the development of both conditions. Although extensive research has been conducted on individual inflammatory pathways in AMI and COPD, the understanding of thrombo-inflammatory crosstalk in comorbid settings remains limited. The effectiveness of various inflammatory components in reducing AMI infarct size or slowing COPD progression has shown promise, yet their efficacy in the context of comorbidity with COPD and AMI is not established. This review focuses on the critical importance of both local and systemic inflammation, highlighting it as a key pathophysiological connection between AMI and COPD/AE-COPD.
Collapse
Affiliation(s)
- Eloise Marriott
- Microcirculation Research Group, Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Aran Singanayagam
- MRC Centre for Molecular Bacteriology & Infection, Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Juma El-Awaisi
- Microcirculation Research Group, Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
12
|
Huang S, Zhang H, Zhuang Z, Guo N, Zhou Q, Duan X, Ge L. Propensity score analysis of red cell distribution width to serum calcium ratio in acute myocardial infarction as a predictor of in-hospital mortality. Front Cardiovasc Med 2023; 10:1292153. [PMID: 38169646 PMCID: PMC10758436 DOI: 10.3389/fcvm.2023.1292153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024] Open
Abstract
Objective Red cell distribution width (RDW) and serum calcium (Ca) levels are predictors of in-hospital mortality in acute myocardial infarction (AMI) patients. However, their sensitivity and specificity are limited. Therefore, this study aimed to determine whether the RDW to Ca ratio (RCR) acquired on admission can be used to predict the in-hospital mortality of AMI patients. Methods This retrospective cohort study extracted clinical information from the Medical Information Market for Intensive IV (MIMIC-IV) database on 2,910 AMI patients enrolled via propensity score matching (PSM). Prognostic values were assessed using a multivariate logistic model and three PSM approaches. Analysis was performed based on stratified variables and interactions among sex, age, ethnicity, anemia, renal disease, percutaneous transluminal coronary intervention (PCI), coronary artery bypass grafting (CABG), atrial fibrillation, congestive heart failure, dementia, diabetes, paraplegia, hypertension, cerebrovascular disease, and Sequential Organ Failure Assessment (SOFA) score. Results A total of 4,105 ICU-admitted AMI patients were analyzed. The optimal cut-off value of the RCR for in-hospital mortality was 1.685. The PSM was performed to identify 1,455 pairs (2,910) of score-matched patients, with balanced differences exhibited for nearly all variables.The patients' median age was 72 years (range, 63-82 years) and 60.9% were male. The risk of in-hospital mortality incidence increased with increasing RCR levels. After adjusting for confounders, the risk ratio for the incidence of in-hospital mortality for high RCR was 1.75 [95% confidence interval (CI): 1.60-1.94, P = 0.0113] compared to that associated with low RCR in the PSM cohort. High RCR was also substantially implicated in in-hospital mortality incidence in the weighted cohorts [odds ratio (OR) = 1.76, 95% CI: 1.62-1.94, P = 0.0129]. Assessment of RCR in three groups showed that patients with high RCR also had a higher risk of in-hospital mortality (OR = 3.04; 95% CI, 2.22-4.16; P < 0.0001) than in patients with RCR in the adjusted model. In the sensitivity analysis, both the original and weighted groups showed similar results. Conclusion The RCR at admission may be useful for predicting in-hospital mortality in ICU-admitted AMI patients.
Collapse
Affiliation(s)
- Sulan Huang
- The First Affiliated Hospital, Jinan University, Guangzhou, China
- Department of Cardiovascular Medicine, The First People's Hospital of Changde City, Changde City, Hunan Province, China
| | - Huijia Zhang
- Department of Rheumatology and Immunology, The First People's Hospital of Changde City, Changde City, Hunan Province, China
| | - Zhijie Zhuang
- Department of Gastroenterology, The First People's Hospital of Changde City, Changde City, Hunan Province, China
| | - Ning Guo
- Department of Cardiovascular Medicine, The First People's Hospital of Changde City, Changde City, Hunan Province, China
| | - Quan Zhou
- Department of Science and Education Section, The First People's Hospital of Changde City, Changde City, Hunan Province, China
| | - Xiangjie Duan
- Department of Infectious Disease, The First People's Hospital of Changde, Changde City, Hunan Province, China
| | - Liangqing Ge
- The First Affiliated Hospital, Jinan University, Guangzhou, China
- Department of Cardiovascular Medicine, The First People's Hospital of Changde City, Changde City, Hunan Province, China
| |
Collapse
|
13
|
Kong MG, Suh J, Lee B, Park HW, Park SY, Moon I, Choi HO, Seo HS, Cho YH, Lee NH, Jang HJ, Kim TH, Kwon SW, Park SD, Oh PC, Moon J, Lee K, Kang WC. Hemo-metabolic impairment in patients with ST-segment elevation myocardial infarction: Data from the INTERSTELLAR registry. Cardiol J 2023; 31:434-441. [PMID: 37964646 PMCID: PMC11229799 DOI: 10.5603/cj.93926] [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: 01/30/2023] [Revised: 08/16/2023] [Accepted: 09/22/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND Not only hemo-dynamic (HD) factors but also hemo-metabolic (HM) risk factors reflecting multi-organ injuries are considered as important prognostic factors in ST-segment elevation myocardial infarction (STEMI). However, studies regarding HM risk factors in STEMI patients are currently limited. METHOD Under analysis were 1,524 patients with STEMI who underwent primary percutaneous coronary intervention in the INTERSTELLAR registry. Patients were divided into HM (≥ 2 risk factors) and non-HM impairment groups. The primary outcome was in-hospital all-cause mortality, and the secondary outcome was 1-year all-cause mortality. RESULTS Of 1,524 patients, 214 (14.0%) and 1,310 (86.0%) patients were in the HM and non-HM impairment groups, respectively. Patients with HM impairment had a higher incidence of in-hospital mortality than those without (24.3% vs. 2.7%, p < 0.001). After adjusting for confounders, HM impairment was independently associated with in-hospital mortality (inverse probability of treatment weighting [IPTW]-adjusted odds ratio: 1.81, 95% confidence interval: 1.08-3.14). In the third door-to-balloon (DTB) time tertile (≥ 82 min), HM impairment was strongly associated with in-hospital mortality. In the first DTB time tertile ( < 62 min), indicating relatively rapid revascularization, HM impairment was consistently associated with increased in-hospital mortality. CONCLUSIONS Hemo-metabolic impairment is significantly associated with increased risk of in-hospital and 1-year mortality in patients with STEMI. It remains a significant prognostic factor, regardless of DTB time.
Collapse
Affiliation(s)
- Min Gyu Kong
- Division of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Jon Suh
- Division of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea.
| | - Bora Lee
- Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea
| | - Hyun Woo Park
- Division of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Su Yeong Park
- Division of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Inki Moon
- Division of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Hyung Oh Choi
- Division of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Hye-Sun Seo
- Division of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Yoon Haeng Cho
- Division of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Nae-Hee Lee
- Division of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Republic of Korea
| | - Ho-Jun Jang
- Division of Cardiology, Sejong General Hospital, Bucheon, Republic of Korea
| | - Tae-Hoon Kim
- Division of Cardiology, Hanil General Hospital, Seoul, Republic of Korea
| | - Sung Woo Kwon
- Division of Cardiology, Inha University Hospital, Incheon, Republic of Korea
| | - Sang-Don Park
- Division of Cardiology, Inha University Hospital, Incheon, Republic of Korea
| | - Pyung Chun Oh
- Division of Cardiology, Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Jeonggeun Moon
- Division of Cardiology, Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Kyounghoon Lee
- Division of Cardiology, Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Woong Chol Kang
- Division of Cardiology, Gil Medical Center, Gachon University College of Medicine, Incheon, Republic of Korea
| |
Collapse
|
14
|
Wu L, Yin W, Wen J, Wang S, Li H, Wang X, Zhang W, Duan S, Zhu Q, Gao E, Wu S, Zhan B, Zhou R, Yang X. Excretory/secretory products from Trichinella spiralis adult worms ameliorate myocardial infarction by inducing M2 macrophage polarization in a mouse model. Parasit Vectors 2023; 16:362. [PMID: 37845695 PMCID: PMC10577921 DOI: 10.1186/s13071-023-05930-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/14/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND Ischemia-induced inflammatory response is the main pathological mechanism of myocardial infarction (MI)-caused heart tissue injury. It has been known that helminths and worm-derived proteins are capable of modulating host immune response to suppress excessive inflammation as a survival strategy. Excretory/secretory products from Trichinella spiralis adult worms (Ts-AES) have been shown to ameliorate inflammation-related diseases. In this study, Ts-AES were used to treat mice with MI to determine its therapeutic effect on reducing MI-induced heart inflammation and the immunological mechanism involved in the treatment. METHODS The MI model was established by the ligation of the left anterior descending coronary artery, followed by the treatment of Ts-AES by intraperitoneal injection. The therapeutic effect of Ts-AES on MI was evaluated by measuring the heart/body weight ratio, cardiac systolic and diastolic functions, histopathological change in affected heart tissue and observing the 28-day survival rate. The effect of Ts-AES on mouse macrophage polarization was determined by stimulating mouse bone marrow macrophages in vitro with Ts-AES, and the macrophage phenotype was determined by flow cytometry. The protective effect of Ts-AES-regulated macrophage polarization on hypoxic cardiomyocytes was determined by in vitro co-culturing Ts-AES-induced mouse bone marrow macrophages with hypoxic cardiomyocytes and cardiomyocyte apoptosis determined by flow cytometry. RESULTS We observed that treatment with Ts-AES significantly improved cardiac function and ventricular remodeling, reduced pathological damage and mortality in mice with MI, associated with decreased pro-inflammatory cytokine levels, increased regulatory cytokine expression and promoted macrophage polarization from M1 to M2 type in MI mice. Ts-AES-induced M2 macrophage polarization also reduced apoptosis of hypoxic cardiomyocytes in vitro. CONCLUSIONS Our results demonstrate that Ts-AES ameliorates MI in mice by promoting the polarization of macrophages toward the M2 type. Ts-AES is a potential pharmaceutical agent for the treatment of MI and other inflammation-related diseases.
Collapse
Affiliation(s)
- Lingqin Wu
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
- Second Affiliated Hospital of Jiaxing University, Jiaxing, 314000, China
| | - Wenhui Yin
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Jutai Wen
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Shuying Wang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Huihui Li
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- Basic Medical College of Bengbu Medical College, Bengbu, 233000, China
| | - Xiaoli Wang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- Basic Medical College of Bengbu Medical College, Bengbu, 233000, China
| | - Weixiao Zhang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Shuyao Duan
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Qiuyu Zhu
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China
| | - Erhe Gao
- Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Shili Wu
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Bin Zhan
- National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Rui Zhou
- First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China.
| | - Xiaodi Yang
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical College, Bengbu, 233000, China.
- Basic Medical College of Bengbu Medical College, Bengbu, 233000, China.
| |
Collapse
|
15
|
Sadeghi R, Haji Aghajani M, Parandin R, Taherpour N, Ahmadzadeh K, Sarveazad A. Leuko-Glycemic Index in the Prognosis of Acute Myocardial Infarction; a Cohort Study on Coronary Angiography and Angioplasty Registry. ARCHIVES OF ACADEMIC EMERGENCY MEDICINE 2023; 11:e63. [PMID: 37840868 PMCID: PMC10568944 DOI: 10.22037/aaem.v11i1.2085] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Introduction The leuko-glycemic index (LGI), a combined index of patient leukocyte counts and blood glucose levels, has been shown to predict the prognosis of myocardial infarction (MI) patients. Our study aims to investigate the performance of LGI in prediction of outcomes in a population of diabetic and non-diabetic MI patients. Methods This observational registry-based cohort study was performed on acute myocardial infarction (AMI) patients. Participants were sub-grouped according to their diabetes status and the calculated optimal LGI cut-off value. The outcomes of the study were the length of hospital stay, and in-hospital and 30-day mortality. Results A total of 296 AMI (112 diabetic and 184 non-diabetic) patients were included in the study. The optimal cut-off value of LGI in the diabetic and non-diabetic groups was calculated as 2970.4 mg/dl.mm3 and 2249.4 mg/dl.mm3, respectively. High LGI was associated with increased hospital admission duration in non-diabetic patients (p = 0.017). The area under the curve (AUC) of LGI for prediction of in-hospital mortality was 0.93 (95% CI: 0.87 to 1.00) in the diabetic group and 0.92 (95% CI: 0.85 to 0.99) in the non-diabetic group. LGI had a sensitivity and specificity of 90.00%, and 93.14% in prediction of in-hospital mortality in the diabetic group compared to 77.77% and 90.85% in the non-diabetic group. We observed 4 post-discharge mortalities in our patient group. Conclusion Our study demonstrated that higher LGI predicts in-hospital mortality in both diabetic and non-diabetic patients, while the length of hospital stay was only predicted by LGI levels in non-diabetic patients.
Collapse
Affiliation(s)
- Roxana Sadeghi
- Prevention of Cardiovascular Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Haji Aghajani
- Prevention of Cardiovascular Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Parandin
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Niloufar Taherpour
- Prevention of Cardiovascular Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Koohyar Ahmadzadeh
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Arash Sarveazad
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
- Nursing Care Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Moustafa A, Hashemi S, Brar G, Grigull J, Ng SHS, Williams D, Schmitt-Ulms G, McDermott JC. The MEF2A transcription factor interactome in cardiomyocytes. Cell Death Dis 2023; 14:240. [PMID: 37019881 PMCID: PMC10076289 DOI: 10.1038/s41419-023-05665-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 12/02/2022] [Accepted: 02/08/2023] [Indexed: 04/07/2023]
Abstract
Transcriptional regulators encoded by the Myocyte Enhancer Factor 2 (MEF2) gene family play a fundamental role in cardiac development, homeostasis and pathology. Previous studies indicate that MEF2A protein-protein interactions serve as a network hub in several cardiomyocyte cellular processes. Based on the idea that interactions with regulatory protein partners underly the diverse roles of MEF2A in cardiomyocyte gene expression, we undertook a systematic unbiased screen of the MEF2A protein interactome in primary cardiomyocytes using an affinity purification-based quantitative mass spectrometry approach. Bioinformatic processing of the MEF2A interactome revealed protein networks involved in the regulation of programmed cell death, inflammatory responses, actin dynamics and stress signaling in primary cardiomyocytes. Further biochemical and functional confirmation of specific protein-protein interactions documented a dynamic interaction between MEF2A and STAT3 proteins. Integration of transcriptome level data from MEF2A and STAT3-depleted cardiomyocytes reveals that the balance between MEF2A and STAT3 activity exerts a level of executive control over the inflammatory response and cardiomyocyte cell survival and experimentally ameliorates Phenylephrine induced cardiomyocyte hypertrophy. Lastly, we identified several MEF2A/STAT3 co-regulated genes, including the MMP9 gene. Herein, we document the cardiomyocyte MEF2A interactome, which furthers our understanding of protein networks involved in the hierarchical control of normal and pathophysiological cardiomyocyte gene expression in the mammalian heart.
Collapse
Affiliation(s)
- Amira Moustafa
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada
| | - Sara Hashemi
- Analytical Sciences, Sanofi, Toronto, ON, M2R 3T4, Canada
- Seneca College, School of Health Sciences, King City, ON, L7B 1B3, Canada
| | - Gurnoor Brar
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada
| | - Jörg Grigull
- Department of Mathematics and Statistics, York University, Toronto, ON, M3J1P3, Canada
| | - Siemon H S Ng
- Analytical Sciences, Sanofi, Toronto, ON, M2R 3T4, Canada
- Analytical Development, Notch Therapeutics, Toronto, ON, M5G 1M1, Canada
| | - Declan Williams
- Tanz Centre for Research in Neurodegenerative Diseases, and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5T 0S8, Canada
| | - Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5T 0S8, Canada
| | - John C McDermott
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada.
- Muscle Health Research Centre (MHRC), York University, Toronto, ON, M3J 1P3, Canada.
- Centre for Research in Biomolecular Interactions (CRBI), York University, Toronto, ON, M3J 1P3, Canada.
| |
Collapse
|
17
|
Ortiz VD, Teixeira RB, Türck P, Corssac GB, Belló-Klein A, de Castro AL, Araujo ASDR. Influence of carvedilol and thyroid hormones on inflammatory proteins and cardioprotective factor HIF-1α in the infarcted heart. Can J Physiol Pharmacol 2023; 101:106-116. [PMID: 36661235 DOI: 10.1139/cjpp-2022-0355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Inflammatory pathways of Toll-like receptor 4 (TLR4) and NLRP3 inflammasome contribute to acute myocardial infarction (AMI) pathophysiology. The hypoxia-inducible factor 1α (HIF-1α), however, is a key transcription factor related to cardioprotection. This study aimed to compare the influence of carvedilol and thyroid hormones (TH) on inflammatory and HIF-1α proteins and on cardiac haemodynamics in the infarcted heart. Male Wistar rats were allocated into five groups: sham-operated group (SHAM), infarcted group (MI), infarcted treated with the carvedilol group (MI + C), infarcted treated with the TH group (MI + TH), and infarcted co-treated with the carvedilol and TH group (MI + C + TH). Haemodynamic analysis was assessed 15 days post-AMI. The left ventricle (LV) was collected for morphometric and Western blot analysis. The MI group presented LV systolic pressure reduction, LV end-diastolic pressure elevation, and contractility index decrease compared to the SHAM group. The MI + C, MI + TH, and MI + C + TH groups did not reveal such alterations compared to the SHAM group. The MI + TH and MI + C + TH groups presented reduced MyD88 and NLRP3 and increased HIF-1α levels. In conclusion, all treatments preserve the cardiac haemodynamic, and only TH, as isolated treatment or in co-treatment with carvedilol, was able to reduce MyD88 and NLRP3 and increase HIF-1α in the infarcted heart.
Collapse
Affiliation(s)
- Vanessa Duarte Ortiz
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Department of Physiology, Institute of Basic Health Science, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Rayane Brinck Teixeira
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Department of Physiology, Institute of Basic Health Science, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Patrick Türck
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Department of Physiology, Institute of Basic Health Science, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Giana Blume Corssac
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Department of Physiology, Institute of Basic Health Science, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Adriane Belló-Klein
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Department of Physiology, Institute of Basic Health Science, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Alexandre Luz de Castro
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Department of Physiology, Institute of Basic Health Science, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Alex Sander da Rosa Araujo
- Laboratory of Cardiovascular Physiology and Reactive Oxygen Species, Department of Physiology, Institute of Basic Health Science, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
18
|
Circulating Galectin-3 in Patients with Cardiogenic Shock Complicating Acute Myocardial Infarction Treated with Mild Hypothermia: A Biomarker Sub-Study of the SHOCK-COOL Trial. J Clin Med 2022; 11:jcm11237168. [PMID: 36498742 PMCID: PMC9740246 DOI: 10.3390/jcm11237168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 11/23/2022] [Accepted: 11/30/2022] [Indexed: 12/05/2022] Open
Abstract
Background: Galectin-3 (Gal-3) is considered a potential cardiovascular inflammatory marker that may provide additional risk stratification for patients with acute heart failure. It is unknown whether mild therapeutic hypothermia (MTH) impacts Gal-3 levels. Therefore, this biomarker study aimed to investigate the effect of MTH on Gal-3. Methods: In the randomized SHOCK-COOL trial, 40 patients with cardiogenic shock (CS) complicating acute myocardial infraction (AMI) were randomly assigned to the MTH (33 °C) or control group in a 1:1 ratio. Blood samples were collected on the day of admission/day 1, day 2, and day 3. Gal-3 level kinetics throughout these time points were compared between the MTH and control groups. Additionally, potential correlations between Gal-3 and clinical patient characteristics were assessed. Multiple imputations were performed to account for missing data. Results: In the control group, Gal-3 levels were significantly lower on day 3 than on day 1 (day 1 vs. day 3: 3.84 [IQR 2.04−13.3] vs. 1.79 [IQR 1.23−3.50] ng/mL; p = 0.049). Gal-3 levels were not significantly different on any day between the MTH and control groups (p for interaction = 0.242). Spearman’s rank correlation test showed no significant correlation between Gal-3 levels and sex, age, smoking, body mass index (BMI), and levels of creatine kinase-MB, creatine kinase, C-reactive protein, creatinine, and white blood cell counts (all p > 0.05). Patients with lower Gal-3 levels on the first day after admission demonstrated a higher risk of all-cause mortality at 30 days (hazard ratio, 2.67; 95% CI, 1.11−6.42; p = 0.029). In addition, Gal-3 levels on day 1 had a good predictive value for 30-day all-cause mortality with an area under the receiver operating characteristic curve of 0.696 (95% CI: 0.513−0.879), with an optimal cut-off point of less than 3651 pg/mL. Conclusions: MTH has no effect on Gal-3 levels in patients with CS complicating AMI compared to the control group. In addition, Gal-3 is a relatively stable biomarker, independent of age, sex, and BMI, and Gal-3 levels at admission might predict the risk of 30-day all-cause mortality.
Collapse
|
19
|
An Z, Tian J, Liu Y, Zhao X, Yang X, Yong J, Liu L, Zhang L, Jiang W, Song X, Zhang H. Exosomes as a Cell-free Therapy for Myocardial Injury Following Acute Myocardial Infarction or Ischemic Reperfusion. Aging Dis 2022; 13:1770-1786. [PMID: 36465167 PMCID: PMC9662265 DOI: 10.14336/ad.2022.0416] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/16/2022] [Indexed: 08/13/2023] Open
Abstract
Exosomes, which contain miRNA, have been receiving growing attention in cardiovascular therapy because of their role in mediating cell-cell communication, autophagy, apoptosis, inflammation, and angiogenesis. Several studies have suggested that miRNA derived from exosomes can be used to detect myocardial infarctions (MI) in patients. Basic research also suggests that exosomes could serve as a potential therapeutic target for treating acute myocardial infarction. Ischemia/reperfusion (IR) injury is associated with adverse cardiac events after acute MI. We aim to review the potential benefits and mechanisms of exosomes in treating MI and IR injury.
Collapse
Affiliation(s)
- Ziyu An
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Jinfan Tian
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Yue Liu
- Cardiovascular disease center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Xin Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Xueyao Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Jingwen Yong
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Libo Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Lijun Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Wenjian Jiang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Xiantao Song
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Hongjia Zhang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
20
|
BMI Modifies Increased Mortality Risk of Post-PCI STEMI Patients with AKI. J Clin Med 2022; 11:jcm11206104. [PMID: 36294425 PMCID: PMC9604849 DOI: 10.3390/jcm11206104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/29/2022] [Accepted: 10/13/2022] [Indexed: 11/25/2022] Open
Abstract
Mortality from acute ST elevation myocardial infarction (STEMI) was significantly reduced with the introduction of percutaneous catheterization intervention (PCI) but remains high in patients who develop acute kidney injury (AKI). Previous studies found overweight to be protective from mortality in patients suffering from STEMI and AKI separately but not as they occur concurrently. This study aimed to establish the relationship between AKI and mortality in STEMI patients after PCI and whether body mass index (BMI) has a protective impact. Between January 2008 and June 2016, two thousand one hundred and forty-one patients with STEMI underwent PCI and were admitted to the Tel Aviv Medical Center Cardiac Intensive Care Unit. Their demographic, laboratory, and clinical data were collected and analyzed. We compared all-cause mortality in patients who developed AKI after PCI for STEMI and those who did not. In total, 178 patients (10%) developed AKI and had higher mortality (p < 0.001). Logistic regression analysis was performed to determine the relationship between AKI, BMI, and mortality. AKI was significantly associated with both 30-day and overall mortality, while BMI had a significant protective effect. Survival analysis found a significant difference in 30-day and overall survival between patients with and without AKI with a significant protective effect of BMI on survival at 30 days. AKI presents a major risk for mortality and poor survival after PCI for STEMI, yet a beneficial effect of increased BMI modifies it.
Collapse
|
21
|
Li Y, Sun X, Liu X, Li J, Li X, Wang G, Liu Y, Lu X, Cui L, Shao M, Wang Y, Wang W, Li C. P2X7R-NEK7-NLRP3 Inflammasome Activation: A Novel Therapeutic Pathway of Qishen Granule in the Treatment of Acute Myocardial Ischemia. J Inflamm Res 2022; 15:5309-5326. [PMID: 36124207 PMCID: PMC9482414 DOI: 10.2147/jir.s373962] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 08/26/2022] [Indexed: 11/23/2022] Open
Abstract
Background Acute myocardial ischemia (AMI) is a common heart disease with increasing morbidity and mortality year by year. Persistent and sterile inflammatory infiltration of myocardial tissue is an important factor triggering of acute myocardial ischemia secondary to acute myocardial infarction, and NLRP3 inflammasome activation is an important part of sterile inflammatory response after acute myocardial ischemia. Previous studies have shown that Qishen granule (QSG) can significantly inhibit the inflammatory injury of myocardial tissue caused by ischemia, but its effect and specific mechanism of inhibiting the activation of NLRP3 inflammasome have not been reported. This study was to investigate the specific mechanism of QSG inhibiting inflammation after AMI, and to validate the possible targets. Methods The myocardial ischemia model in mice was established by ligation of the left anterior descending coronary artery. Echocardiography was used to evaluate the cardiac function of the mice. Plasma CK-MB and cTnl were detected by ELISA to evaluate the degree of myocardial injury. The extent of myocardial tissue inflammation in mice was assessed by HE staining and immunohistochemistry of IL-18, IL-1β. The expressions of NLRP3, ASC, Caspase-1, and CD86 were detected by immunofluorescence; detection of key pathway proteins P2X7R, NEK7, NLRP3, ASC, Caspase-1, and effector proteins IL-18, IL-1β by Western blot. In vitro experiments, ATP+LPS was used to construct a RAW264.7 macrophage NLRP3 inflammasome activation model. Immunofluorescence and Western blot analysis were performed to detect the expression of NLRP3 pathway activator and effector proteins. Plasmid-transfected P2X7R overexpression and immunoprecipitation assays were used to evaluate the QSG-regulated NLRP3 inflammasome activation pathway. Results QSG rescued cardiac function and further reduced inflammatory effects in mice by inhibiting NLRP3 inflammasome activation. In vitro, QSG inhibited LPS combined with ATP-induced NLRP3 inflammasome activation in RAW264.7 macrophages by downregulating the expression of NLRP3 inflammasome key pathway proteins. In addition, inhibition or overexpression of P2X7R in RAW264.7 macrophages and immunoprecipitated protein interactions further confirmed that QSG reduces macrophages inflammasome activation via the P2X7R-NEK7-NLRP3 pathway. Conclusion P2X7R-NEK7-NLRP3 inflammasome activation is a novel therapeutic mechanism of QSG in the treatment of acute myocardial ischemia.
Collapse
Affiliation(s)
- Yanqin Li
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Xiaoqian Sun
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Xiangning Liu
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Junjun Li
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Xuan Li
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Gang Wang
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Yizhou Liu
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Xiangyu Lu
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Lingwen Cui
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Mingyan Shao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Yong Wang
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China.,School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China.,Beijing Key Laboratory of TCM Syndrome and Formula, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| | - Wei Wang
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China.,Beijing Key Laboratory of TCM Syndrome and Formula, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China.,Guangzhou University of Chinese Medicine, Guangdong, 510006, People's Republic of China
| | - Chun Li
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China.,Beijing Key Laboratory of TCM Syndrome and Formula, Beijing University of Chinese Medicine, Beijing, 100029, People's Republic of China
| |
Collapse
|
22
|
Haghshenas MR, Zamir MR, Sadeghi M, Fattahi MJ, Mirshekari K, Ghaderi A. Clinical relevance and therapeutic potential of IL-38 in immune and non-immune-related disorders. Eur Cytokine Netw 2022; 33:54-69. [PMID: 37052152 PMCID: PMC10134710 DOI: 10.1684/ecn.2022.0480] [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] [Accepted: 07/30/2022] [Indexed: 04/14/2023]
Abstract
Interleukin-38 (IL-38) is the most recent member of the IL-1 family that acts as a natural inflammatory inhibitor by binding to cognate receptors, particularly the IL-36 receptor. In vitro, animal and human studies on autoimmune, metabolic, cardiovascular and allergic diseases, as well sepsis and respiratory viral infections, have shown that IL-38 exerts an anti-inflammatory activity by modulating the generation and function of inflammatory cytokines (e.g. IL-6, IL-8, IL-17 and IL-36) and regulating dendritic cells, M2 macrophages and regulatory T cells (Tregs). Accordingly, IL-38 may possess therapeutic potential for these types of diseases. IL-38 down-regulates CCR3+ eosinophil cells, CRTH2+ Th2 cells, Th17 cells, and innate lymphoid type 2 cells (ILC2), but up-regulates Tregs, and this has influenced the design of immunotherapeutic strategies based on regulatory cells/cytokines for allergic asthma in future studies. In auto-inflammatory diseases, IL-38 alleviates skin inflammation by regulating γδ T cells and limiting the production of IL-17. Due to its ability to suppress IL-1β, IL-6 and IL-36, this cytokine could reduce COVID-19 severity, and might be employed as a therapeutic tool. IL-38 may also influence host immunity and/or the components of the cancer microenvironment, and has been shown to improve the outcome of colorectal cancer, and may participate in tumour progression in lung cancer possibly by modulating CD8 tumour infiltrating T cells and PD-L1 expression. In this review, we first briefly present the biological and immunological functions of IL-38, and then discuss the important roles of IL-38 in various types of diseases, and finally highlight its use in therapeutic strategies.
Collapse
Affiliation(s)
- Mohammad Reza Haghshenas
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mina Roshan Zamir
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahboubeh Sadeghi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Javad Fattahi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Kimia Mirshekari
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Ghaderi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
23
|
John RV, Devasia T, N M, Lukose J, Chidangil S. Micro-Raman spectroscopy study of blood samples from myocardial infarction patients. Lasers Med Sci 2022; 37:3451-3460. [PMID: 35821543 DOI: 10.1007/s10103-022-03604-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 06/28/2022] [Indexed: 12/15/2022]
Abstract
Acute myocardial infarction (MI) is found to be a major causative factor for global mortality and morbidity. This situation demands necessity of developing efficient and rapid diagnostic tools to detect acute MI. Raman spectroscopy is a non-destructive optical diagnostic technique, which has high potential in probing biochemical changes in clinical samples during initiation and progress of diseases. In this work, blood was taken as the sample to examine inflammation in acute MI patients using Raman spectroscopy. Ratio of Raman peak intensities that corresponds to phenylalanine (1000 cm-1) and tyrosine (825 cm-1) can facilitate indirect information about tetrahydrobiopterin (BH4) availability, which can indicate inflammatory status in patients. This ratio obtained was higher for MI patients in comparison with control subjects. The decrease in phenylalanine and tyrosine ratio (Phe-Tyr ratio) is attributed to the prognosis of standard of care (medications like antiplatelets including aspirin, statin and revascularisation) leading to inflammation reduction. Phe-Tyr ratio estimated from the Raman spectra of blood can be exploited as a reliable method to probe inflammation due to MI. The method is highly objective, require only microliters of sample and minimal sample preparation, signifying its clinical utility.
Collapse
Affiliation(s)
- Reena V John
- Centre of Excellence for Biophotonics, Department of Atomic and Molecular Physics, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Tom Devasia
- Department of Cardiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Mithun N
- Centre of Excellence for Biophotonics, Department of Atomic and Molecular Physics, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Jijo Lukose
- Centre of Excellence for Biophotonics, Department of Atomic and Molecular Physics, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Santhosh Chidangil
- Centre of Excellence for Biophotonics, Department of Atomic and Molecular Physics, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
24
|
Kalinskaya A, Dukhin O, Lebedeva A, Maryukhnich E, Rusakovich G, Vorobyeva D, Shpektor A, Margolis L, Vasilieva E. Circulating Cytokines in Myocardial Infarction Are Associated With Coronary Blood Flow. Front Immunol 2022; 13:837642. [PMID: 35242141 PMCID: PMC8886043 DOI: 10.3389/fimmu.2022.837642] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/21/2022] [Indexed: 01/08/2023] Open
Abstract
Background The level of systemic inflammation correlates with the severity of the clinical course of acute myocardial infarction (AMI). It has been shown that circulating cytokines and endothelial dysfunction play an important role in the process of clot formation. The aim of our study was to assess the concentration of various circulating cytokines, endothelial function and blood clotting in AMI patients depending on the blood flow through the infarction-related artery (IRA). Methods We included 75 patients with AMI. 58 presented with ST-elevation myocardial infarction (STEMI) and 17 had non-ST-elevation myocardial infarction (non-STEMI). A flow-mediated dilation test (FMD test), thrombodynamics and rotational thromboelastometry as well as assessment of 14 serum cytokines using xMAP technology were performed. Findings Non-STEMI-patients were characterized by higher levels of MDC, MIP-1β, TNF-α. Moreover, we observed that patients with impaired blood flow through the IRA (TIMI flow 0-1) had higher average and initial clot growth rates, earlier onset of spontaneous clots, C-reactive protein (CRP) and IL-10 compared to patients with preserved blood flow through the IRA (TIMI flow 2-3). Patients with TIMI 2-3 blood flow had higher level of IP-10. IL-10 correlated with CRP and pro-inflammatory cytokines levels, initial clot growth rate and clot lysis time in TIMI 0-1 patients. All these differences were statistically significant. Interpretation We demonstrated that concentrations of the inflammatory cytokines correlate not only with the form of myocardial infarction (STEMI or non-STEMI), but also with the blood flow through the infarct-related artery. Inflammatory response, functional state of endothelium, and clot formation are closely linked with each other. A combination of these parameters affects the patency of the infarct-related artery.
Collapse
Affiliation(s)
- Anna Kalinskaya
- Laboratory of Atherothrombosis, Cardiology Department, Moscow State University of Medicine and Dentistry, Moscow, Russia.,Clinical City Hospital named after I.V. Davydovsky, Moscow Department of Healthcare, Moscow, Russia
| | - Oleg Dukhin
- Laboratory of Atherothrombosis, Cardiology Department, Moscow State University of Medicine and Dentistry, Moscow, Russia.,Clinical City Hospital named after I.V. Davydovsky, Moscow Department of Healthcare, Moscow, Russia
| | - Anna Lebedeva
- Laboratory of Atherothrombosis, Cardiology Department, Moscow State University of Medicine and Dentistry, Moscow, Russia
| | - Elena Maryukhnich
- Laboratory of Atherothrombosis, Cardiology Department, Moscow State University of Medicine and Dentistry, Moscow, Russia
| | - Georgy Rusakovich
- Laboratory of Atherothrombosis, Cardiology Department, Moscow State University of Medicine and Dentistry, Moscow, Russia
| | - Daria Vorobyeva
- Laboratory of Atherothrombosis, Cardiology Department, Moscow State University of Medicine and Dentistry, Moscow, Russia
| | - Alexander Shpektor
- Laboratory of Atherothrombosis, Cardiology Department, Moscow State University of Medicine and Dentistry, Moscow, Russia.,Clinical City Hospital named after I.V. Davydovsky, Moscow Department of Healthcare, Moscow, Russia
| | - Leonid Margolis
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Elena Vasilieva
- Laboratory of Atherothrombosis, Cardiology Department, Moscow State University of Medicine and Dentistry, Moscow, Russia.,Clinical City Hospital named after I.V. Davydovsky, Moscow Department of Healthcare, Moscow, Russia
| |
Collapse
|
25
|
Toll-Like Receptors/TNF-α Pathway Crosstalk and Impact on Different Sites of Recurrent Myocardial Infarction in Elderly Patients. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1280350. [PMID: 35425840 PMCID: PMC9005286 DOI: 10.1155/2022/1280350] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/17/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022]
Abstract
Background Recurrent myocardial infarction is associated with increased mortality. Risk and predictive factors of recurrent myocardial infarction in elderly patients after coronary stenting are not well known. This research sought to investigate the effects of proinflammatory cytokines and toll-like receptor on recurrent myocardial infarction after coronary stenting in elderly patients. Methods We measured the levels of toll-like receptor 2 (TLR2), toll-like receptor 3 (TLR3), toll-like receptor 4 (TLR4), tumor necrosis factor-α (TNF-α), soluble tumor necrosis factor-α receptor-1 (sTNFR-1), soluble tumor necrosis factor-α receptor-2 (sTNFR-2), endothelial progenitor cells (EPCs), and vascular endothelial growth factor (VEGF) in elderly patients with recurrent myocardial infarction and assessed the changes of proinflammatory cytokines and toll-like receptors in elderly patients with recurrent myocardial infarction after coronary stenting. Results Levels of TLR2, TLR3, TLR4, TNF-α, sTNFR-1, and sTNFR-2 were remarkably increased (P < 0.001), and EPCs and VEGF were remarkably lowered (P < 0.001) in the elderly patients with recurrent myocardial infarction after coronary stent implantation. Increased expressions of proinflammatory cytokines and toll-like receptors induced recurrent myocardial infarction after coronary stenting. Elevated expressions of proinflammatory cytokines and toll-like receptors may be used to identify elderly patients who have an increased risk of developing recurrent myocardial infarction after coronary stenting. Conclusion The increase levels of proinflammatory cytokines and toll-like receptors were associated with recurrent myocardial infarction after coronary stenting. Increased expressions of proinflammatory cytokines and toll-like receptors may be clinically useful biomarkers for predicting recurrent myocardial infarction in the elderly patients after coronary stent implantation.
Collapse
|
26
|
Foglio E, Pellegrini L, Russo MA, Limana F. HMGB1-Mediated Activation of the Inflammatory-Reparative Response Following Myocardial Infarction. Cells 2022; 11:cells11020216. [PMID: 35053332 PMCID: PMC8773872 DOI: 10.3390/cells11020216] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/04/2022] [Accepted: 01/08/2022] [Indexed: 02/06/2023] Open
Abstract
Different cell types belonging to the innate and adaptive immune system play mutually non-exclusive roles during the different phases of the inflammatory-reparative response that occurs following myocardial infarction. A timely and finely regulation of their action is fundamental for the process to properly proceed. The high-mobility group box 1 (HMGB1), a highly conserved nuclear protein that in the extracellular space can act as a damage-associated molecular pattern (DAMP) involved in a large variety of different processes, such as inflammation, migration, invasion, proliferation, differentiation, and tissue regeneration, has recently emerged as a possible regulator of the activity of different immune cell types in the distinct phases of the inflammatory reparative process. Moreover, by activating endogenous stem cells, inducing endothelial cells, and by modulating cardiac fibroblast activity, HMGB1 could represent a master regulator of the inflammatory and reparative responses following MI. In this review, we will provide an overview of cellular effectors involved in these processes and how HMGB1 intervenes in regulating each of them. Moreover, we will summarize HMGB1 roles in regulating other cell types that are involved in the different phases of the inflammatory-reparative response, discussing how its redox status could affect its activity.
Collapse
Affiliation(s)
- Eleonora Foglio
- Technoscience, Parco Scientifico e Tecnologico Pontino, 04100 Latina, Italy;
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy;
| | - Laura Pellegrini
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy;
| | - Matteo Antonio Russo
- IRCCS San Raffaele Roma and MEBIC Consortium, 00166 Rome, Italy;
- San Raffaele University of Rome, 00166 Rome, Italy
| | - Federica Limana
- San Raffaele University of Rome, 00166 Rome, Italy
- Laboratory of Cellular and Molecular Pathology, IRCCS San Raffaele Roma, 00166 Rome, Italy
- Correspondence:
| |
Collapse
|
27
|
Chen M, Liao L, Yan J, Lin FQ. Predictive Value of Red Blood Cell Distribution Width for 1-Year All-Cause Mortality in Critically Ill Patients with Acute Myocardial Infarction. Int J Gen Med 2022; 15:465-471. [PMID: 35046707 PMCID: PMC8760980 DOI: 10.2147/ijgm.s345109] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/16/2021] [Indexed: 12/19/2022] Open
Abstract
Objective Red blood cell distribution width (RDW) on admission is a prognostic factor in cardiovascular disease. This study investigated the prognostic value of the RDW measured within 24 hours before discharge (24h dRDW) on 1-year all-cause mortality in critically ill patients with acute myocardial infarction (AMI), and compared the effect of 24h dRDW in anemia and non-anemia patients. Materials and Methods Altogether, 4088 patients with AMI were studied retrospectively. Data from the MIMIC-III database were collected and analyzed. The Kaplan–Meier method, Cox regression models, and receiver operating characteristic (ROC) analysis were used to assess the impact of 24h dRDW on all-cause mortality in AMI patients, and a stratified analysis was performed to investigate the prognostic value of 24h dRDW in anemia and non-anemia patients. Results Of the 4088 patients, there were 704 non-survivors (17.2%). The non-survivors had a higher RDW than the survivors (p<0.001). Cox regression analysis showed that 24h dRDW had a significant independent association with 1‐year all-cause mortality in critically ill patients with AMI (quintile 5 vs quintile 1, HR, 95% CI: 2.847, 2.014–4.023). The area under the ROC curve of 24h dRDW was 0.710 (95% CI, 0.689–0.730). In the stratified analysis, a significant prognostic value of 24h dRDW was found in anemia patients for 1-year all-cause mortality, but not in non-anemia patients. Conclusion Elevated 24h dRDW values are significantly associated with increased hazards of all‐cause mortality in critically ill patients with AMI. Significant prognostic value of 24h dRDW was found in AMI patients with anemia, but not in those without anemia.
Collapse
Affiliation(s)
- Min Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Lin Liao
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Jie Yan
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Fa-Quan Lin
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
- Correspondence: Fa-Quan Lin Email
| |
Collapse
|
28
|
Korn A, Baylan U, Simsek S, Schalkwijk CG, Niessen HWM, Krijnen PAJ. Myocardial infarction coincides with increased NOX2 and N ε-(carboxymethyl) lysine expression in the cerebral microvasculature. Open Heart 2021; 8:openhrt-2021-001842. [PMID: 34819349 PMCID: PMC8614153 DOI: 10.1136/openhrt-2021-001842] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/05/2021] [Indexed: 12/24/2022] Open
Abstract
Background Myocardial infarction (MI) is associated with mental health disorders, in which neuroinflammation and cerebral microvascular dysfunction may play a role. Previously, we have shown that the proinflammatory factors Nε-(carboxymethyl)lysine (CML) and NADPH oxidase 2 (NOX2) are increased in the human infarcted heart microvasculature. The aim of this study was to analyse the presence of CML and NOX2 in the cerebral microvasculature of patients with MI. Methods Brain tissue was obtained at autopsy from 24 patients with MI and nine control patients. According to their infarct age, patients with MI were divided into three groups: 3–6 hours old (phase I), 6 hours–5 days old (phase II) and 5–14 days old (phase III). CML and NOX2 in the microvasculature were studied through immunohistochemical analysis. Results We observed a 2.5-fold increase in cerebral microvascular CML in patients with phase II and phase III MI (phase II: 21.39±7.91, p=0.004; phase III: 24.21±10.37, p=0.0007) compared with non-MI controls (8.55±2.98). NOX2 was increased in microvessels in patients with phase II MI (p=0.002) and phase III MI (p=0.04) compared with controls. No correlation was found between CML and NOX2 (r=0.58, p=0.13). Conclusions MI coincides with an increased presence of CML and NOX2 in the brain microvasculature. These data point to proinflammatory alterations in the brain microvasculature that may underlie MI-associated mental health disorders.
Collapse
Affiliation(s)
- Amber Korn
- Department of Pathology, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands .,Amsterdam Cardiovascular Sciences, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Umit Baylan
- Department of Pathology, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Suat Simsek
- Department of Internal Medicine, Noordwest Ziekenhuisgroep, Alkmaar, The Netherlands.,Department of Internal Medicine, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands
| | - Casper G Schalkwijk
- Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands.,Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Hans W M Niessen
- Department of Pathology, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| | - Paul A J Krijnen
- Department of Pathology, Amsterdam UMC Locatie VUmc, Amsterdam, The Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam UMC Location VUmc, Amsterdam, The Netherlands
| |
Collapse
|
29
|
M1 Bone Marrow-Derived Macrophage-Derived Extracellular Vesicles Inhibit Angiogenesis and Myocardial Regeneration Following Myocardial Infarction via the MALAT1/MicroRNA-25-3p/CDC42 Axis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9959746. [PMID: 34745428 PMCID: PMC8570847 DOI: 10.1155/2021/9959746] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 09/08/2021] [Accepted: 10/04/2021] [Indexed: 11/18/2022]
Abstract
Myocardial infarction (MI) is a severe cardiovascular disease. Some M1 macrophage-derived extracellular vesicles (EVs) are involved in the inhibition of angiogenesis and acceleration dysfunction during MI. However, the potential mechanism of M1 phenotype bone marrow-derived macrophages- (BMMs-) EVs (M1-BMMs-EVs) in MI is largely unknown. This study sought to investigate whether M1-BMMs-EVs increased CDC42 expression and activated the MEK/ERK pathway by carrying lncRNA MALAT1 and competitively binding to miR-25-3p, thus inhibiting angiogenesis and myocardial regeneration after MI. After EV treatment, the cardiac function, infarct size, fibrosis, angiogenesis, and myocardial regeneration of MI mice and the viability, proliferation and angiogenesis of oxygen-glucose deprivation- (OGD-) treated myocardial microvascular endothelial cells (MMECs) were assessed. MALAT1 expression in MI mice, cells, and EVs was detected. MALAT1 downstream microRNAs (miRs), genes, and pathways were predicted and verified. MALAT1 and miR-25-3p were intervened to evaluate EV effects on OGD-treated cells. In MI mice, EV treatment aggravated MI and inhibited angiogenesis and myocardial regeneration. In OGD-treated cells, EV treatment suppressed cell viability, proliferation, and angiogenesis. MALAT1 was highly expressed in MI mice, OGD-treated MMECs, M1-BMMs, and EVs. Silencing MALAT1 weakened the inhibition of EV treatment on OGD-treated cells. MALAT1 sponged miR-25-3p to upregulate CDC42. miR-25-3p overexpression promoted OGD-treated cell viability, proliferation, and angiogenesis. The MEK/ERK pathway was activated after EV treatment. Collectively, M1-BMMs-EVs inhibited angiogenesis and myocardial regeneration following MI via the MALAT1/miR-25-3p/CDC42 axis and the MEK/ERK pathway activation.
Collapse
|
30
|
Jong J, Packard RRS. 18F-FDG PET imaging of myocardial inflammation and viability following experimental infarction and anti-inflammatory treatment with compound MCC950. J Nucl Cardiol 2021; 28:2358-2360. [PMID: 32333277 DOI: 10.1007/s12350-020-02104-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 03/09/2020] [Indexed: 10/24/2022]
Affiliation(s)
- Jeremy Jong
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, 10833 Le Conte Ave., CHS Building Room 17-054A, Los Angeles, CA, 90095, USA
| | - René R Sevag Packard
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, 10833 Le Conte Ave., CHS Building Room 17-054A, Los Angeles, CA, 90095, USA.
- Ronald Reagan UCLA Medical Center, Los Angeles, CA, USA.
- Veterans Affairs West Los Angeles Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
31
|
Ji Z, Wang C, Tong Q. Role of miRNA-324-5p-Modified Adipose-Derived Stem Cells in Post-Myocardial Infarction Repair. Int J Stem Cells 2021; 14:298-309. [PMID: 34158416 PMCID: PMC8429947 DOI: 10.15283/ijsc21025] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/26/2021] [Accepted: 04/30/2021] [Indexed: 11/09/2022] Open
Abstract
Background and Objectives To seek out the role of mircoRNA (miR)-324-5p-modified adipose-derived stem cells (ADSCs) in post-myocardial infarction (MI) myocardial repair. Methods and Results Rat ADSCs were cultivated and then identified by morphologic observation, osteogenesis and adipogenesis induction assays and flow cytometry. Afterwards, ADSCs were modified by miR-324-5p lentiviral vector, with ADSC proliferation and migration measured. Then, rat MI model was established, which was treated by ADSCs or miR-324-5p-modified ADSCs. Subsequently, the function of miR-324-5p-modified ADSCs in myocardial repair of MI rats was assessed through functional assays. Next, the binding relation of miR-324-5p and Toll-interacting protein (TOLLIP) was validated. Eventually, functional rescue assay of TOLLIP was performed to verify the role of TOLLIP in MI. First, rat ADSCs were harvested. Overexpressed miR-324-5p improved ADSC viability. ADSC transplantation moderately enhanced cardiac function of MI rats, reduced enzyme levels and decreased infarct size and apoptosis; while miR-324-5p-modified ADSCs could better promote post-MI repair. Mechanically, miR-324-5p targeted TOLLIP in myocardial tissues. Moreover, TOLLIP overexpression debilitated the promotive role of miR-324-5p-modified ADSCs in post-MI repair in rats. Conclusions miR-324-5p-modified ADSCs evidently strengthened post-MI myocardial repair by targeting TOLLIP in myocardial tissues.
Collapse
Affiliation(s)
- Zhou Ji
- Department of Cardiovascular Medicine, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Chan Wang
- Jinzhou Hospital of Traditional Chinese Medicine, Jinzhou, China
| | - Qing Tong
- Office of Academic Research, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
32
|
Poznyak AV, Bezsonov EE, Popkova TV, Starodubova AV, Orekhov AN. Immunity in Atherosclerosis: Focusing on T and B Cells. Int J Mol Sci 2021; 22:ijms22168379. [PMID: 34445084 PMCID: PMC8395064 DOI: 10.3390/ijms22168379] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 07/27/2021] [Accepted: 07/30/2021] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis is the major cause of the development of cardiovascular disease, which, in turn, is one of the leading causes of mortality worldwide. From the point of view of pathogenesis, atherosclerosis is an extremely complex disease. A huge variety of processes, such as violation of mitophagy, oxidative stress, damage to the endothelium, and others, are involved in atherogenesis; however, the main components of atherogenesis are considered to be inflammation and alterations of lipid metabolism. In this review, we want to focus on inflammation, and more specifically on the cellular elements of adaptive immunity, T and B cells. It is known that various T cells are widely represented directly in atherosclerotic plaques, while B cells can be found, for example, in the adventitia layer. Of course, such widespread and well-studied cells have attracted attention as potential therapeutic targets for the treatment of atherosclerosis. Various approaches have been developed and tested for their efficacy.
Collapse
Affiliation(s)
- Anastasia V. Poznyak
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia
- Correspondence: (A.V.P.); (A.N.O.)
| | - Evgeny E. Bezsonov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia;
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
| | - Tatyana V. Popkova
- V.A. Nasonova Institute of Rheumatology, 34A Kashirskoye Shosse, 115522 Moscow, Russia;
| | - Antonina V. Starodubova
- Federal Research Centre for Nutrition, Biotechnology and Food Safety, 2/14 Ustinsky Passage, 109240 Moscow, Russia;
- Medical Faculty, Pirogov Russian National Research Medical University, 1 Ostrovitianov Street, 117997 Moscow, Russia
| | - Alexander N. Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia;
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia
- Correspondence: (A.V.P.); (A.N.O.)
| |
Collapse
|
33
|
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
|
34
|
Khalifa AA, Rashad RM, El-Hadidy WF. Thymoquinone protects against cardiac mitochondrial DNA loss, oxidative stress, inflammation and apoptosis in isoproterenol-induced myocardial infarction in rats. Heliyon 2021; 7:e07561. [PMID: 34355083 PMCID: PMC8322274 DOI: 10.1016/j.heliyon.2021.e07561] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 03/28/2021] [Accepted: 07/09/2021] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Myocardial infarction (MI) is an ischemic life-threatening disease with exaggerated oxidative stress state that vigorously damages the cardiomyocyte membrane and subcellular structures, including the vital mitochondrial DNA (mtDNA). The mtDNA is responsible for the proper functionality of the mitochondria, which are abundant in cardiomyocytes due to their dynamic nature and energy production requirements. Furthermore, oxidative stress triggers an inflammatory cascade and eventual apoptosis, which exacerbates cardiac injuries and dysfunction. AIM The present study used an isoproterenol (ISP)-induced MI rat model to investigate the role of the main active constituent of Nigella Sativa seeds, thymoquinone (TQ), in preserving the cardiac mtDNA content and ameliorating oxidative stress, inflammation, and apoptosis. METHODS Rats in the (TQ + ISP) group were pre-treated with TQ (20 mg/kg/day) for 21 days before the MI induction using ISP (85 mg/kg/day). In addition, negative control and ISP groups were included in the study for comparison. A histopathological examination was performed and serum cardiac parameters (cTnI and LDH) were assessed. In addition, mtDNA content, oxidative stress parameters (MDA, GSH, SOD, GPx, and CAT), inflammatory mediators (IL-6, IL-1β, and TNF-α), and apoptosis markers (BAX, Bcl2, and caspase-3) were detected. RESULTS The results showed that pre- and co-treatment with TQ in the (TQ + ISP) group reversed the histoarchitecture changes, caused a significant decrease in serum cardiac markers, oxidative stress markers, inflammatory cytokines, the apoptosis process, and preserved the cardiac mtDNA content. CONCLUSION TQ is a cardioprotective agent with an extended effect on preserving the cardiac mtDNA content, in addition to its powerful antioxidant, anti-inflammatory, and anti-apoptotic action.
Collapse
Affiliation(s)
- Asmaa A. Khalifa
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Egypt
| | - Radwa M. Rashad
- Department of Pathology, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Wessam F. El-Hadidy
- Department of Pharmacology and Therapeutics, Faculty of Pharmacy, Pharos University in Alexandria, Egypt
- Department of Pharmacology and Experimental Therapeutics, Medical Research Institute, Alexandria University, Alexandria, Egypt
| |
Collapse
|
35
|
Hospitalization for acute coronary syndrome increases the long-term risk of pneumonia: a population-based cohort study. Sci Rep 2021; 11:9696. [PMID: 33958673 PMCID: PMC8102567 DOI: 10.1038/s41598-021-89038-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/15/2021] [Indexed: 11/30/2022] Open
Abstract
It is well established that the risk of acute coronary syndrome (ACS) increases after respiratory infection. However, the reverse association has not been evaluated. We tested the hypothesis that the long-term risk of pneumonia is increased after a new ACS event. A matched-cohort study was conducted using a nationally representative dataset. We identified patients with admission for ACS between 2004 and 2014, without a previous history of ACS or pneumonia. Incidence density sampling was used to match patients, on the basis of age and sex, to 3 controls who were also free from both ACS and pneumonia. We examined the incidence of pneumonia after ACS until the end of the cohort observation (Dec 31, 2014). The analysis cohort consisted of 5469 ACS cases and 16,392 controls (median age, 64 years; 68.3% men). The incidence rate ratios of the first and the total pneumonia episodes in the ACS group relative to the control group was 1.25 (95% confidence interval [CI], 1.11–1.41) and 1.23(95% CI 1.11–1.36), respectively. A significant ACS-related increase in the incidence of pneumonia was observed in the Cox-regression, shared frailty, and joint frailty model analyses, with hazard ratios of 1.25 (95% CI 1.09–1.42), 1.35 (95% CI 1.15–1.58), and 1.24 (95% CI 1.10–1.39), respectively. In this population-based cohort of patients who were initially free from both ACS and pneumonia, we found that hospitalization for ACS substantially increased the long term risk of pneumonia. This should be considered when formulating post-discharge care plans and preventive vaccination strategies in patients with ACS.
Collapse
|
36
|
Shentu W, Ozawa K, Nguyen TA, Wu MD, Packwood W, Xie A, Muller MA, Brown E, Hagen MW, López JA, Lindner JR. Echocardiographic Molecular Imaging of the Effect of Anticytokine Therapy for Atherosclerosis. J Am Soc Echocardiogr 2021; 34:433-442.e3. [PMID: 33253812 PMCID: PMC8026579 DOI: 10.1016/j.echo.2020.11.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/13/2020] [Accepted: 11/18/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND Echocardiographic molecular imaging techniques are beginning to be applied to evaluate preclinical efficacy of new drugs. In a large clinical trial, anti-interleukin-1β (IL-1β) immunotherapy reduced atherosclerotic events, yet treatment effects were modest, and the mechanisms of action were not fully elucidated. We tested the hypothesis that echocardiographic molecular imaging can assess changes in vascular thromboinflammatory status in response to anti-IL-1β therapy. METHODS In wild-type and atherosclerotic mice deficient for the low-density lipoprotein-receptor and Apobec-1, closed-chest myocardial infarction (MI) was performed to mimic high-risk clinical cohorts. Control animals had sham surgery. Post-MI animals were randomized to either no therapy or anti-IL-1β immunotherapy, which was continued weekly. At post-MI day 3 or 21, in vivo ultrasound molecular imaging of aortic VCAM-1, P-selectin, von Willebrand factor A1-domain, and platelet GPIbα in the thoracic aorta was performed. Aortic histology and NF-κB activity were assessed in atherosclerotic mice. RESULTS In both atherosclerotic and wild-type mice, MI produced a several-fold increase (P < .05) in aortic molecular signals for P-selectin, VCAM-1, von Willebrand factor, and GPIbα. In atherosclerotic mice, signal remained elevated at day 21. Anti-IL-1β therapy completely abolished the post-MI increase in signal for all endothelial targets (P < .05 vs nontreated) at day 3 and 21. In atherosclerotic mice, MI triggered an increase in aortic plaque growth and macrophage content, a decrease in plaque collagen, and elevated aortic NF-κB (P < .05 for all changes). All of these remote plaque adverse changes were inhibited by anti-IL-1β therapy. CONCLUSIONS Echocardiographic molecular imaging of the vascular endothelium can quantify the beneficial effects of therapies designed to suppress the proatherosclerotic arterial thromboinflammatory effects of alarmins such as IL-1β. This approach could potentially be used to evaluate the biologic variables that influence response in preclinical studies, and possibly to select patients most likely to benefit from therapy.
Collapse
Affiliation(s)
- Weihui Shentu
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - Koya Ozawa
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - The Anh Nguyen
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - Melinda D Wu
- Division of Pediatric Hematology and Oncology, Oregon Health and Science University, Portland, Oregon
| | - William Packwood
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - Aris Xie
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - Matthew A Muller
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - Eran Brown
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - Matthew W Hagen
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon
| | - José A López
- Knight Cardiovascular Institute, Oregon Health and Science University, Portland, Oregon; Bloodworks Research Institute, Seattle, Washington
| | - Jonathan R Lindner
- Oregon National Primate Research Center, Oregon Health and Science University, Portland, Oregon.
| |
Collapse
|
37
|
Szabo D, Sarszegi Z, Polgar B, Saghy E, Nemeth A, Reglodi D, Makkos A, Gorbe A, Helyes Z, Ferdinandy P, Herczeg R, Gyenesei A, Cziraki A, Tamas A. PACAP-38 in Acute ST-Segment Elevation Myocardial Infarction in Humans and Pigs: A Translational Study. Int J Mol Sci 2021; 22:2883. [PMID: 33809145 PMCID: PMC8002092 DOI: 10.3390/ijms22062883] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/03/2021] [Accepted: 03/06/2021] [Indexed: 12/14/2022] Open
Abstract
Acute myocardial infarction (MI) is one of the most common causes of death worldwide. Pituitary adenylate cyclase activating polypeptide (PACAP) is a cardioprotective neuropeptide expressing its receptors in the cardiovascular system. The aim of our study was to examine tissue PACAP-38 in a translational porcine MI model and plasma PACAP-38 levels in patients with ST-segment elevation myocardial infarction (STEMI). Significantly lower PACAP-38 levels were detected in the non-ischemic region of the left ventricle (LV) in MI heart compared to the ischemic region of MI-LV and also to the Sham-operated LV in porcine MI model. In STEMI patients, plasma PACAP-38 level was significantly higher before percutaneous coronary intervention (PCI) compared to controls, and decreased after PCI. Significant negative correlation was found between plasma PACAP-38 and troponin levels. Furthermore, a significant effect was revealed between plasma PACAP-38, hypertension and HbA1c levels. This was the first study showing significant changes in cardiac tissue PACAP levels in a porcine MI model and plasma PACAP levels in STEMI patients. These results suggest that PACAP, due to its cardioprotective effects, may play a regulatory role in MI and could be a potential biomarker or drug target in MI.
Collapse
Affiliation(s)
- Dora Szabo
- Heart Institute, Medical School, University of Pecs, 7624 Pecs, Hungary; (D.S.); (Z.S.); (A.N.); (A.C.)
- Department of Anatomy, MTA-PTE PACAP Research Team, Centre for Neuroscience, Medical School, University of Pecs, 7624 Pecs, Hungary;
| | - Zsolt Sarszegi
- Heart Institute, Medical School, University of Pecs, 7624 Pecs, Hungary; (D.S.); (Z.S.); (A.N.); (A.C.)
| | - Beata Polgar
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, 7624 Pecs, Hungary;
| | - Eva Saghy
- MTA-SE System Pharmacology Research Group and Cardiovascular and Metabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (E.S.); (A.M.); (A.G.); (P.F.)
| | - Adam Nemeth
- Heart Institute, Medical School, University of Pecs, 7624 Pecs, Hungary; (D.S.); (Z.S.); (A.N.); (A.C.)
| | - Dora Reglodi
- Department of Anatomy, MTA-PTE PACAP Research Team, Centre for Neuroscience, Medical School, University of Pecs, 7624 Pecs, Hungary;
| | - Andras Makkos
- MTA-SE System Pharmacology Research Group and Cardiovascular and Metabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (E.S.); (A.M.); (A.G.); (P.F.)
| | - Aniko Gorbe
- MTA-SE System Pharmacology Research Group and Cardiovascular and Metabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (E.S.); (A.M.); (A.G.); (P.F.)
- Pharmahungary Group, 6722 Szeged, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pecs, 7624 Pecs, Hungary;
- Szentagothai Research Centre, University of Pecs, 7624 Pecs, Hungary; (R.H.); (A.G.)
- PharmInVivo Ltd., 7629 Pecs, Hungary
| | - Peter Ferdinandy
- MTA-SE System Pharmacology Research Group and Cardiovascular and Metabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (E.S.); (A.M.); (A.G.); (P.F.)
- Pharmahungary Group, 6722 Szeged, Hungary
| | - Robert Herczeg
- Szentagothai Research Centre, University of Pecs, 7624 Pecs, Hungary; (R.H.); (A.G.)
| | - Attila Gyenesei
- Szentagothai Research Centre, University of Pecs, 7624 Pecs, Hungary; (R.H.); (A.G.)
| | - Attila Cziraki
- Heart Institute, Medical School, University of Pecs, 7624 Pecs, Hungary; (D.S.); (Z.S.); (A.N.); (A.C.)
| | - Andrea Tamas
- Department of Anatomy, MTA-PTE PACAP Research Team, Centre for Neuroscience, Medical School, University of Pecs, 7624 Pecs, Hungary;
| |
Collapse
|
38
|
Tan L, Liu L, Yao J, Piao C. miR-145-5p attenuates inflammatory response and apoptosis in myocardial ischemia-reperfusion injury by inhibiting (NADPH) oxidase homolog 1. Exp Anim 2021; 70:311-321. [PMID: 33658472 PMCID: PMC8390312 DOI: 10.1538/expanim.20-0160] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Myocardial ischemia-reperfusion (I/R) injury is a common complication following reperfusion therapy that involves a series of immune or apoptotic reactions. Studies have revealed the potential roles of miRNAs in I/R injury. Herein, we established a myocardial I/R model in rats and a hypoxia/reoxygenation (H/R) model in H9c2 cells and investigated the effect of miR-145-5p on myocardial I/R injury. After 3 h or 24 h of reperfusion, left ventricular end-systolic pressure (LVESP), ejection fraction (EF), and fractional shortening (FS) were obviously decreased, and left ventricular end-diastolic pressure (LVEDP) was increased. Meanwhile, I/R induced an increase in myocardial infarction area. Moreover, a decrease in miR-145-5p and increase in (NADPH) oxidase homolog 1 (NOH-1) were observed following I/R injury. With this in mind, we performed a luciferase reporter assay and demonstrated that miR-145-5p directly bound to NOH-1 3' untranslated region (UTR). Furthermore, miR-145-5p mimics decreased the levels of tumor necrosis factor (TNF)-α, IL-1β, and IL-6 via oxygen and glucose deprivation/reperfusion (OGD/R) stimulation. Upregulation of miR-145-5p increased cell viability and reduced apoptosis accompanied by downregulation of Bax, cleaved caspase-3, cleaved poly(ADP-ribose) polymerase (PARP) and upregulation of Bcl2. In addition, miR-145-5p overexpression increased superoxide dismutase (SOD) activity and reduced reactive oxygen species (ROS) and malondialdehyde (MDA) content under OGD/R stress. Notably, NOH-1 could significantly abrogate the above effects, suggesting that it is involved in miR-145-5p-regulated I/R injury. In summary, our findings indicated that miR-145-5p/NOH-1 has a protective effect on myocardial I/R injury by inhibiting the inflammatory response and apoptosis.
Collapse
Affiliation(s)
- Lili Tan
- Department of Cardiology, The Second Affiliated Hospital of Shenyang Medical College, 20 Beijiu Road, Shenyang 110002, P.R. China
| | - Limin Liu
- Department of Cardiology, The Second Affiliated Hospital of Shenyang Medical College, 20 Beijiu Road, Shenyang 110002, P.R. China
| | - Jian Yao
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Shenyang Medical College, 20 Beijiu Road, Shenyang 110002, P.R. China
| | - Chenghao Piao
- Department of Cardiology, The Second Affiliated Hospital of Shenyang Medical College, 20 Beijiu Road, Shenyang 110002, P.R. China
| |
Collapse
|
39
|
Procopio MC, Lauro R, Nasso C, Carerj S, Squadrito F, Bitto A, Di Bella G, Micari A, Irrera N, Costa F. Role of Adenosine and Purinergic Receptors in Myocardial Infarction: Focus on Different Signal Transduction Pathways. Biomedicines 2021; 9:biomedicines9020204. [PMID: 33670488 PMCID: PMC7922652 DOI: 10.3390/biomedicines9020204] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 02/15/2021] [Indexed: 12/24/2022] Open
Abstract
Myocardial infarction (MI) is a dramatic event often caused by atherosclerotic plaque erosion or rupture and subsequent thrombotic occlusion of a coronary vessel. The low supply of oxygen and nutrients in the infarcted area may result in cardiomyocytes necrosis, replacement of intact myocardium with non-contractile fibrous tissue and left ventricular (LV) function impairment if blood flow is not quickly restored. In this review, we summarized the possible correlation between adenosine system, purinergic system and Wnt/β-catenin pathway and their role in the pathogenesis of cardiac damage following MI. In this context, several pathways are involved and, in particular, the adenosine receptors system shows different interactions between its members and purinergic receptors: their modulation might be effective not only for a normal functional recovery but also for the treatment of heart diseases, thus avoiding fibrosis, reducing infarcted area and limiting scaring. Similarly, it has been shown that Wnt/β catenin pathway is activated following myocardial injury and its unbalanced activation might promote cardiac fibrosis and, consequently, LV systolic function impairment. In this regard, the therapeutic benefits of Wnt inhibitors use were highlighted, thus demonstrating that Wnt/β-catenin pathway might be considered as a therapeutic target to prevent adverse LV remodeling and heart failure following MI.
Collapse
Affiliation(s)
- Maria Cristina Procopio
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Rita Lauro
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Chiara Nasso
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Scipione Carerj
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Francesco Squadrito
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Gianluca Di Bella
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Antonio Micari
- Department of Biomedical and Dental Sciences and Morphological and Functional Imaging, University of Messina, A.O.U. Policlinic “G. Martino”, 98165 Messina, Italy;
| | - Natasha Irrera
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
- Correspondence: ; Tel.: +39-090-221-3093; Fax: +39-090-221-23-81
| | - Francesco Costa
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| |
Collapse
|
40
|
Yang H, Liu S, Du H, Hong Z, Lv Y, Nie C, Yang W, Gao Y. Hydrogen Attenuates Myocardial Injury in Rats by Regulating Oxidative Stress and NLRP3 Inflammasome Mediated Pyroptosis. Int J Med Sci 2021; 18:3318-3325. [PMID: 34400901 PMCID: PMC8364469 DOI: 10.7150/ijms.61329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/16/2021] [Indexed: 11/26/2022] Open
Abstract
Purpose: Hydrogen (H2) is an antioxidant with anti-inflammatory and apoptosis functions.This study aimed to estimate the effects of H2 on acute myocardial infarction (AMI) in rats and its association with the inhibition of oxidative stress and cardiomyocyte pyroptosis. Methods: Sixty-four rats were randomly divided into three groups (Sham, AMI, and H2). The left anterior descending coronary artery (LAD) of rats in the AMI and H2 groups was ligated, while rats in the Sham group were threaded without ligation. In addition, 2% H2 was administered by inhalation for 24 h after ligation in the H2 group. Transthoracic echocardiography was performed after H2 inhalation, followed by collection of the serum and cardiac tissue of all rats. Results: H2 inhalation ameliorated the cardiac dysfunction, infarct size and inflammatory cell infiltration caused by AMI. Meanwhile, H2 inhalation reduced the concentration of serum Troponin I (TnI), brain natriuretic peptide (BNP), reactive oxygen species (ROS), cardiac malondialdehyde (MDA), and 8-OHdG. In addition, H2 inhalation inhibited cardiac inflammation and pyroptosis relative proteins expression. Conclusion: H2 effectively promoted heart functions in AMI rats by regulating oxidative stress and pyroptosis.
Collapse
Affiliation(s)
- Hongxiao Yang
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shuang Liu
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Huijun Du
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zi Hong
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yajing Lv
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Chaoqun Nie
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Wei Yang
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yunan Gao
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
41
|
Lai XX, Zhang N, Chen LY, Luo YY, Shou BY, Xie XX, Liu RH. Latifolin protects against myocardial infarction by alleviating myocardial inflammatory via the HIF-1α/NF-κB/IL-6 pathway. PHARMACEUTICAL BIOLOGY 2020; 58:1156-1166. [PMID: 33222562 PMCID: PMC7717487 DOI: 10.1080/13880209.2020.1840597] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 09/02/2020] [Accepted: 10/16/2020] [Indexed: 06/11/2023]
Abstract
CONTEXT The Traditional Chinese herb medicine Dalbergia odorifera T. Chen (Fabaceae), exerted a protective effect on myocardial ischaemia. Latifolin is a neoflavonoid extracted from Dalbergia odorifera. It has been reported to have the effects of anti-inflammation and cardiomyocyte protection. OBJECTIVE To investigate whether latifolin can improve myocardial infarction (MI) through attenuating myocardial inflammatory and to explore its possible mechanisms. MATERIALS AND METHODS Left coronary artery was ligated to induce a rat model of MI, and the rats were treated with sodium carboxymethyl cellulose (CMC-Na) or different doses of latifolin (25, 50, 100 mg/kg/d) by oral gavage for 28 days. Serum contents of myocardial enzyme were measured at seven and fourteen days after treatment. Cardiac function, infarct size, histopathological changes and inflammatory cells infiltration was assessed at 28 days after treatment. Western blotting was used to investigate the underlying mechanisms. RESULTS Latifolin treatment markedly decreased the contents of myocardial enzymes, and increased left ventricular ejection fraction (85.27% vs. 59.11%) and left ventricular fractional shortening (62.71% vs. 45.53%). Latifolin was found to significantly reduced infarction size (27.78% vs. 39.07%), myocardial fibrosis and the numbers of macrophage infiltration (436 cells/mm2 vs. 690 cells/mm2). In addition, latifolin down-regulated the expression levels of hypoxia-inducible factor-1α (0.95-fold), phospho-nuclear factor-κB (0.2-fold) and interleukin-6 (1.11-fold). DISCUSSION AND CONCLUSIONS Latifolin can protect against myocardial infarction by improving myocardial inflammation through the HIF-1α/NF-κB/IL-6 signalling pathway. Accordingly, latifolin may be a promising drug for pharmacological treatment of ischaemic cardiovascular disease.
Collapse
Affiliation(s)
- Xiao-Xiao Lai
- National Pharmaceutical Engineering Centre for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Ni Zhang
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Lan-Ying Chen
- National Pharmaceutical Engineering Centre for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Ying-Ying Luo
- National Pharmaceutical Engineering Centre for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Bin-Yao Shou
- National Pharmaceutical Engineering Centre for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Xin-Xu Xie
- National Pharmaceutical Engineering Centre for Solid Preparation of Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Rong-Hua Liu
- School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| |
Collapse
|
42
|
Che J, Najer A, Blakney AK, McKay PF, Bellahcene M, Winter CW, Sintou A, Tang J, Keane TJ, Schneider MD, Shattock RJ, Sattler S, Stevens MM. Neutrophils Enable Local and Non-Invasive Liposome Delivery to Inflamed Skeletal Muscle and Ischemic Heart. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2003598. [PMID: 33103807 PMCID: PMC7613371 DOI: 10.1002/adma.202003598] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/03/2020] [Indexed: 05/24/2023]
Abstract
Uncontrolled inflammation is a major pathological factor underlying a range of diseases including autoimmune conditions, cardiovascular disease, and cancer. Improving localized delivery of immunosuppressive drugs to inflamed tissue in a non-invasive manner offers significant promise to reduce severe side effects caused by systemic administration. Here, a neutrophil-mediated delivery system able to transport drug-loaded nanocarriers to inflamed tissue by exploiting the inherent ability of neutrophils to migrate to inflammatory tissue is reported. This hybrid system (neutrophils loaded with liposomes ex vivo) efficiently migrates in vitro following an inflammatory chemokine gradient. Furthermore, the triggered release of loaded liposomes and reuptake by target macrophages is studied. The migratory behavior of liposome-loaded neutrophils is confirmed in vivo by demonstrating the delivery of drug-loaded liposomes to an inflamed skeletal muscle in mice. A single low-dose injection of the hybrid system locally reduces inflammatory cytokine levels. Biodistribution of liposome-loaded neutrophils in a human-disease-relevant myocardial ischemia reperfusion injury mouse model after i.v. injection confirms the ability of injected neutrophils to carry loaded liposomes to inflammation sites. This strategy shows the potential of nanocarrier-loaded neutrophils as a universal platform to deliver anti-inflammatory drugs to promote tissue regeneration in inflammatory diseases.
Collapse
Affiliation(s)
- Junyi Che
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Adrian Najer
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Anna K Blakney
- Department of Infectious Diseases, Imperial College London, London, W2 1PG, UK
| | - Paul F McKay
- Department of Infectious Diseases, Imperial College London, London, W2 1PG, UK
| | - Mohamed Bellahcene
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Charles W Winter
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Amalia Sintou
- National Heart and Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Jiaqing Tang
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Timothy J Keane
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Michael D Schneider
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Robin J Shattock
- Department of Infectious Diseases, Imperial College London, London, W2 1PG, UK
| | - Susanne Sattler
- National Heart and Lung Institute, Imperial College London, London, W12 0NN, UK
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering, and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
43
|
Osmak G, Baulina N, Koshkin P, Favorova O. Collapsing the list of myocardial infarction-related differentially expressed genes into a diagnostic signature. J Transl Med 2020; 18:231. [PMID: 32517814 PMCID: PMC7285786 DOI: 10.1186/s12967-020-02400-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/03/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Myocardial infarction (MI) is one of the most severe manifestations of coronary artery disease (CAD) and the leading cause of death from non-infectious diseases worldwide. It is known that the central component of CAD pathogenesis is a chronic vascular inflammation. However, the mechanisms underlying the changes that occur in T, B and NK lymphocytes, monocytes and other immune cells during CAD and MI are still poorly understood. One of those pathogenic mechanisms might be the dysregulation of intracellular signaling pathways in the immune cells. METHODS In the present study we performed a transcriptome profiling in peripheral blood mononuclear cells of MI patients and controls. The machine learning algorithm was then used to search for MI-associated signatures, that could reflect the dysregulation of intracellular signaling pathways. RESULTS The genes ADAP2, KLRC1, MIR21, PDGFD and CD14 were identified as the most important signatures for the classification model with L1-norm penalty function. The classifier output quality was equal to 0.911 by Receiver Operating Characteristic metric on test data. These results were validated on two independent open GEO datasets. Identified MI-associated signatures can be further assisted in MI diagnosis and/or prognosis. CONCLUSIONS Thus, our study presents a pipeline for collapsing the list of differential expressed genes, identified by high-throughput techniques, in order to define disease-associated diagnostic signatures.
Collapse
Affiliation(s)
- German Osmak
- National Medical Research Center for Cardiology, Moscow, 121552, Russia.
| | - Natalia Baulina
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| | - Philipp Koshkin
- National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | - Olga Favorova
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| |
Collapse
|
44
|
Ozaki Tan SJ, Floriano JF, Nicastro L, Emanueli C, Catapano F. Novel Applications of Mesenchymal Stem Cell-derived Exosomes for Myocardial Infarction Therapeutics. Biomolecules 2020; 10:E707. [PMID: 32370160 PMCID: PMC7277090 DOI: 10.3390/biom10050707] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/22/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of mortality and morbidity globally, representing approximately a third of all deaths every year. The greater part of these cases is represented by myocardial infarction (MI), or heart attack as it is better known, which occurs when declining blood flow to the heart causes injury to cardiac tissue. Mesenchymal stem cells (MSCs) are multipotent stem cells that represent a promising vector for cell therapies that aim to treat MI due to their potent regenerative effects. However, it remains unclear the extent to which MSC-based therapies are able to induce regeneration in the heart and even less clear the degree to which clinical outcomes could be improved. Exosomes, which are small extracellular vesicles (EVs) known to have implications in intracellular communication, derived from MSCs (MSC-Exos), have recently emerged as a novel cell-free vector that is capable of conferring cardio-protection and regeneration in target cardiac cells. In this review, we assess the current state of research of MSC-Exos in the context of MI. In particular, we place emphasis on the mechanisms of action by which MSC-Exos accomplish their therapeutic effects, along with commentary on the current difficulties faced with exosome research and the ongoing clinical applications of stem-cell derived exosomes in different medical contexts.
Collapse
Affiliation(s)
- Sho Joseph Ozaki Tan
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK; (S.J.O.T.); (J.F.F.); (L.N.)
| | - Juliana Ferreria Floriano
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK; (S.J.O.T.); (J.F.F.); (L.N.)
- Botucatu Medical School, Sao Paulo State University, Botucatu 18618687, Brazil
| | - Laura Nicastro
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK; (S.J.O.T.); (J.F.F.); (L.N.)
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK; (S.J.O.T.); (J.F.F.); (L.N.)
| | - Francesco Catapano
- National Heart and Lung Institute, Imperial College London, London W12 0NN, UK; (S.J.O.T.); (J.F.F.); (L.N.)
| |
Collapse
|
45
|
Oishi Y, Manabe I. Organ System Crosstalk in Cardiometabolic Disease in the Age of Multimorbidity. Front Cardiovasc Med 2020; 7:64. [PMID: 32411724 PMCID: PMC7198858 DOI: 10.3389/fcvm.2020.00064] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 03/27/2020] [Indexed: 12/11/2022] Open
Abstract
The close association among cardiovascular, metabolic, and kidney diseases suggests a common pathological basis and significant interaction among these diseases. Metabolic syndrome and cardiorenal syndrome are two examples that exemplify the interlinked development of disease or dysfunction in two or more organs. Recent studies have been sorting out the mechanisms responsible for the crosstalk among the organs comprising the cardiovascular, metabolic, and renal systems, including heart-kidney and adipose-liver signaling, among many others. However, it is also becoming clear that this crosstalk is not limited to just pairs of organs, and in addition to organ-organ crosstalk, there are also organ-system and organ-body interactions. For instance, heart failure broadly impacts various organs and systems, including the kidney, liver, lung, and nervous system. Conversely, systemic dysregulation of metabolism, immunity, and nervous system activity greatly affects heart failure development and prognosis. This is particularly noteworthy, as more and more patients present with two or more coexisting chronic diseases or conditions (multimorbidity) due in part to the aging of society. Advances in treatment also contribute to the increase in multimorbidity, as exemplified by cardiovascular disease in cancer survivors. To understand the mechanisms underlying the increasing burden of multimorbidity, it is vital to elucidate the multilevel crosstalk and communication within the body at the levels of organ systems, tissues, and cells. In this article, we focus on chronic inflammation as a key common pathological basis of cardiovascular and metabolic diseases, and discuss emerging mechanisms that drive chronic inflammation in the context of multimorbidity.
Collapse
Affiliation(s)
- Yumiko Oishi
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Ichiro Manabe
- Department of Disease Biology and Molecular Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| |
Collapse
|
46
|
Papageorgiou C, Synetos A, Tampakis K, Anninos H, Kontogiannis C, Kapelouzou A, Kanakakis I, Tousoulis D, Paraskevaidis I, Toutouzas K. Activated Clotting Time as a Marker of Inflammation in Hospitalized Patients. Clin Appl Thromb Hemost 2020; 26:1076029620929090. [PMID: 32479107 PMCID: PMC7427012 DOI: 10.1177/1076029620929090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/23/2020] [Accepted: 04/29/2020] [Indexed: 01/06/2023] Open
Abstract
Inflammation and coagulation pathways are implicated in circulatory disease, but their interaction has not been completely deciphered yet. In this study, we investigated the association of coagulation and inflammation indices (activated clotting time [ACT], C-reactive protein, neutrophils) in hospitalized patients. Blood samples were drawn from consecutive patients at admission and at 48 hours for the assessment of the aforementioned parameters (n = 63). Healthy controls matched for sex and age were also examined (n = 39). Activated clotting time positively correlated with CRP on admission (r = 0.354, P = .005), while the correlation was more robust on the second day (r = 0.775, P < .001). Activated clotting time was significantly more prolonged in patients with abnormal CRP or abnormal absolute neutrophil count compared to patients with normal inflammatory markers (U = 55.0, P < .001 and U = 310.5, P = .035, respectively). At 48 hours, a positive relationship was observed between ACT and relative percentage of neutrophils (r = 0.358, P = .004). These findings suggest a link between ACT and inflammation indices for the first time in humans. Further research is needed to determine whether these interrelations can be used to improve patient management.
Collapse
Affiliation(s)
- Christos Papageorgiou
- Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Andreas Synetos
- 1st Department of Cardiology, Hippokration Hospital, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Konstantinos Tampakis
- Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Hector Anninos
- Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Christos Kontogiannis
- Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Alkistis Kapelouzou
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Greece
| | - Ioannis Kanakakis
- Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Dimitrios Tousoulis
- 1st Department of Cardiology, Hippokration Hospital, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Ioannis Paraskevaidis
- Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - Konstantinos Toutouzas
- 1st Department of Cardiology, Hippokration Hospital, School of Medicine, National and Kapodistrian University of Athens, Greece
| |
Collapse
|
47
|
Liu Y, Wang X, Wang H, Hu T. Identification of key genes and pathways in abdominal aortic aneurysm by integrated bioinformatics analysis. J Int Med Res 2019; 48:300060519894437. [PMID: 31885343 PMCID: PMC7783286 DOI: 10.1177/0300060519894437] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Objectives To identify key genes associated with abdominal aortic aneurysm (AAA) by
integrating a microarray profile and a single-cell RNA-seq dataset. Methods The microarray profile of GSE7084 and the single-cell RNA-seq dataset were
obtained from the Gene Express Omnibus database. Differentially expressed
genes (DEGs) were chosen using the R package and annotated by Gene Ontology
and Kyoto Encyclopedia of Genes and Genomics analysis. The hub genes were
identified based on their degrees of interaction in the protein-protein
interaction (PPI) network. Expression of hub genes was determined using
single-cell RNA-seq analysis. Results In total, 507 upregulated and 842 downregulated DEGs were identified and
associated with AAA. The upregulated DEGs were enriched into 9 biological
processes and 10 biological pathways, which were closely involved in the
pathogenesis and progression of AAA. Based on the PPI network, we focused on
six hub genes, four of which were novel target genes compared with the known
aneurysm gene database. Using single-cell RNA-seq analysis, we explored the
four genes expressed in vascular cells of AAA: CANX,
CD44, DAXX, and
STAT1. Conclusions We identified key genes that may provide insight into the mechanism of AAA
pathogenesis and progression and that have potential to be therapeutic
targets.
Collapse
Affiliation(s)
- Yihai Liu
- Department of Cardiology, The
Affiliated Huaian No. 1 People’s Hospital of
Nanjing
Medical University, Huaian, China
| | - Xixi Wang
- Department of Neurology, Affiliated
Shanghai First People’s Hospital of
Nanjing
Medical University, Nanjing, China
| | - Hongye Wang
- Department of Cardiology, The
Affiliated Huaian No. 1 People’s Hospital of
Nanjing
Medical University, Huaian, China
| | - Tingting Hu
- Department of Cardiology, The
Affiliated Huaian No. 1 People’s Hospital of
Nanjing
Medical University, Huaian, China
- Tingting Hu, Department of Cardiology, the
Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Beijing
West Road 6, Huaian 223001, China.
| |
Collapse
|
48
|
Yue Y, Wang C, Benedict C, Huang G, Truongcao M, Roy R, Cimini M, Garikipati VNS, Cheng Z, Koch WJ, Kishore R. Interleukin-10 Deficiency Alters Endothelial Progenitor Cell-Derived Exosome Reparative Effect on Myocardial Repair via Integrin-Linked Kinase Enrichment. Circ Res 2019; 126:315-329. [PMID: 31815595 DOI: 10.1161/circresaha.119.315829] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Rationale: Systemic inflammation compromises the reparative properties of endothelial progenitor cell (EPC) and their exosomes on myocardial repair, although the underlying mechanism of loss of function of exosomes from inflamed EPCs is still obscure. Objective: To determine the mechanisms of IL-10 (interleukin-10) deficient-EPC-derived exosome dysfunction in myocardial repair and to investigate if modification of specific exosome cargo can rescue reparative activity. Methods and Results: Using IL-10 knockout mice mimicking systemic inflammation condition, we compared therapeutic effect and protein cargo of exosomes isolated from wild-type EPC and IL-10 knockout EPC. In a mouse model of myocardial infarction (MI), wild-type EPC-derived exosome treatment significantly improved left ventricle cardiac function, inhibited cell apoptosis, reduced MI scar size, and promoted post-MI neovascularization, whereas IL-10 knockout EPC-derived exosome treatment showed diminished and opposite effects. Mass spectrometry analysis revealed wild-type EPC-derived exosome and IL-10 knockout EPC-derived exosome contain different protein expression pattern. Among differentially expressed proteins, ILK (integrin-linked kinase) was highly enriched in both IL-10 knockout EPC-derived exosome as well as TNFα (tumor necrosis factor-α)-treated mouse cardiac endothelial cell-derived exosomes (TNFα inflamed mouse cardiac endothelial cell-derived exosome). ILK-enriched exosomes activated NF-κB (nuclear factor κB) pathway and NF-κB-dependent gene transcription in recipient endothelial cells and this effect was partly attenuated through ILK knockdown in exosomes. Intriguingly, ILK knockdown in IL-10 knockout EPC-derived exosome significantly rescued their reparative dysfunction in myocardial repair, improved left ventricle cardiac function, reduced MI scar size, and enhanced post-MI neovascularization in MI mouse model. Conclusions: IL-10 deficiency/inflammation alters EPC-derived exosome function, content and therapeutic effect on myocardial repair by upregulating ILK enrichment in exosomes, and ILK-mediated activation of NF-κB pathway in recipient cells, whereas ILK knockdown in exosomes attenuates NF-κB activation and reduces inflammatory response. Our study provides new understanding of how inflammation may alter stem cell-exosome-mediated cardiac repair and identifies ILK as a target kinase for improving progenitor cell exosome-based cardiac therapies.
Collapse
Affiliation(s)
- Yujia Yue
- From the Center for Translational Medicine (Y.Y., C.W., C.B., G.H., M.T., R.R., M.C. V.N.S.G., Z.C., W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Chunlin Wang
- From the Center for Translational Medicine (Y.Y., C.W., C.B., G.H., M.T., R.R., M.C. V.N.S.G., Z.C., W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Cindy Benedict
- From the Center for Translational Medicine (Y.Y., C.W., C.B., G.H., M.T., R.R., M.C. V.N.S.G., Z.C., W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Grace Huang
- From the Center for Translational Medicine (Y.Y., C.W., C.B., G.H., M.T., R.R., M.C. V.N.S.G., Z.C., W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - May Truongcao
- From the Center for Translational Medicine (Y.Y., C.W., C.B., G.H., M.T., R.R., M.C. V.N.S.G., Z.C., W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Rajika Roy
- From the Center for Translational Medicine (Y.Y., C.W., C.B., G.H., M.T., R.R., M.C. V.N.S.G., Z.C., W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Maria Cimini
- From the Center for Translational Medicine (Y.Y., C.W., C.B., G.H., M.T., R.R., M.C. V.N.S.G., Z.C., W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Venkata Naga Srikanth Garikipati
- From the Center for Translational Medicine (Y.Y., C.W., C.B., G.H., M.T., R.R., M.C. V.N.S.G., Z.C., W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Zhongjian Cheng
- From the Center for Translational Medicine (Y.Y., C.W., C.B., G.H., M.T., R.R., M.C. V.N.S.G., Z.C., W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Walter J Koch
- From the Center for Translational Medicine (Y.Y., C.W., C.B., G.H., M.T., R.R., M.C. V.N.S.G., Z.C., W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA.,Department of Pharmacology and Medicine (W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | - Raj Kishore
- From the Center for Translational Medicine (Y.Y., C.W., C.B., G.H., M.T., R.R., M.C. V.N.S.G., Z.C., W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA.,Department of Pharmacology and Medicine (W.J.K., R.K.), Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| |
Collapse
|
49
|
Wei Y, Lan Y, Zhong Y, Yu K, Xu W, Zhu R, Sun H, Ding Y, Wang Y, Zeng Q. Interleukin-38 alleviates cardiac remodelling after myocardial infarction. J Cell Mol Med 2019; 24:371-384. [PMID: 31746138 PMCID: PMC6933378 DOI: 10.1111/jcmm.14741] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/12/2019] [Accepted: 09/23/2019] [Indexed: 12/21/2022] Open
Abstract
Excessive immune‐mediated inflammatory reaction plays a deleterious role in ventricular remodelling after myocardial infarction (MI). Interleukin (IL)‐38 is a newly characterized cytokine of the IL‐1 family and has been reported to exert a protective effect in some autoimmune diseases. However, its role in cardiac remodelling post‐MI remains unknown. In this study, we found that the expression of IL‐38 was increased in infarcted heart after MI induced in C57BL/6 mice by permanent ligation of the left anterior descending artery. In addition, our data showed that ventricular remodelling after MI was significantly ameliorated after recombinant IL‐38 injection in mice. This amelioration was demonstrated by better cardiac function, restricted inflammatory response, attenuated myocardial injury and decreased myocardial fibrosis. Our results in vitro revealed that IL‐38 affects the phenotype of dendritic cells (DCs) and IL‐38 plus troponin I (TNI)‐treated tolerogenic DCs dampened adaptive immune response when co‐cultured with CD4+T cells. In conclusion, IL‐38 plays a protective effect in ventricular remodelling post‐MI, one possibility by influencing DCs to attenuate inflammatory response. Therefore, targeting IL‐38 may hold a new therapeutic potential in treating MI.
Collapse
Affiliation(s)
- Yuzhen Wei
- Institute of Cardiology, Union Hospital, TongJi Medical College, Huahzong University of Science and Technology, Wuhan, China
| | - Yin Lan
- Institute of Cardiology, Union Hospital, TongJi Medical College, Huahzong University of Science and Technology, Wuhan, China
| | - Yucheng Zhong
- Institute of Cardiology, Union Hospital, TongJi Medical College, Huahzong University of Science and Technology, Wuhan, China
| | - Kunwu Yu
- Institute of Cardiology, Union Hospital, TongJi Medical College, Huahzong University of Science and Technology, Wuhan, China
| | - Wenbin Xu
- Institute of Cardiology, Union Hospital, TongJi Medical College, Huahzong University of Science and Technology, Wuhan, China
| | - Ruirui Zhu
- Institute of Cardiology, Union Hospital, TongJi Medical College, Huahzong University of Science and Technology, Wuhan, China
| | - Haitao Sun
- Institute of Cardiology, Union Hospital, TongJi Medical College, Huahzong University of Science and Technology, Wuhan, China
| | - Yan Ding
- Institute of Cardiology, Union Hospital, TongJi Medical College, Huahzong University of Science and Technology, Wuhan, China
| | - Yue Wang
- Institute of Cardiology, Union Hospital, TongJi Medical College, Huahzong University of Science and Technology, Wuhan, China
| | - Qiutang Zeng
- Institute of Cardiology, Union Hospital, TongJi Medical College, Huahzong University of Science and Technology, Wuhan, China
| |
Collapse
|
50
|
Moccetti F, Brown E, Xie A, Packwood W, Qi Y, Ruggeri Z, Shentu W, Chen J, López JA, Lindner JR. Myocardial Infarction Produces Sustained Proinflammatory Endothelial Activation in Remote Arteries. J Am Coll Cardiol 2019; 72:1015-1026. [PMID: 30139430 DOI: 10.1016/j.jacc.2018.06.044] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 06/07/2018] [Accepted: 06/10/2018] [Indexed: 01/13/2023]
Abstract
BACKGROUND In the months after acute myocardial infarction (MI), risk for acute atherothrombotic events in nonculprit arteries increases several fold. OBJECTIVES This study investigated whether sustained proinflammatory and prothrombotic endothelial alterations occur in remote vessels after MI. METHODS Wild-type mice, atherosclerotic mice with double knockout (DKO) of the low-density lipoprotein receptor and Apobec-1, and DKO mice treated with the Nox-inhibitor apocynin were studied at baseline and at 3 and 21 days after closed-chest MI. Ultrasound molecular imaging of P-selectin, vascular cell adhesion molecule (VCAM)-1, von Willebrand factor (VWF) A1-domain, and platelet GPIbα was performed. Intravital microscopy was used to characterize post-MI leukocyte and platelet recruitment in the remote microcirculation after MI. RESULTS Aortic molecular imaging for P-selectin, VCAM-1, VWF-A1, and platelets was increased several-fold (p < 0.01) 3 days post-MI for both wild-type and DKO mice. At 21 days, these changes resolved in wild-type mice but persisted in DKO mice. Signal for platelet adhesion was abolished 1 h after administration of ADAMTS13, which regulates VWF multimerization. In DKO and wild-type mice, apocynin significantly attenuated the post-MI increase for molecular targets, and platelet depletion significantly reduced P-selectin and VCAM-1 signal. On intravital microscopy, MI resulted in remote vessel leukocyte adhesion and platelet string or net complexes. On histology, high-risk inflammatory features in aortic plaque increased in DKO mice 21 days post-MI, which were completely prevented by apocynin. CONCLUSIONS Acute MI stimulates a spectrum of changes in remote vessels, including up-regulation of endothelial inflammatory adhesion molecules and platelet-endothelial adhesion from endothelial-associated VWF multimers. These remote arterial alterations persist longer in the presence of hyperlipidemia, are associated with accelerated plaque growth and inflammation, and are attenuated by Nox inhibition.
Collapse
Affiliation(s)
- Federico Moccetti
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Eran Brown
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Aris Xie
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - William Packwood
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Yue Qi
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | - Zaverio Ruggeri
- Department of Molecular and Experimental Medicine, Scripps Research Institute, La Jolla, California
| | - Weihui Shentu
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon
| | | | | | - Jonathan R Lindner
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon; Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon.
| |
Collapse
|