1
|
Zhou Z, Li T, Qin H, Wang X, He S, Fan Z, Ye Q, Du Y. Acacetin as a natural cardiovascular therapeutic: mechanisms and preclinical evidence. Front Pharmacol 2025; 16:1493981. [PMID: 40255574 PMCID: PMC12006078 DOI: 10.3389/fphar.2025.1493981] [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: 09/10/2024] [Accepted: 02/06/2025] [Indexed: 04/22/2025] Open
Abstract
Globally, cardiovascular disease (CVD) has emerged as a leading cause of mortality and morbidity. As the world's population ages, CVD incidence is on the rise, and extensive attention has been drawn to optimizing the therapeutic regimens. Acacetin, a natural flavonoid derived from various plants, has been demonstrated to have a wide spectrum of pharmacological properties, such as antioxidant, anti-inflammatory, anti-bacterial, and anti-tumor activities, as well as protective effects on diverse tissues and organs. Recently, increasing numbers of studies (mostly preclinical) have indicated that acacetin has potential cardiovascular protective effects and might become a novel therapeutic strategy for CVDs. The importance of acacetin in CVD treatment necessitates a systematic and comprehensive review of its protective effects on the cardiovascular system and the underlying mechanisms involved. Here, we first provide an overview of some basic properties of acacetin. Subsequently, the protective effects of acacetin on multiple CVDs, like arrhythmias, cardiac ischemia/reperfusion injury, atherosclerosis, myocardial hypertrophy and fibrosis, drug-induced cardiotoxicity, diabetic cardiomyopathy, hypertension, and cardiac senescence, are discussed in detail. The underlying mechanisms by which acacetin exhibits cardiovascular protection appear to involve suppressing oxidative stress, reducing inflammation, preventing cardiomyocyte apoptosis and endothelial cell injury, as well as regulating mitochondrial autophagy and lipid metabolism. Meanwhile, several critical signaling pathways have also been found to mediate the protection of acacetin against CVDs, including phosphoinositide 3-kinase/protein kinase B/mechanistic target of rapamycin (PI3K/Akt/mTOR), sirtuin 1/AMP-activated protein kinase/peroxisome proliferator-activated receptor-γ coactivator-1α (Sirt1/AMPK/PGC-1α), transforming growth factor-β1/small mothers against decapentaplegic 3 (TGF-β1/Smad3), protein kinase B/endothelial nitric oxide synthase (Akt/eNOS), and others. Finally, we highlight the existing problems associated with acacetin that need to be addressed, such as the requirement for clinical evidence and enhanced bioavailability, as well as its potential as a promising cardiovascular drug candidate.
Collapse
Affiliation(s)
- Zihe Zhou
- Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Department of Clinical Medicine, School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Tao Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological key Laboratory of Sichuan Province, Institute of Cardiovascular Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Helin Qin
- Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Xinyu Wang
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological key Laboratory of Sichuan Province, Institute of Cardiovascular Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Shanshan He
- Department of Basic Medicine, School of Basic Medical Science, Southwest Medical University, Luzhou, Sichuan, China
| | - Zhongcai Fan
- Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Qiang Ye
- Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Yanfei Du
- Department of Cardiology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological key Laboratory of Sichuan Province, Institute of Cardiovascular Medicine, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
2
|
Liakos CI, Aggeli C, Toutouzas KP, Markou MI, Vyssoulis GP, Tsioufis C. Acute exercise-induced inflammatory and thrombotic response in hypertensive patients. Eur J Appl Physiol 2025; 125:339-351. [PMID: 39249538 DOI: 10.1007/s00421-024-05599-9] [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: 03/09/2024] [Accepted: 08/24/2024] [Indexed: 09/10/2024]
Abstract
PURPOSE Vigorous physical activity may acutely trigger the onset of an acute coronary syndrome especially in sedentary persons with established cardiovascular risk factors such as arterial hypertension. The rupture of an inflamed coronary plaque and the activation of the coagulation cascade are the main underlying mechanisms. The present study aimed to determine the effect of acute exercise on the inflammatory and thrombotic response in patients with arterial hypertension as compared to normotensive peers. METHODS After excluding patients with any inflammatory or/and coronary artery disease, a total of 60 non-treated hypertensive patients and 65 normotensive individuals underwent a maximal treadmill exercise testing. Βlood samples were drawn at rest and immediately after peak exercise. High-sensitivity C-reactive protein (hsCRP), serum amyloid A (SAA), white blood cell (WBC), interleukin-6 (IL-6), and total fibrinogen (TF) levels, as well as plasminogen activator inhibitor-1 (PAI-1) activity, were measured. RESULTS All biomarkers increased with exercise, except PAI-1, which decreased (P < 0.05 for the change between resting and peak exercise for all biomarkers). Αfter adjusting for relevant confounders (duration of exercise, metabolic equivalents, systolic BP, and rate-pressure product achieved at peak exercise), the normotensive group had less marked (P < 0.05) exercise-induced changes than the hypertensive group in hsCRP (7.7 vs. 8.6%), SAA (5.6 vs. 11.9%), WBC (45.0 vs. 51.7%), and PAI-1 (-17.3 vs. -20.1%) and a similar (P = NS) change in IL-6 (23.8 vs. 23.0%) and TF (8.5 vs. 8.5%). CONCLUSION In conclusion, the acute exercise-induced inflammatory and thrombotic response seems to be more pronounced in non-treated hypertensive patients than in normotensive controls. Possible clinical implications of this finding merit further examination.
Collapse
Affiliation(s)
- Charalampos I Liakos
- Department of Medicine, School of Health Sciences, 1st Cardiology Clinic, Hippokration General Hospital, National and Kapodistrian University of Athens, 114 V. Sofias Avenue, 11527, Athens, Greece.
| | - Constantina Aggeli
- Department of Medicine, School of Health Sciences, 1st Cardiology Clinic, Hippokration General Hospital, National and Kapodistrian University of Athens, 114 V. Sofias Avenue, 11527, Athens, Greece
| | - Konstantinos P Toutouzas
- Department of Medicine, School of Health Sciences, 1st Cardiology Clinic, Hippokration General Hospital, National and Kapodistrian University of Athens, 114 V. Sofias Avenue, 11527, Athens, Greece
| | - Maria I Markou
- 1st Internal Medicine Clinic, NIMTS Hospital, 11521, Athens, Greece
| | - Gregory P Vyssoulis
- Department of Medicine, School of Health Sciences, 1st Cardiology Clinic, Hippokration General Hospital, National and Kapodistrian University of Athens, 114 V. Sofias Avenue, 11527, Athens, Greece
| | - Constantinos Tsioufis
- Department of Medicine, School of Health Sciences, 1st Cardiology Clinic, Hippokration General Hospital, National and Kapodistrian University of Athens, 114 V. Sofias Avenue, 11527, Athens, Greece
| |
Collapse
|
3
|
Cai Y, Chen Q. Resveratrol: A Narrative Review Regarding Its Mechanisms in Mitigating Obesity-Associated Metabolic Disorders. Phytother Res 2025; 39:999-1019. [PMID: 39715730 DOI: 10.1002/ptr.8416] [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: 07/31/2024] [Revised: 12/04/2024] [Accepted: 12/07/2024] [Indexed: 12/25/2024]
Abstract
Resveratrol (RSV) is a naturally occurring astragalus-like polyphenolic compound with remarkable weight loss properties. However, the mechanism of RSV in treating obesity is unclear. In this narrative review, we explored electronic databases (PubMed) for research articles from 2021 to the present using the keywords "resveratrol" and "obesity". This article explores the mechanisms involved in the alleviation of obesity-related metabolic disorders by RSV. RSV affects obesity by modulating mitochondrial function, insulin signaling, and gut microbiota, regulating lipid metabolism, inhibiting oxidative stress, and regulating epigenetic regulation. Administering RSV to pregnant animals exhibits maternal and first-generation offspring benefits, and RSV administration to lactating animals has long-term benefits, which involve the epigenetic modulations by RSV. A comprehensive understanding of the epigenetic mechanisms of RSV regulation could help in developing drugs suitable for pregnancy preparation groups, pregnant women, and nursing infants.
Collapse
Affiliation(s)
- Yichen Cai
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiu Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
4
|
Chudek J, Pośpiech M, Chudek A, Holecki M, Puzianowska-Kuźnicka M. Osteoprotegerin as an Emerging Biomarker of Carotid Artery Stenosis? A Scoping Review with Meta-Analysis. Diagnostics (Basel) 2025; 15:219. [PMID: 39857103 PMCID: PMC11764218 DOI: 10.3390/diagnostics15020219] [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: 12/26/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Objective: In developed countries, stroke is the fifth cause of death, with a high mortality rate, and with recovery to normal neurological function in one-third of survivors. Atherosclerotic occlusive disease of the extracranial part of the internal carotid artery and related embolic complications are common preventable causes of ischemic stroke (IS), attributable to 7-18% of all first-time cases. Osteoprotegerin (OPG), a soluble member of the tumor necrosis factor receptor (TNFR) superfamily, is considered a modulator of vascular calcification linked to vascular smooth muscle cell proliferation and collagen production in atherosclerotic plaques. Therefore, OPG emerges as a potential biomarker (BM) of calcified carotid plaques and carotid artery stenosis (CAS). Methods: We performed a literature search of PubMed on OPG in CAS and atherosclerosis published until 2024. Results: Increased levels of serum OPG were reported in both patients with symptomatic and asymptomatic CAS, and higher values were observed in those with unstable atherosclerotic plaques. Notably, increased OPG levels were observed regardless of the location of atherosclerosis, including coronary and other peripheral arteries. In addition, chronic kidney disease, the most significant confounder disturbing the association between vascular damage and circulating OPG levels, decreases the usefulness of OPG as a BM in CAS. Conclusions: Osteoprotegerin may be considered an emerging BM of global rather than cerebrovascular atherosclerosis. Its diagnostic significance in identifying patients with asymptomatic CAS and their monitoring is limited.
Collapse
Affiliation(s)
- Jerzy Chudek
- Department of Internal Medicine and Oncological Chemotherapy, Medical Faculty in Katowice, Medical University of Silesia, 40-027 Katowice, Poland;
| | - Marta Pośpiech
- Department of Internal Medicine and Oncological Chemotherapy, Medical Faculty in Katowice, Medical University of Silesia, 40-027 Katowice, Poland;
| | - Anna Chudek
- Health Promotion and Obesity Management Unit, Department of Pathophysiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland;
| | - Michał Holecki
- Department of Internal, Autoimmune and Metabolic Diseases, School of Medicine, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Monika Puzianowska-Kuźnicka
- Department of Human Epigenetics, Mossakowski Medical Research Institute, Polish Academy of Sciences, 02-106 Warsaw, Poland;
- Department of Geriatrics and Gerontology, Medical Centre of Postgraduate Education, 01-813 Warsaw, Poland
| |
Collapse
|
5
|
Li F, Zhang Y, Wang Y, Cai X, Fan X. Cytokine Gene Variants as Predisposing Factors for the Development and Progression of Coronary Artery Disease: A Systematic Review. Biomolecules 2024; 14:1631. [PMID: 39766338 PMCID: PMC11726869 DOI: 10.3390/biom14121631] [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: 11/15/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025] Open
Abstract
Coronary artery disease (CAD) is the most prevalent form of cardiovascular disease. A growing body of research shows that interleukins (ILs), such as IL-8, IL-18 and IL-16, elicit pro-inflammatory responses and may play critical roles in the pathologic process of CAD. Single nucleotide polymorphisms (SNPs), capable of generating functional modifications in IL genes, appear to be associated with CAD risk. This study aims to evaluate the associations of ten previously identified SNPs of the three cytokines with susceptibility to or protection of CAD. A systematic review and meta-analysis were conducted using Pubmed, EMBASE, WOS, CENTRAL, CNKI, CBM, Weipu, WANFANG Data and Google Scholar databases for relevant literature published up to September 2024. Odds ratios (ORs) with 95% confidence intervals (CIs) were calculated for the four genetic models of the investigated SNPs in overall and subgroups analyses. Thirty-eight articles from 16 countries involving 14574 cases and 13001 controls were included. The present meta-analysis revealed no significant association between CAD and IL-8-rs2227306 or five IL-16 SNPs (rs8034928, rs3848180, rs1131445, rs4778889 and rs11556218). However, IL-8-rs4073 was significantly associated with an increased risk of CAD across all genetic models. In contrast, three IL-18 (rs187238, rs1946518 and rs1946519) variants containing minor alleles were associated with decreased risks of CAD under all models. Subgroups analyses by ethnicity indicated that IL-8-rs4073 conferred a significantly higher risk of CAD among Asians, including East, South and West Asians (allelic OR = 1.46, homozygous OR = 1.96, heterozygous OR = 1.47, dominant OR = 1.65), while it showed an inversely significant association with CAD risk in Caucasians (homozygous OR = 0.82, dominant OR = 0.85). Additionally, IL-18-rs187238 and IL-18-rs1946518 were significantly associated with reduced CAD risks in East Asians (for rs187238: allelic OR = 0.72, homozygous OR = 0.33, heterozygous OR = 0.73, dominant OR = 0.71; for rs1946518: allelic OR = 0.62, homozygous OR = 0.38, heterozygous OR = 0.49, dominant OR = 0.45). IL-18-rs187238 also demonstrated protective effects in Middle Eastern populations (allelic OR = 0.76, homozygous OR = 0.63, heterozygous OR = 0.72, dominant OR = 0.71). No significant associations were observed in South Asians or Caucasians for these IL-18 SNPs. Consistent with the overall analysis results, subgroups analyses further highlighted a significant association between IL-8-rs4073 and increased risk of acute coronary syndrome (heterozygous OR = 0.72). IL-18-rs187238 was significantly associated with decreased risks of myocardial infarction (MI) (allelic OR = 0.81, homozygous OR = 0.55, dominant OR = 0.80) and multiple vessel stenosis (allelic OR = 0.54, heterozygous OR = 0.45, dominant OR = 0.45). Similarly, IL-18-rs1946518 was significantly associated with reduced MI risk (allelic OR = 0.75, heterozygous OR = 0.68). These findings support the role of cytokine gene IL-8 and IL-18 variants as predisposing factors for the development and progression of CAD.
Collapse
Affiliation(s)
- Fang Li
- Correspondence: (F.L.); (X.F.); Tel.: +86-731-88872780 (F.L. & X.F.)
| | | | | | | | - Xiongwei Fan
- The Laboratory of Heart Development Research, College of Life Sciences, Hunan Normal University, Changsha 410081, China; (Y.Z.); (Y.W.); (X.C.)
| |
Collapse
|
6
|
Onnis C, Virmani R, Madra A, Nardi V, Salgado R, Montisci R, Cau R, Boi A, Lerman A, De Cecco CN, Libby P, Saba L. Whys and Wherefores of Coronary Arterial Positive Remodeling. Arterioscler Thromb Vasc Biol 2024; 44:2416-2427. [PMID: 39479766 PMCID: PMC11594009 DOI: 10.1161/atvbaha.124.321504] [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: 11/28/2024]
Abstract
Positive remodeling (PR) is an atherosclerotic plaque feature defined as an increase in arterial caliber at the level of an atheroma, in response to increasing plaque burden. The mechanisms that lead to its formation are incompletely understood, but its role in coronary atherosclerosis has major clinical implications. Indeed, plaques with PR have elevated risk of provoking acute cardiac events. Hence, PR figures among the high-risk plaque features that cardiac imaging studies should report. This review aims to provide an overview of the current literature on coronary PR. It outlines the pathophysiology of PR, the different techniques used to assess its presence, and the imaging findings associated to PR, on both noninvasive and invasive studies. This review also summarizes clinical observations, trials, and studies, focused on the impact of PR on clinical outcome.
Collapse
Affiliation(s)
- Carlotta Onnis
- Department of Radiology, Azienda Ospedaliero Universitaria, Cagliari, Italy (C.O., R.C., L.S.)
| | - Renu Virmani
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD (R.V., A.M.)
| | - Anna Madra
- Department of Cardiovascular Pathology, CVPath Institute, Gaithersburg, MD (R.V., A.M.)
| | - Valentina Nardi
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (V.N., A.L.)
| | - Rodrigo Salgado
- Department of Radiology, Antwerp University Hospital and Antwerp University Lier, Belgium (R.S.)
| | - Roberta Montisci
- Clinical Cardiology, Department of Medical Science and Public Health, University of Cagliari, Italy (R.M.)
| | - Riccardo Cau
- Department of Radiology, Azienda Ospedaliero Universitaria, Cagliari, Italy (C.O., R.C., L.S.)
| | - Alberto Boi
- Department of Cardiology, Azienda Ospedaliera Brotzu, Cagliari, Italy (A.B.)
| | - Amir Lerman
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (V.N., A.L.)
| | - Carlo N. De Cecco
- Division of Cardiothoracic Imaging and Biomedical Informatics, Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA (C.N.D.C.)
| | - Peter Libby
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA (P.L.)
| | - Luca Saba
- Department of Radiology, Azienda Ospedaliero Universitaria, Cagliari, Italy (C.O., R.C., L.S.)
| |
Collapse
|
7
|
Zhao M, Feng L, Li W. Network Pharmacology and Experimental Verification: SanQi-DanShen Treats Coronary Heart Disease by Inhibiting the PI3K/AKT Signaling Pathway. Drug Des Devel Ther 2024; 18:4529-4550. [PMID: 39399124 PMCID: PMC11471080 DOI: 10.2147/dddt.s480248] [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: 07/26/2024] [Accepted: 09/11/2024] [Indexed: 10/15/2024] Open
Abstract
Objective To employee network pharmacology to predict the components and pathways of SanQi-DanShen (SQDS) in treating coronary heart disease, followed by in vitro experiments to validate the molecular mechanism of SQDS in treating coronary heart disease. Methods We sourced the active ingredients and targets of Panax notoginseng and Danshen from the Traditional Chinese Medicine Systems Pharmacology database. Coronary heart disease related genes were retrieved from the OMIM, Genecards, and Therapeutic Target databases. Using Cytoscape 3.7.2 software, we constructed a network diagram illustrating the components and targets of SQDS. The associated targets were then imported into the STRING database to build a protein-protein interaction network. The Metascape database and WeChat software were utilized for Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses. Lastly, we performed molecular docking between the key components and related targets using AutoDock Vina. To validate the potential mechanism of SQDS in treating coronary heart disease, we established an acute coronary heart disease rat model via tail vein injection of pituitrin. Results Network pharmacology analysis revealed that 65 active ingredients and 167 targets of SQDS are implicated in the treatment of coronary heart disease. The key targets identified include AKT1, TNF, TP53, IL6, and VEGFA. Notably, the PI3K/AKT signaling pathway emerged as the primary pathway. Furthermore, animal experiments showed that, compared to the model group, SQDS significantly reduced levels of TNF-α, IL-6, Bax, and cardiac troponin I, while increasing Bcl-2 content. It also notably suppressed the expression of p-PI3K and p-AKT, thereby offering protection to myocardial tissue. Conclusion Through the integrated approach of network pharmacology and molecular docking, we have established that SQDS exerts a multi-component, multi-target, and multi-pathway synergistic therapeutic effect on coronary heart disease. Its mechanism may involve the inhibition of the PI3K/AKT signaling pathway and the reduction of inflammatory factor expression.
Collapse
Affiliation(s)
- Min Zhao
- School of Medicine, Lijiang University of Culture and Tourism, Lijiang, Yunnan, 674100, People’s Republic of China
| | - Liuxiang Feng
- People’s Hospital of Yulong Naxi Autonomous County of Lijiang City, Lijiang, Yunnan, 674112, People’s Republic of China
| | - Wenhua Li
- School of Medicine, Xizang Minzu University, Xianyang Shaanxi, 712082, People’s Republic of China
| |
Collapse
|
8
|
Aksu U, Korucu C, Ekşi RA, Gökaslan ÇÖ. The relationship between the systemic immune inflammatory index and computerized tomography guided coronary lesion severity in diabetic patients. JOURNAL OF CLINICAL ULTRASOUND : JCU 2024; 52:675-679. [PMID: 38578039 DOI: 10.1002/jcu.23680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/16/2024] [Accepted: 03/25/2024] [Indexed: 04/06/2024]
Abstract
PURPOSE The incidence of cardiovascular events is high in diabetic patients. In diabetic patients, the levels of inflammatory parameters in the circulation are increased, which is associated with poor outcome. In this study, we investigated the relationship between the systemic immune inflammatory index (SII), which is a sensitive indicator of the inflammatory response, and the severity of coronary atherosclerosis in diabetic patients. MATERIALS AND METHODS Diabetic patients who underwent coronary computed tomography for chest pain were included in the study. The patients were divided into two groups according to their median systemic immune inflammatory index values, and the predictors of SII elevation were investigated. RESULTS A total 210 patients were included in the study. The mean age of the patients was 52.6 ± 9.3 and 44.3% were male. In univariate analysis, HDL, triglyceride, lesion severity, and CAR were associated with high SII. In the regression analysis, lesion severity and lower HDL levels were determined as predictor of high SII. CONCLUSION Inflammation plays an important role in the development of coronary atherosclerosis. Diabetic patients with elevated SII levels may require further investigation for significant atherosclerosis.
Collapse
Affiliation(s)
- Uğur Aksu
- Faculty of Medicine, Department of Cardiology, Afyonkarahisar Health Sciences University, Afyon, Turkey
| | - Cem Korucu
- Faculty of Medicine, Department of Cardiology, Afyonkarahisar Health Sciences University, Afyon, Turkey
| | - Ramazan Anıl Ekşi
- Faculty of Medicine, Department of Cardiology, Afyonkarahisar Health Sciences University, Afyon, Turkey
| | - Çiğdem Özer Gökaslan
- Faculty of Medicine, Department of Radiology, Afyonkarahisar Health Sciences University, Afyon, Turkey
| |
Collapse
|
9
|
Mesa RA, Damas OM, Schneiderman N, Palacio AM, Gallo LC, Talavera GA, Sotres-Alvarez D, Daviglus ML, Pirzada A, Llabre MM, Elfassy T. Association Between High-Sensitivity C-Reactive Protein and Metabolic Syndrome Among Hispanic/Latino Participants of the Hispanic Community Health Study/Study of Latinos. Metab Syndr Relat Disord 2024; 22:327-336. [PMID: 38563777 DOI: 10.1089/met.2023.0298] [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: 04/04/2024] Open
Abstract
Purpose: To determine whether high-sensitivity C-reactive protein (hsCRP) is associated with incident Metabolic Syndrome (MetS) among U.S. Hispanic/Latino adults. Patients and Methods: The Hispanic Community Health Study/Study of Latinos is a longitudinal observational cohort assessing cardiovascular health among diverse U.S. Hispanic/Latino adults. hsCRP was measured at visit 1 (2008-2011) and classified as low, moderate, or high, based on the Centers for Disease Control and Prevention and American Heart Association (CDC/AHA) guidelines. All MetS components [abdominal obesity, triglycerides, high-density lipoprotein (HDL) cholesterol, blood pressure, and fasting glucose] were measured at visit 1 and visit 2 (2014-2017). MetS was defined as the presence of three or more components based on the 2005 definition from the modified Third Report of the National Cholesterol Education Program Adult Treatment Panel (modified NCEP ATP III). Participants free of MetS at visit 1 and with complete data on hsCRP and all MetS components were included (n = 6121 participants). We used Poisson regression analysis to determine whether hsCRP was associated with incident MetS after adjusting for demographic, behavioral, and clinical factors. All analyses accounted for the complex survey design of the study. Results: In fully adjusted models, moderate versus low hsCRP was associated with a 33% increased risk of MetS [incidence rate ratio (IRR): 1.33, 95% confidence interval (CI): 1.10-1.61], while high versus low hsCRP was associated with a 89% increased risk of MetS (IRR: 1.89, 95% CI: 1.58-2.25). Conclusions: Greater levels of hsCRP were associated with new onset of MetS in a diverse sample of U.S. Hispanic/Latino adults. Results suggest that hsCRP may be an independent risk factor for MetS.
Collapse
Affiliation(s)
- Robert A Mesa
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Oriana M Damas
- Division of Gastroenterology, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | - Ana M Palacio
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Linda C Gallo
- Department of Psychology, San Diego State University, San Diego, California, USA
| | - Gregory A Talavera
- Department of Psychology, Graduate School of Public Health, San Diego State University, San Diego, California, USA
| | - Daniela Sotres-Alvarez
- Collaborative Studies Coordinating Center, Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Martha L Daviglus
- Institute for Minority Health Research, University of Illinois Chicago, Chicago, Illinois, USA
| | - Amber Pirzada
- Institute for Minority Health Research, University of Illinois Chicago, Chicago, Illinois, USA
| | - Maria M Llabre
- Department of Psychology, University of Miami, Miami, Florida, USA
| | - Tali Elfassy
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
10
|
De Meyer GRY, Zurek M, Puylaert P, Martinet W. Programmed death of macrophages in atherosclerosis: mechanisms and therapeutic targets. Nat Rev Cardiol 2024; 21:312-325. [PMID: 38163815 DOI: 10.1038/s41569-023-00957-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/02/2023] [Indexed: 01/03/2024]
Abstract
Atherosclerosis is a progressive inflammatory disorder of the arterial vessel wall characterized by substantial infiltration of macrophages, which exert both favourable and detrimental functions. Early in atherogenesis, macrophages can clear cytotoxic lipoproteins and dead cells, preventing cytotoxicity. Efferocytosis - the efficient clearance of dead cells by macrophages - is crucial for preventing secondary necrosis and stimulating the release of anti-inflammatory cytokines. In addition, macrophages can promote tissue repair and proliferation of vascular smooth muscle cells, thereby increasing plaque stability. However, advanced atherosclerotic plaques contain large numbers of pro-inflammatory macrophages that secrete matrix-degrading enzymes, induce death in surrounding cells and contribute to plaque destabilization and rupture. Importantly, macrophages in the plaque can undergo apoptosis and several forms of regulated necrosis, including necroptosis, pyroptosis and ferroptosis. Regulated necrosis has an important role in the formation and expansion of the necrotic core during plaque progression, and several triggers for necrosis are present within atherosclerotic plaques. This Review focuses on the various forms of programmed macrophage death in atherosclerosis and the pharmacological interventions that target them as a potential means of stabilizing vulnerable plaques and improving the efficacy of currently available anti-atherosclerotic therapies.
Collapse
Affiliation(s)
- Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium.
| | - Michelle Zurek
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Pauline Puylaert
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
11
|
Iusupova AO, Slepova OA, Pakhtusov NN, Popova LV, Ageev AA, Lishuta AS, Privalova EV, Khabarova NV, Dadashovа GМ, Belenkov YN. Assessment of the Level of Matrix Metalloproteinases, VEGF and MicroRNA-34a in Patients With Non-obstructive and Obstructive Lesions of the Coronary Arteries. KARDIOLOGIIA 2024; 64:14-21. [PMID: 38742511 DOI: 10.18087/cardio.2024.4.n2622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/26/2024] [Indexed: 05/16/2024]
Abstract
AIM To assess the levels of matrix metalloproteinases (MMP), vascular endothelial growth factor (VEGF), and miRNA-34a expression in patients with ischemic heart disease (IHD) and obstructive and nonobstructive coronary artery (CA) disease. MATERIAL AND METHODS This cross-sectional observational study included 64 patients with IHD (diagnosis verified by coronary angiography or multislice computed tomography coronary angiography), of which 33 (51.6%) were men aged 64.9±8.1 years. 20 patients had nonobstructive CA disease (stenosis <50%), and 44 had hemodynamically significant stenoses. The control group consisted of 30 healthy volunteers. MMP-1, -9, -13, and -14, miRNA-34a, and VEGF were measured in all patients. RESULTS The concentration of MMP-1 was significantly higher in patients with ischemia and nonobstructive CA disease (INOCAD) (p=0.016), and the concentration of MMP-9 was the highest in the group with obstructive CA disease (p<0.001). The concentrations of MMP-13 and MMP-14 did not differ significantly between the groups. The highest VEGF concentrations were observed in the INOCAD group (p<0.001). The expression of miRNA-34a significantly differed between the IHD groups with different types of CA disease and controls (p <0.001). Patients with hemodynamically significant stenosis showed moderate relationships between the concentrations of MMP-14 and VEGF (ρ=0.418; p=0.024), as well as between VEGF and miRNA-34a (ρ=0.425; p=0.022). Patients with INOCAD had a significant negative correlation between the concentrations of MMP-13 and VEGF (ρ= -0.659; p=0.003). Correlation analysis showed in all IHD patients a moderate relationship of the concentrations of MMP-1 and MMP-14 with VEGF (ρ=0.449; p=0.002 and p=0.341; p=0.019, respectively). According to ROC analysis, a MMP-9 concentration above 4.83 ng/ml can be a predictor for the presence of hemodynamically significant CA obstruction in IHD patients; a VEGF concentration higher than 27.23 pg/ml suggests the absence of hemodynamically significant CA stenosis. CONCLUSION IHD patients with INOCAD had the greatest increase in MMP-1, whereas patients with obstructive CA disease had the highest level of MMP-9. According to our data, concentrations of MMP-9 and VEGF can be used to predict the degree of CA obstruction. The expression of miRNA-34a was significantly higher in IHD patients with INOCAD and CA obstruction than in the control group, which suggested a miRNA-34a contribution to the development and progression of coronary atherosclerosis. In the future, it may be possible to use this miRNA as a diagnostic marker for IHD.
Collapse
Affiliation(s)
- A O Iusupova
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - O A Slepova
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - N N Pakhtusov
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - L V Popova
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - A A Ageev
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - A S Lishuta
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - E V Privalova
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - N V Khabarova
- Sechenov First Moscow State Medical University, Moscow, Russia
| | | | - Yu N Belenkov
- Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
12
|
Meloni A, Maffei E, Clemente A, De Gori C, Occhipinti M, Positano V, Berti S, La Grutta L, Saba L, Cau R, Bossone E, Mantini C, Cavaliere C, Punzo B, Celi S, Cademartiri F. Spectral Photon-Counting Computed Tomography: Technical Principles and Applications in the Assessment of Cardiovascular Diseases. J Clin Med 2024; 13:2359. [PMID: 38673632 PMCID: PMC11051476 DOI: 10.3390/jcm13082359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Spectral Photon-Counting Computed Tomography (SPCCT) represents a groundbreaking advancement in X-ray imaging technology. The core innovation of SPCCT lies in its photon-counting detectors, which can count the exact number of incoming x-ray photons and individually measure their energy. The first part of this review summarizes the key elements of SPCCT technology, such as energy binning, energy weighting, and material decomposition. Its energy-discriminating ability represents the key to the increase in the contrast between different tissues, the elimination of the electronic noise, and the correction of beam-hardening artifacts. Material decomposition provides valuable insights into specific elements' composition, concentration, and distribution. The capability of SPCCT to operate in three or more energy regimes allows for the differentiation of several contrast agents, facilitating quantitative assessments of elements with specific energy thresholds within the diagnostic energy range. The second part of this review provides a brief overview of the applications of SPCCT in the assessment of various cardiovascular disease processes. SPCCT can support the study of myocardial blood perfusion and enable enhanced tissue characterization and the identification of contrast agents, in a manner that was previously unattainable.
Collapse
Affiliation(s)
- Antonella Meloni
- Bioengineering Unit, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.M.); (V.P.)
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.C.); (C.D.G.); (M.O.)
| | - Erica Maffei
- Department of Radiology, Istituto di Ricovero e Cura a Carattere Scientifico SYNLAB SDN, 80131 Naples, Italy; (E.M.); (C.C.); (B.P.)
| | - Alberto Clemente
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.C.); (C.D.G.); (M.O.)
| | - Carmelo De Gori
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.C.); (C.D.G.); (M.O.)
| | - Mariaelena Occhipinti
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.C.); (C.D.G.); (M.O.)
| | - Vicenzo Positano
- Bioengineering Unit, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.M.); (V.P.)
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.C.); (C.D.G.); (M.O.)
| | - Sergio Berti
- Diagnostic and Interventional Cardiology Department, Fondazione G. Monasterio CNR-Regione Toscana, 54100 Massa, Italy;
| | - Ludovico La Grutta
- Department of Radiology, University Hospital “P. Giaccone”, 90127 Palermo, Italy;
| | - Luca Saba
- Department of Radiology, University Hospital of Cagliari, 09042 Monserrato (CA), Italy; (L.S.); (R.C.)
| | - Riccardo Cau
- Department of Radiology, University Hospital of Cagliari, 09042 Monserrato (CA), Italy; (L.S.); (R.C.)
| | - Eduardo Bossone
- Department of Cardiology, Ospedale Cardarelli, 80131 Naples, Italy;
| | - Cesare Mantini
- Department of Radiology, “G. D’Annunzio” University, 66100 Chieti, Italy;
| | - Carlo Cavaliere
- Department of Radiology, Istituto di Ricovero e Cura a Carattere Scientifico SYNLAB SDN, 80131 Naples, Italy; (E.M.); (C.C.); (B.P.)
| | - Bruna Punzo
- Department of Radiology, Istituto di Ricovero e Cura a Carattere Scientifico SYNLAB SDN, 80131 Naples, Italy; (E.M.); (C.C.); (B.P.)
| | - Simona Celi
- BioCardioLab, Fondazione G. Monasterio CNR-Regione Toscana, 54100 Massa, Italy;
| | - Filippo Cademartiri
- Department of Radiology, Fondazione G. Monasterio CNR-Regione Toscana, 56124 Pisa, Italy; (A.C.); (C.D.G.); (M.O.)
| |
Collapse
|
13
|
Chen K, Liu Y, Xu B, Ye T, Chen L, Wu G, Zong G. Relationship between the lymphocyte to C‑reactive protein ratio and coronary artery disease severity. Exp Ther Med 2024; 27:60. [PMID: 38234629 PMCID: PMC10790159 DOI: 10.3892/etm.2023.12348] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/23/2023] [Indexed: 01/19/2024] Open
Abstract
Coronary atherosclerosis is a chronic systemic inflammatory disease. Laboratory parameters such as the neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR) and systemic immune inflammation index (SII) have been used to assess inflammation degree and coronary artery disease (CAD) severity. The lymphocyte-to-C-reactive protein ratio (LCR) is a new SII. However, its relationship with CAD development and severity is unclear. A total of 1,107 patients (479 in control group, 628 in CAD group) underwent coronary angiography. The routine and biochemical indices of the venous blood of patients were assessed before coronary angiography. LCR, SII, NLR and PLR were calculated and statistical analyses were performed. Propensity score matching (PSM) and a logistic regression model were used to analyze the relationship between LCR and CAD. After the PSM, 384 pairs of patients with or without CAD were successfully matched. After the median binary classification of all indicators, uni- and multivariate logistic regression analyses showed that platelet count was an independent risk factor and LCR was an independent protective factor. Using the same method, in the coronary heart disease severity group, 212 pairs were successfully matched and NLR and PLR were independent risk factors, while LCR was an independent protective factor. In conclusion, LCR is an independent protective factor against CAD development and severity.
Collapse
Affiliation(s)
- Ke Chen
- Department of Cardiology, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, P.R. China
- Department of Cardiology, The 904th Hospital of Joint Logistic Support Force of People's Liberation Army, Wuxi, Jiangsu 214044, P.R. China
| | - Yehong Liu
- Department of Cardiology, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, P.R. China
- Department of Cardiology, The 904th Hospital of Joint Logistic Support Force of People's Liberation Army, Wuxi, Jiangsu 214044, P.R. China
| | - Baida Xu
- Department of Cardiology, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, P.R. China
- Department of Cardiology, The 904th Hospital of Joint Logistic Support Force of People's Liberation Army, Wuxi, Jiangsu 214044, P.R. China
| | - Ting Ye
- Department of Cardiology, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, P.R. China
- Department of Cardiology, The 904th Hospital of Joint Logistic Support Force of People's Liberation Army, Wuxi, Jiangsu 214044, P.R. China
| | - Liang Chen
- Department of Cardiology, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, P.R. China
- Department of Cardiology, The 904th Hospital of Joint Logistic Support Force of People's Liberation Army, Wuxi, Jiangsu 214044, P.R. China
| | - Gangyong Wu
- Department of Cardiology, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, P.R. China
- Department of Cardiology, The 904th Hospital of Joint Logistic Support Force of People's Liberation Army, Wuxi, Jiangsu 214044, P.R. China
| | - Gangjun Zong
- Department of Cardiology, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, P.R. China
- Department of Cardiology, The 904th Hospital of Joint Logistic Support Force of People's Liberation Army, Wuxi, Jiangsu 214044, P.R. China
| |
Collapse
|
14
|
Chen R, Su S, Wang C, Liu Y, Huang W, Luo S, Yang F, Luo J. Systemic immune-inflammation index predicts the clinical outcomes in patients with acute uncomplicated type-B aortic dissection undergoing optimal medical therapy. BMC Cardiovasc Disord 2024; 24:7. [PMID: 38166807 PMCID: PMC10763462 DOI: 10.1186/s12872-023-03596-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 11/05/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Optimal medical therapy (OMT) for uncomplicated type B aortic dissection (uTBAD) provides excellent short-term outcomes during follow up; however, its long-term therapeutic effectiveness is unsatisfactory. This study evaluated the predictive value of systemic immune-inflammation index (SII) for adverse events among patients with acute uTBAD undergoing OMT. METHODS We performed a retrospective analysis of a prospectively maintained database between 2013 and 2020. The primary end point in this study was composite outcomes including aortic intervention, all-cause mortality, retrograde type A aortic dissection (rTAAD) and aortic diameter growth > 5 mm. The patients were divided into high and low SII groups according to the optimal cut-off value of SII as determined using the receiver operating characteristic curve. Cox proportional hazards models were constructed to estimate the hazards ratios and identify the predictors of composite outcomes. RESULTS A total of 124 patients with acute uTBAD who underwent OMT were enrolled. One patient died during hospitalisation. At the end of a mean follow-up duration of 51 ± 23 months, 53 (43.1%) patients experienced composite outcomes, 15 patients (12.2%) died, 31 (25.2%) underwent aortic intervention, 21 (17.1%) exhibited diameter growth of > 5 mm, and 2 developed rTAAD. The patients were divided into low SII group (n = 78, 62.9%) and high SII group (n = 46, 37.1%) as per the optimal cut-off SII value of 1449. The incidence of composite outcomes in high SII group was significantly higher than that in low SII (28 [60.9%] vs. 26[33.3%], p < 0.01). Patients with high SII demonstrated significantly higher mortality rate than those with a low SII (11 [23.9%] vs. 5 [6.4%], respectively; p < 0.01). In addition, the high SII group had significantly higher rate of aortic-related reinterventions than the low SII group (16 [34.8%] vs. 15 [19.2%], p = 0.03). Multivariable Cox analyses showed that a high SII score was independently associated with composite outcomes rate (hazard ratio, 2.15; 95% confidence interval, 1.22-3.78; p < 0.01). CONCLUSIONS The long-term therapeutic effectiveness of OMT alone in patients with acute uTBAD is unsatisfactory. An SII > 1449 at the time of diagnosis is an independent predictor of OMT failure.
Collapse
Affiliation(s)
- Ruirong Chen
- Department of Anesthesiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Sheng Su
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Changjin Wang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yuan Liu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wenhui Huang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Songyuan Luo
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Fan Yang
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Jianfang Luo
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
15
|
Macarie RD, Tucureanu MM, Ciortan L, Gan AM, Butoi E, Mânduțeanu I. Ficolin-2 amplifies inflammation in macrophage-smooth muscle cell cross-talk and increases monocyte transmigration by mechanisms involving IL-1β and IL-6. Sci Rep 2023; 13:19431. [PMID: 37940674 PMCID: PMC10632380 DOI: 10.1038/s41598-023-46770-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 11/04/2023] [Indexed: 11/10/2023] Open
Abstract
Ficolin-2, recently identified in atherosclerotic plaques, has been correlated with future acute cardiovascular events, but its role remains unknown. We hypothesize that it could influence plaque vulnerability by interfering in the cross-talk between macrophages (MØ) and smooth muscle cells (SMC). To examine its role and mechanism of action, we exposed an in-vitro co-culture system of SMC and MØ to ficolin-2 (10 µg/mL) and then performed cytokine array, protease array, ELISA, qPCR, Western Blot, and monocyte transmigration assay. Carotid plaque samples from atherosclerotic patients with high plasma levels of ficolin-2 were analyzed by immunofluorescence. We show that ficolin-2: (i) promotes a pro-inflammatory phenotype in SMC following interaction with MØ by elevating the gene expression of MCP-1, upregulating gene and protein expression of IL-6 and TLR4, and by activating ERK/MAPK and NF-KB signaling pathways; (ii) increased IL-1β, IL-6, and MIP-1β in MØ beyond the level induced by cellular interaction with SMC; (iii) elevated the secretion of IL-1β, IL-6, and CCL4 in the conditioned medium; (iv) enhanced monocyte transmigration and (v) in atherosclerotic plaques from patients with high plasma levels of ficolin-2, we observed co-localization of ficolin-2 with SMC marker αSMA and the cytokines IL-1β and IL-6. These findings shed light on previously unknown mechanisms underlying ficolin-2-dependent pathological inflammation in atherosclerotic plaques.
Collapse
Affiliation(s)
- Răzvan Daniel Macarie
- Biopathology and Therapy of Inflammation Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Monica Mădălina Tucureanu
- Biopathology and Therapy of Inflammation Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania.
| | - Letiția Ciortan
- Biopathology and Therapy of Inflammation Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Ana-Maria Gan
- Biopathology and Therapy of Inflammation Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Elena Butoi
- Biopathology and Therapy of Inflammation Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Ileana Mânduțeanu
- Biopathology and Therapy of Inflammation Department, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| |
Collapse
|
16
|
Yan A, Gotlieb AI. The microenvironment of the atheroma expresses phenotypes of plaque instability. Cardiovasc Pathol 2023; 67:107572. [PMID: 37595697 DOI: 10.1016/j.carpath.2023.107572] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/06/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023] Open
Abstract
Data from histopathology studies of human atherosclerotic tissue specimens and from vascular imaging studies support the concept that the local arterial microenvironment of a stable atheroma promotes destabilizing conditions that result in the transition to an unstable atheroma. Destabilization is characterized by several different plaque phenotypes that cause major clinical events such as acute coronary syndrome and cerebrovascular strokes. There are several rupture-associated phenotypes causing thrombotic vascular occlusion including simple fibrous cap rupture of an atheroma, fibrous cap rupture at site of previous rupture-and-repair of an atheroma, and nodular calcification with rupture. Endothelial erosion without rupture has more recently been shown to be a common phenotype to promote thrombosis as well. Microenvironment features that are linked to these phenotypes of plaque instability are neovascularization arising from the vasa vasorum network leading to necrotic core expansion, intraplaque hemorrhage, and cap rupture; activation of adventitial and perivascular adipose tissue cells leading to secretion of cytokines, growth factors, adipokines in the outer artery wall that destabilize plaque structure; and vascular smooth muscle cell phenotypic switching through transdifferentiation and stem/progenitor cell activation resulting in the promotion of inflammation, calcification, and secretion of extracellular matrix, altering fibrous cap structure, and necrotic core growth. As the technology evolves, studies using noninvasive vascular imaging will be able to investigate the transition of stable to unstable atheromas in real time. A limitation in the field, however, is that reliable and predictable experimental models of spontaneous plaque rupture and/or erosion are not currently available to study the cell and molecular mechanisms that regulate the conversion of the stable atheroma to an unstable plaque.
Collapse
Affiliation(s)
- Angela Yan
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | - Avrum I Gotlieb
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
17
|
Li J, Xu J, Zhang W, Li P, Zhang W, Wang H, Tang B. Detection and Imaging of Active Substances in Early Atherosclerotic Lesions Using Fluorescent Probes. Chembiochem 2023; 24:e202300105. [PMID: 36898970 DOI: 10.1002/cbic.202300105] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/12/2023]
Abstract
Atherosclerosis (AS) is a vascular disease caused by chronic inflammation and lipids that is the main cause of myocardial infarction, stroke and other cardiovascular diseases. Atherosclerosis is often difficult to detect in its early stages due to the absence of clinically significant vascular stenosis. This is not conducive to early intervention or treatment of the disease. Over the past decade, researchers have developed various imaging methods for the detection and imaging of atherosclerosis. At the same time, more and more biomarkers are being found that can be used as targets for detecting atherosclerosis. Therefore, the development of a variety of imaging methods and a variety of targeted imaging probes is an important project to achieve early assessment and treatment of atherosclerosis. This paper provides a comprehensive review of the optical probes used to detect and target atherosclerosis imaging in recent years, and describes the current challenges and future development directions.
Collapse
Affiliation(s)
- Jin Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for, Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Jiheng Xu
- School of Materials Science and Engineering, Shandong University, Jinan, 250014, P. R. China
| | - Wei Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for, Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Ping Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for, Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Wen Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for, Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Hui Wang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for, Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for, Chemical Imaging in Universities of Shandong, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, P. R. China
| |
Collapse
|
18
|
Yazdani AN, Pletsch M, Chorbajian A, Zitser D, Rai V, Agrawal DK. Biomarkers to monitor the prognosis, disease severity, and treatment efficacy in coronary artery disease. Expert Rev Cardiovasc Ther 2023; 21:675-692. [PMID: 37772751 PMCID: PMC10615890 DOI: 10.1080/14779072.2023.2264779] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/26/2023] [Indexed: 09/30/2023]
Abstract
INTRODUCTION Coronary Artery Disease (CAD) is a prevalent condition characterized by the presence of atherosclerotic plaques in the coronary arteries of the heart. The global burden of CAD has increased significantly over the years, resulting in millions of deaths annually and making it the leading health-care expenditure and cause of mortality in developed countries. The lack of cost-effective strategies for monitoring the prognosis of CAD warrants a pressing need for accurate and efficient markers to assess disease severity and progression for both reducing health-care costs and improving patient outcomes. AREA COVERED To effectively monitor CAD, prognostic biomarkers and imaging techniques play a vital role in risk-stratified patients during acute treatment and over time. However, with over 1,000 potential markers of interest, it is crucial to identify the key markers with substantial utility in monitoring CAD progression and evaluating therapeutic interventions. This review focuses on identifying and highlighting the most relevant markers for monitoring CAD prognosis and disease severity. We searched for relevant literature using PubMed and Google Scholar. EXPERT OPINION By utilizing the markers discussed, health-care providers can improve patient care, optimize treatment plans, and ultimately reduce health-care costs associated with CAD management.
Collapse
Affiliation(s)
- Armand N. Yazdani
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766
| | - Michaela Pletsch
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766
| | - Abraham Chorbajian
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766
| | - David Zitser
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766
| | - Vikrant Rai
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766
| | - Devendra K. Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766
| |
Collapse
|
19
|
Wei Y, Li W, Luan H, Tuerhongjiang G, Yuan Z, Wu Y. The association of glycated hemoglobin A1c with coronary artery disease, myocardial infarction, and severity of coronary lesions. J Investig Med 2023; 71:202-211. [PMID: 36803042 DOI: 10.1177/10815589221140593] [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: 01/07/2023]
Abstract
Glycated hemoglobin A1c (HbA1c) has been recognized as a predictor of cardiovascular events. However, the relationship between HbA1c and coronary artery disease (CAD) in the Chinese population has yet to be systematically explored. In addition, factors associated with HbA1c were generally analyzed linearly, thereby failing to appreciate more complex nonlinear associations. The study aimed to evaluate the relationship between the HbA1c value and the presence and severity of coronary artery stenosis. A total of 7192 consecutive patients who underwent coronary angiography were enrolled. Their biological parameters, including HbA1c, were measured. The severity of coronary stenosis was evaluated using Gensini score. After correcting for baseline confounding factors, a multivariate logistic regression analysis was used to evaluate the relationship between HbA1c and CAD severity. Restricted cubic splines were applied to explore the relation of HbA1c with the presence of CAD, myocardial infarction (MI), and the severity of coronary lesions. HbA1c was significantly associated with the presence and severity of CAD in patients without diagnosed diabetes (odds ratio: 1.306, 95% confidence interval: 1.053-1.619, p = 0.015). Spline analysis showed a U-shaped association of HbA1c with the presence of MI. Both HbA1c > 7.2% and HbA1c < 5.7% were associated with the presence of MI. In conclusion, HbA1c value was highly associated with the severity of coronary artery stenosis in the whole study population, and in CAD patients without diagnosed diabetes. Compared with patients with HbA1c levels between 6.0% and 7.0%, HbA1c < 5.7% and HbA1c > 7.2% were associated with higher presence of MI.
Collapse
Affiliation(s)
- Yuanyuan Wei
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Wenyuan Li
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hui Luan
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shannxi, China
| | - Gulinigaer Tuerhongjiang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shannxi, China
| | - Zuyi Yuan
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shannxi, China
| | - Yue Wu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shannxi, China
| |
Collapse
|
20
|
Photon-Counting Computed Tomography (PCCT): Technical Background and Cardio-Vascular Applications. Diagnostics (Basel) 2023; 13:diagnostics13040645. [PMID: 36832139 PMCID: PMC9955798 DOI: 10.3390/diagnostics13040645] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/28/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Photon-counting computed tomography (PCCT) is a new advanced imaging technique that is going to transform the standard clinical use of computed tomography (CT) imaging. Photon-counting detectors resolve the number of photons and the incident X-ray energy spectrum into multiple energy bins. Compared with conventional CT technology, PCCT offers the advantages of improved spatial and contrast resolution, reduction of image noise and artifacts, reduced radiation exposure, and multi-energy/multi-parametric imaging based on the atomic properties of tissues, with the consequent possibility to use different contrast agents and improve quantitative imaging. This narrative review first briefly describes the technical principles and the benefits of photon-counting CT and then provides a synthetic outline of the current literature on its use for vascular imaging.
Collapse
|
21
|
McGarrah RW, Ferencik M, Giamberardino SN, Hoffmann U, Foldyna B, Karady J, Ginsburg GS, Kraus WE, Douglas PS, Shah SH. Lipoprotein Subclasses Associated With High-Risk Coronary Atherosclerotic Plaque: Insights From the PROMISE Clinical Trial. J Am Heart Assoc 2022; 12:e026662. [PMID: 36565187 PMCID: PMC9973611 DOI: 10.1161/jaha.122.026662] [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] [Indexed: 12/25/2022]
Abstract
BACKGROUND More than half of major adverse cardiovascular events (MACE) occur in the absence of obstructive coronary artery disease and are often attributed to the rupture of high-risk coronary atherosclerotic plaque (HRP). Blood-based biomarkers that associate with imaging-defined HRP and predict MACE are lacking. METHODS AND RESULTS Nuclear magnetic resonance-based lipoprotein particle profiling was performed in the biomarker substudy of the PROMISE (Prospective Multicenter Imaging Study for Evaluation of Chest Pain) trial (N=4019) in participants who had stable symptoms suspicious for coronary artery disease. Principal components analysis was used to reduce the number of correlated lipoproteins into uncorrelated lipoprotein factors. The association of lipoprotein factors and individual lipoproteins of significantly associated factors with core laboratory determined coronary computed tomographic angiography features of HRP was determined using logistic regression models. The association of HRP-associated lipoproteins with MACE was assessed in the PROMISE trial and validated in an independent coronary angiography biorepository (CATHGEN [Catheterization Genetics]) using Cox proportional hazards models. Lipoprotein factors composed of high-density lipoprotein (HDL) subclasses were associated with HRP. In these factors, large HDL (odds ratio [OR], 0.70 [95% CI, 0.56-0.85]; P<0.001) and medium HDL (OR, 0.84 [95% CI, 0.72-0.98]; P=0.028) and HDL size (OR, 0.82 [95% CI, 0.69-0.96]; P=0.018) were associated with HRP in multivariable models. Medium HDL was associated with MACE in PROMISE (hazard ratio [HR], 0.76 [95% CI, 0.63-0.92]; P=0.004), which was validated in the CATHGEN biorepository (HR, 0.91 [95% CI, 0.88-0.94]; P<0.001). CONCLUSIONS Large and medium HDL subclasses and HDL size inversely associate with HRP features, and medium HDL subclasses inversely associate with MACE in PROMISE trial participants. These findings may aid in the risk stratification of individuals with chest pain and provide insight into the pathobiology of HRP. REGISTRATION URL: https://clinicaltrials.gov; Unique identifier: NCT01174550.
Collapse
Affiliation(s)
- Robert W. McGarrah
- Division of Cardiology, Department of MedicineDuke University School of MedicineDurhamNC,Duke Molecular Physiology InstituteDuke University School of MedicineDurhamNC
| | - Maros Ferencik
- Knight Cardiovascular InstituteOregon Health and Science UniversityPortlandOR
| | | | - Udo Hoffmann
- Cardiovascular Imaging Research CenterHarvard Medical School–Massachusetts General HospitalBostonMA
| | - Borek Foldyna
- Cardiovascular Imaging Research CenterHarvard Medical School–Massachusetts General HospitalBostonMA
| | - Julia Karady
- Cardiovascular Imaging Research CenterHarvard Medical School–Massachusetts General HospitalBostonMA,MTA‐SE Cardiovascular Imaging Research Group, Heart and Vascular CenterSemmelweis UniversityBudapestHungary
| | - Geoffrey S. Ginsburg
- Duke Center for Applied Genomics & Precision MedicineDuke University School of MedicineDurhamNC
| | - William E. Kraus
- Division of Cardiology, Department of MedicineDuke University School of MedicineDurhamNC,Duke Molecular Physiology InstituteDuke University School of MedicineDurhamNC
| | - Pamela S. Douglas
- Division of Cardiology, Department of MedicineDuke University School of MedicineDurhamNC,Duke Clinical Research InstituteDuke University School of MedicineDurhamNC
| | - Svati H. Shah
- Division of Cardiology, Department of MedicineDuke University School of MedicineDurhamNC,Duke Molecular Physiology InstituteDuke University School of MedicineDurhamNC,Duke Clinical Research InstituteDuke University School of MedicineDurhamNC
| |
Collapse
|
22
|
Bordeianu G, Mitu I, Stanescu RS, Ciobanu CP, Petrescu-Danila E, Marculescu AD, Dimitriu DC. Circulating Biomarkers for Laboratory Diagnostics of Atherosclerosis-Literature Review. Diagnostics (Basel) 2022; 12:diagnostics12123141. [PMID: 36553147 PMCID: PMC9777004 DOI: 10.3390/diagnostics12123141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis is still considered a disease burden with long-term damaging processes towards the cardiovascular system. Evaluation of atherosclerotic stages requires the use of independent markers such as those already considered traditional, that remain the main therapeutic target for patients with atherosclerosis, together with emerging biomarkers. The challenge is finding models of predictive markers that are particularly tailored to detect and evaluate the evolution of incipient vascular lesions. Important advances have been made in this field, resulting in a more comprehensible and stronger linkage between the lipidic profile and the continuous inflammatory process. In this paper, we analysed the most recent data from the literature studying the molecular mechanisms of biomarkers and their involvement in the cascade of events that occur in the pathophysiology of atherosclerosis.
Collapse
Affiliation(s)
| | - Ivona Mitu
- Correspondence: (I.M.); (R.S.S.); Tel.: +40-75206-1747 (I.M.)
| | | | | | | | | | | |
Collapse
|
23
|
Perrotta I. Atherosclerosis: From Molecular Biology to Therapeutic Perspective 2.0. Int J Mol Sci 2022; 23:ijms232315158. [PMID: 36499481 PMCID: PMC9740737 DOI: 10.3390/ijms232315158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease of large- and medium-sized arteries involving aberrant immune-inflammatory responses, dysfunctional molecular pathways, and impaired tissue repair mechanisms [...].
Collapse
Affiliation(s)
- Ida Perrotta
- Department of Biology, Ecology and Earth Sciences, Centre for Microscopy and Microanalysis (CM2), University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy
| |
Collapse
|
24
|
Sphingosine 1-Phosphate and Apolipoprotein M Levels and Their Correlations with Inflammatory Biomarkers in Patients with Untreated Familial Hypercholesterolemia. Int J Mol Sci 2022; 23:ijms232214065. [PMID: 36430543 PMCID: PMC9697457 DOI: 10.3390/ijms232214065] [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: 10/18/2022] [Revised: 11/08/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
High-density lipoprotein (HDL)-bound apolipoprotein M/sphingosine 1-phosphate (ApoM/S1P) complex in cardiovascular diseases serves as a bridge between HDL and endothelial cells, maintaining a healthy endothelial barrier. To date, S1P and ApoM in patients with untreated heterozygous familial hypercholesterolemia (HeFH) have not been extensively studied. Eighty-one untreated patients with HeFH and 32 healthy control subjects were included in this study. Serum S1P, ApoM, sCD40L, sICAM-1, sVCAM-1, oxLDL, and TNFα concentrations were determined by ELISA. PON1 activities were measured spectrophotometrically. Lipoprotein subfractions were detected by Lipoprint. We diagnosed FH using the Dutch Lipid Clinic Network criteria. Significantly higher serum S1P and ApoM levels were found in HeFH patients compared to controls. S1P negatively correlated with large HDL and positively with small HDL subfractions in HeFH patients and the whole study population. S1P showed significant positive correlations with sCD40L and MMP-9 levels and PON1 arylesterase activity, while we found significant negative correlation between sVCAM-1 and S1P in HeFH patients. A backward stepwise multiple regression analysis showed that the best predictors of serum S1P were large HDL subfraction and arylesterase activity. Higher S1P and ApoM levels and their correlations with HDL subfractions and inflammatory markers in HeFH patients implied their possible role in endothelial protection.
Collapse
|
25
|
Li T, Yuan D, Wang P, Jia S, Zhang C, Zhu P, Song Y, Tang X, Zhao X, Gao Z, Yang Y, Gao R, Xu B, Yuan J. Associations of lipid measures with total occlusion in patients with established coronary artery disease: a cross-sectional study. Lipids Health Dis 2022; 21:118. [DOI: 10.1186/s12944-022-01733-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/04/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
Background
Total occlusion is the most severe coronary lesion, indicating heavy ischemic burden and poor prognosis. The lipid profile is central to the development of atherosclerotic coronary lesions. Evidence on the optimal lipid measure to be monitored and managed in patients with established coronary artery disease (CAD) is inconclusive.
Methods
Total cholesterol (TC), total triglyceride (TG), low-density lipoprotein cholesterol (LDL-c), nonhigh-density lipoprotein cholesterol (non-HDL-c), lipoprotein (a) [Lp(a)], apolipoprotein B (apoB), non-HDL-c/HDL-c, and apoB/apoA-1 were analyzed in quintiles and as continuous variables. The associations of lipid measures with total occlusion were tested using logistic regression models, visualized with restricted cubic splines, and compared by areas under the receiver operating characteristic curves (AUROC). Discordance analysis was performed when apoB/apoA-1 and non-HDL-c/HDL-c were not in concordance.
Results
The prospective cohort study included 10,003 patients (mean age: 58 years; women: 22.96%), with 1879 patients having total occlusion. The risks of total occlusion significantly increased with quintiles of Lp(a), non-HDL-c/HDL-c, and apoB/apoA-1 (all p for trend < 0.001). TG had no association with total occlusion. Restricted cubic splines indicate significant positive linear relations between the two ratios and total occlusion [odds ratio per 1-standard deviation increase (95% confidence interval): non-HDL-c/HDL-c: 1.135 (1.095–1.176), p < 0.001; apoB/apoA-1: 2.590 (2.049–3.274), p < 0.001]. The AUROCs of apoB/apoA-1 and non-HDL-c/HDL-c were significantly greater than those of single lipid measures. Elevation in the apoB/apoA-1 tertile significantly increased the risk of total occlusion at a given non-HDL-c/HDL-c tertile but not vice versa.
Conclusion
ApoB/apoA-1 confers better predictive power for total occlusion than non-HDL-c/HDL-c and single lipid measures in established CAD patients.
Collapse
|
26
|
Hagiwara Y, Takao N, Usuki N, Yoshie T, Takaishi S, Shimizu T, Ueda T, Hasegawa Y, Yamano Y. Carotid ultrasound using superb microvascular imaging to identify patients developing in-stent restenosis after CAS. J Stroke Cerebrovasc Dis 2022; 31:106627. [DOI: 10.1016/j.jstrokecerebrovasdis.2022.106627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 06/23/2022] [Accepted: 06/26/2022] [Indexed: 10/17/2022] Open
|
27
|
Gargani L, Baldini M, Berchiolli R, Bort IR, Casolo G, Chiappino D, Cosottini M, D'Angelo G, De Santis M, Erba P, Fabiani I, Fabiani P, Gabbriellini I, Galeotti GG, Ghicopulos I, Goncalves I, Lapi S, Masini G, Morizzo C, Napoli V, Nilsson J, Orlandi G, Palombo C, Pieraccini F, Ricci S, Siciliano G, Slart RHJA, De Caterina R. Detecting the vulnerable carotid plaque: the Carotid Artery Multimodality imaging Prognostic study design. J Cardiovasc Med (Hagerstown) 2022; 23:466-473. [PMID: 35763768 DOI: 10.2459/jcm.0000000000001314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Carotid artery disease is highly prevalent and a main cause of ischemic stroke and vascular dementia. There is a paucity of information on predictors of serious vascular events. Besides percentage diameter stenosis, international guidelines also recommend the evaluation of qualitative characteristics of carotid artery disease as a guide to treatment, but with no agreement on which qualitative features to assess. This inadequate knowledge leads to a poor ability to identify patients at risk, dispersion of medical resources, and unproven use of expensive and resource-consuming techniques, such as magnetic resonance imaging, positron emission tomography, and computed tomography. OBJECTIVES The Carotid Artery Multimodality imaging Prognostic (CAMP) study will: prospectively determine the best predictors of silent and overt ischemic stroke and vascular dementia in patients with asymptomatic subcritical carotid artery disease by identifying the noninvasive diagnostic features of the 'vulnerable carotid plaque'; assess whether 'smart' use of low-cost diagnostic methods such as ultrasound-based evaluations may yield at least the same level of prospective information as more expensive techniques. STUDY DESIGN We will compare the prognostic/predictive value of all proposed techniques with regard to silent or clinically manifest ischemic stroke and vascular dementia. The study will include ≥300 patients with asymptomatic, unilateral, intermediate degree (40-60% diameter) common or internal carotid artery stenosis detected at carotid ultrasound, with a 2-year follow-up. The study design has been registered on Clinicaltrial.gov on December 17, 2020 (ID number NCT04679727).
Collapse
Affiliation(s)
- Luna Gargani
- Institute of Clinical Physiology, National Research Council
| | | | - Raffaella Berchiolli
- Vascular Surgery Unit, Cardio Thoracic and Vascular Department, University of Pisa
| | | | | | | | | | | | - Mariella De Santis
- Cardiology Unit, Cardio-Thoracic and Vascular Department, University of Pisa, Pisa, Italy
| | - Paola Erba
- Department of Nuclear Medicine, University of Pisa, Pisa, Italy
- Medical Imaging Center, Department of Nuclear Medicine & Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Plinio Fabiani
- Internal Medicine, S.M. Annunziata Hospital, Florence, Italy
| | - Ilaria Gabbriellini
- Neurology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gian Giacomo Galeotti
- Cardiology Unit, Cardio-Thoracic and Vascular Department, University of Pisa, Pisa, Italy
| | - Irene Ghicopulos
- Neurology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Isabel Goncalves
- Department of Clinical Sciences - Malmö University Hospital, University of Lund, Malmö, Sweden
| | - Simone Lapi
- BMS Multispecialistic Biobank-Biobank Unit, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Gabriele Masini
- Cardiology Unit, Cardio-Thoracic and Vascular Department, University of Pisa, Pisa, Italy
| | - Carmela Morizzo
- Cardiology Unit, Cardio-Thoracic and Vascular Department, University of Pisa, Pisa, Italy
| | - Vinicio Napoli
- Diagnostic and Interventional Radiology, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Jan Nilsson
- Department of Clinical Sciences - Malmö University Hospital, University of Lund, Malmö, Sweden
| | - Giovanni Orlandi
- Neurology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Carlo Palombo
- Cardiology Unit, Cardio-Thoracic and Vascular Department, University of Pisa, Pisa, Italy
| | | | - Stefano Ricci
- Department of Information Engineering (DINFO), University of Florence, Florence, Italy
| | - Gabriele Siciliano
- Neurology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Riemer H J A Slart
- Medical Imaging Center, Department of Nuclear Medicine & Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Biomedical Photonic Imaging, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Raffaele De Caterina
- Cardiology Unit, Cardio-Thoracic and Vascular Department, University of Pisa, Pisa, Italy
| |
Collapse
|
28
|
Demir M, Özbek M. A novel predictor in patients with coronary chronic total occlusion: systemic immune-inflammation index: a single-center cross-sectional study. Rev Assoc Med Bras (1992) 2022; 68:579-585. [DOI: 10.1590/1806-9282.20211097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/11/2022] [Indexed: 11/22/2022] Open
|
29
|
Su S, Liu J, Chen L, Xie E, Geng Q, Zeng H, Yuan Q, Yang F, Luo J. Systemic immune-inflammation index predicted the clinical outcome in patients with type-B aortic dissection undergoing thoracic endovascular repair. Eur J Clin Invest 2022; 52:e13692. [PMID: 34695253 DOI: 10.1111/eci.13692] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/11/2021] [Accepted: 10/04/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND The systemic immune-inflammation index (SII) has been reported to have prognostic ability in various cardiovascular diseases; however, it has not been studied in type-B aortic dissection (TBAD). We aimed to explore the relation of SII with short-term and long-term outcomes in TBAD patients undergoing thoracic endovascular repair (TEVAR). METHODS We performed a retrospective analysis of a prospectively maintained database from 2010 to 2017. The patients were divided into two groups (high SII and low SII) as per the optimal cut-off value determined using the receiver operating characteristic curve. Multivariate logistic and Cox regression analyses were performed to analyse the relationship between the SII and the short-term and long-term outcomes. RESULTS A total of 805 TBAD patients who underwent TEVAR were enrolled. Twenty-six (3.2%) patients died during hospitalisation. At the end of a median follow-up duration of 48.80 mon, 70 (9.8%) patients had died. The patients were divided into the high-SII group [n = 333 (41.4%%)] and the low-SII group [n = 472 (58.6%)] as per the optimal cut-off value of 1,062. Multivariable logistic analyses showed that a high-SII score was independently associated with major adverse cardiovascular events (MACEs) in-hospital (odd ratio [OR], 1.67; 95% confidence interval [CI], 1.13-2.47; p = .01). In addition, multivariable Cox analyses showed that a high-SII score could be an independent indicator for follow-up adverse events (hazard ratio [HR], 1.70; 95% CI, 1.14-2.56, p = .01). CONCLUSIONS Systemic immune-inflammation index is associated with both in-hospital and long-term outcomes in patients with TBAD undergoing TEVAR. Therefore, SII may serve as valuable tool for risk stratification before intervention.
Collapse
Affiliation(s)
- Sheng Su
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jitao Liu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Lyufan Chen
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Enmin Xie
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Qingshan Geng
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hongke Zeng
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qiong Yuan
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Institute of Pharmaceutical Innovation, College of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Fan Yang
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Jianfang Luo
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
30
|
Systemic immune-inflammation index predicts in-hospital and long-term outcomes in patients with ST-segment elevation myocardial infarction. Coron Artery Dis 2022; 33:251-260. [PMID: 35044330 DOI: 10.1097/mca.0000000000001117] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE This study examines the predictive value of the novel systemic immune-inflammation index (SII) in patients with ST-segment elevation myocardial infarction (STEMI). METHODS A total of 1660 patients with STEMI who underwent primary percutaneous coronary intervention (pPCI) were enrolled in the study. In-hospital and 3-year outcomes were compared between the four groups (Q1-4). The SII was calculated using the following formula: neutrophil*platelet/lymphocyte. RESULTS The frequency of in-hospital cardiogenic shock, acute respiratory failure, acute kidney injury, ventricular arrhythmia, stent thrombosis, recurrent myocardial infarction, major adverse cardiac events and mortality were significantly higher in the high SII groups (Q3 and Q4). Logistic regression models demonstrated that Q3 and Q4 had an independent risk of mortality and Q4 had an independent risk of cardiogenic shock compared to Q1. Receiver operating characteristic analysis showed that the best cutoff value of SII to predict the in-hospital mortality was 1781 with 66% sensitivity and 74% specificity. Kaplan-Meier overall survivals for Q1, Q2, Q3 and Q4 were 97.6, 96.9, 91.6 and 81.0%, respectively. Cox proportional analysis for 3-year mortality demonstrated that Q3 and Q4 had an independent risk for mortality compared to Q1. CONCLUSION SII, a novel inflammatory index, was found to be a better predictor for in-hospital and long-term outcomes than traditional risk factors in patients with STEMI undergoing pPCI.
Collapse
|
31
|
Udaya R, Sivakanesan R. Synopsis of Biomarkers of Atheromatous Plaque Formation, Rupture and Thrombosis in the Diagnosis of Acute Coronary Syndromes. Curr Cardiol Rev 2022; 18:53-62. [PMID: 35410616 PMCID: PMC9896418 DOI: 10.2174/1573403x18666220411113450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/08/2021] [Accepted: 01/15/2022] [Indexed: 11/22/2022] Open
Abstract
Acute coronary syndrome is the main cause of mortality and morbidity worldwide and early diagnosis is a challenge for clinicians. Though cardiac Troponin, the most commonly used biomarker, is the gold standard for myocardial necrosis, it is blind for ischemia without necrosis. Therefore, ideal biomarkers are essential in the care of patients presenting with symptoms suggestive of cardiac ischemia. The ideal biomarker or group of biomarkers of atheromatous plaque formation, rupture and thrombosis for timely and accurate diagnosis of acute coronary syndrome is a current need. Therefore, we discuss the existing understanding and future of biomarkers of atheromatous plaque formation, rupture and thrombosis of acute coronary syndrome in this review. Keywords were searched from Medline, ISI, IBSS and Google Scholar databases. Further, the authors conducted a manual search of other relevant journals and reference lists of primary articles. The development of high-sensitivity troponin assays facilitates earlier exclusion of acute coronary syndrome, contributing to a reduced length of stay at the emergency department, and earlier treatment resulting in better outcomes. Although researchers have investigated biomarkers of atheromatous plaque formation, rupture and thrombosis to help early diagnosis of cardiac ischemia, most of them necessitate validation from further analysis. Among these biomarkers, pregnancy-associated plasma protein-A, intercellular adhesion molecule-1, and endothelial cell-specific molecule- 1(endocan) have shown promising results in the early diagnosis of acute coronary syndrome but need further evaluation. However, the use of a combination of biomarkers representing varying pathophysiological mechanisms of cardiac ischemia will support risk assessment, diagnosis and prognosis in these patients and this is the way forward.
Collapse
Affiliation(s)
- Ralapanawa Udaya
- Address correspondence to this author at the Department of Medicine, University of Peradeniya, Galaha Rd, 20400, Sri Lanka; Tel: 0718495682; E-mail:
| | | |
Collapse
|
32
|
NR1D1 Deletion Induces Rupture-Prone Vulnerable Plaques by Regulating Macrophage Pyroptosis via the NF- κB/NLRP3 Inflammasome Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5217572. [PMID: 34956438 PMCID: PMC8702349 DOI: 10.1155/2021/5217572] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 11/11/2021] [Indexed: 12/18/2022]
Abstract
Vulnerable plaque rupture is the main trigger of most acute cardiovascular events. But the underlying mechanisms responsible for the transition from stable to vulnerable plaque remain largely unknown. Nuclear receptor subfamily 1 group D member 1 (NR1D1), also known as REV-ERB α, is a nuclear receptor that has shown the protective role in cardiovascular system. However, the effect of NR1D1 on vulnerable plaque rupture and its underlying mechanisms are still unclear. By generating the rupture-prone vulnerable plaque model in hypercholesterolemic ApoE−/− mice and NR1D1−/−ApoE−/− mice, we demonstrated that NR1D1 deficiency significantly augmented plaque vulnerability/rupture, with higher incidence of intraplaque hemorrhage (78.26% vs. 47.82%, P = 0.0325) and spontaneous plaque rupture with intraluminal thrombus formation (65.21% vs. 39.13%, P = 0.1392). In vivo experiments indicated that NR1D1 exerted a protective role in the vasculature. Mechanically, NR1D1 deficiency aggravates macrophage infiltration, inflammation, and oxidative stress. Compared with the ApoE−/− mice, NR1D1−/−ApoE−/− mice exhibited a significantly higher expression level of pyroptosis-related genes in macrophages within the plaque. Further investigation based on mice bone marrow-derived macrophages (BMDMs) confirmed that NR1D1 exerted a protective effect by inhibiting macrophage pyroptosis in a NLRP3-inflammasome-dependent manner. Besides, pharmacological activation of NR1D1 by SR9009, a specific NR1D1 agonist, prevented plaque vulnerability/rupture. In general, our findings provide further evidences that NR1D1 plays a protective role in the vasculature, regulates inflammation and oxidative stress, and stabilizes rupture-prone vulnerable plaques.
Collapse
|
33
|
Montanaro M, Scimeca M, Toschi N, Bonanno E, Giacobbi E, Servadei F, Ippoliti A, Santeusanio G, Mauriello A, Anemona L. Effects of Risk Factors on In Situ Expression of Proinflammatory Markers Correlated to Carotid Plaque Instability. Appl Immunohistochem Mol Morphol 2021; 29:741-749. [PMID: 34039839 DOI: 10.1097/pai.0000000000000947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 04/13/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND AND AIMS Several studies demonstrated a role of active chronic inflammatory infiltrate in carotid plaques progression suggesting a possible link between cardiovascular risk factors and inflammation-related plaque instability. The aim of this study is therefore to evaluate the possible effects of cardiovascular risk factors on in situ expression of proinflammatory markers associated with carotid plaque instability. METHODS AND RESULTS A tissue microarray containing carotid plaques from 36 symptomatic (major stroke or transient ischemic attack) and 37 asymptomatic patients was built. Serial sections were employed to evaluate the expression of some inflammatory markers by immunohistochemistry [CD3, CD4a, CD8, CD20, CD86, CD163, interleukin (IL)-2, IL-6, IL-17]. Immunohistochemical data were analyzed to study the possible associations between in situ expression of inflammatory biomarker and the main cardiovascular risk factors. Our data demonstrated that plaque instability is associated with the high in situ expression of some cytokines, such as IL-2, IL-6, IL-17. Besides the female sex, none of the risk factors analyzed showed a significant association between the in situ expression of these markers and unstable plaques. A significant increase of IL-6-positive and IL-17-positive cells was observed in unstable atheromatous plaques of female patients, as compared with unstable plaques of male patients. CONCLUSIONS Plaque destabilization is certainly correlated with the presence of the major cardiovascular risk factors, however, our results showed that, with the exception of sex, their action in the evolutive process of plaque instability seems rather nonspecific, favoring a general release of proinflammatory cytokines.
Collapse
Affiliation(s)
| | - Manuel Scimeca
- Departments of Experimental Medicine
- University of San Raffaele
- Saint Camillus International University of Health Sciences, Rome, Italy
| | - Nicola Toschi
- Biomedicine and Prevention, University of Rome "Tor Vergata"
- Imaging Martinos Center for Biomedical Imaging
- Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Lee M, Lee K, Kim DW, Cho JS, Kim TS, Kwon J, Kim CJ, Park CS, Kim HY, Yoo KD, Jeon DS, Chang K, Kim MC, Jeong MH, Ahn Y, Park MW. Relationship of serial hsCRP changes to long-term clinical outcomes in stabilized post myocardial infarction patients. Can J Cardiol 2021; 38:92-101. [PMID: 34737035 DOI: 10.1016/j.cjca.2021.10.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/11/2021] [Accepted: 10/17/2021] [Indexed: 11/02/2022] Open
Abstract
BACKGROUND Little is known about the association between serial hsCRP (high-sensitivity C-reactive protein) measurements and long-term outcomes in post-myocardial infarction(MI) patients. We aimed to investigate the usefulness of serial hsCRP measurements for risk stratification in stabilized post-MI patients following percutaneous coronary intervention (PCI). METHODS A total of 1,018 patients who had hsCRP values at both baseline and 1 year following MI were included. High inflammatory status was defined as an hsCRP>2 mg/L. Patients were classified into four groups: persistently low, falling (first high then low hsCRP), rising (first low then high hsCRP), or persistently high hsCRP. The primary outcome was major adverse cardiac and cerebrovascular events (MACCE: a composite of all-cause of death, MI, and cerebrovascular accident) within 4 years after the second hsCRP measurement. RESULTS At 1 year after MI, the number of patients in the persistently low, falling, rising, and persistently high hsCRP groups was 394(38.7%),358(35.2%),69(6.8%), and 197(19.4%), respectively. The incidence of MACCE was progressively elevated from the persistently low to the falling, rising, and persistently high hsCRP groups.(respectively,4.8%,8.1%,10.1%, and 13.2%; p=0.004). Persistently high hsCRP was an independent predictor of MACCE (adjusted hazard ratio:2.55; 95% confidence interval:1.35 to 4.81; p=0.004) and provided incremental prognostic value beyond that of the baseline clinical risk model (NRI=0.397, IDI=0.025, all p < 0.001). CONCLUSIONS Among stabilized post-MI patients who underwent PCI, persistently high hsCRP was frequently seen 1 year after MI and was strongly associated with long-term adverse clinical outcomes. Serial measurements of hsCRP during clinical follow-up after MI may help to identify patients at higher risk for mortality and morbidity.
Collapse
Affiliation(s)
- Myunhee Lee
- Division of Cardiology, Daejeon St. Mary's hospital, College of Medicine, The Catholic University of Korea, Daejeon, Republic of Korea
| | - Kyusup Lee
- Division of Cardiology, Daejeon St. Mary's hospital, College of Medicine, The Catholic University of Korea, Daejeon, Republic of Korea
| | - Dae-Won Kim
- Division of Cardiology, Daejeon St. Mary's hospital, College of Medicine, The Catholic University of Korea, Daejeon, Republic of Korea
| | - Jung Sun Cho
- Division of Cardiology, Daejeon St. Mary's hospital, College of Medicine, The Catholic University of Korea, Daejeon, Republic of Korea
| | - Tae-Seok Kim
- Division of Cardiology, Daejeon St. Mary's hospital, College of Medicine, The Catholic University of Korea, Daejeon, Republic of Korea
| | - Jongbum Kwon
- Department of Thoracic and Cardiovascular Surgery, Daejeon St. Mary's Hospital, the Catholic University of Korea, Daejeon, Republic of Korea
| | - Chan Joon Kim
- Division of Cardiology, Department of Internal Medicine, Uijeongbu St Mary's Hospital, The Catholic University of Korea,Uijeongbu, Republic of Korea
| | - Chul Soo Park
- Division of Cardiology, Department of Internal Medicine, Yeouido St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hee Yeol Kim
- Division of Cardiology, Department of Internal Medicine, Bucheon St. Mary's Hospital, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Ki-Dong Yoo
- Division of Cardiology, Department of Internal Medicine, St. Vincent's hospital, The Catholic University of Korea, Suwon, Republic of Korea
| | - Doo Soo Jeon
- Division of Cardiology, Department of Internal Medicine, Incheon St. Mary's Hospital, The Catholic University of Korea, Incheon, Republic of Korea
| | - Kiyuk Chang
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Min Chul Kim
- Cardiovascular Center, Chonnam National University Hospital, Chonnam National University, Gwangju, Republic of Korea
| | - Myung Ho Jeong
- Cardiovascular Center, Chonnam National University Hospital, Chonnam National University, Gwangju, Republic of Korea
| | - Youngkeun Ahn
- Cardiovascular Center, Chonnam National University Hospital, Chonnam National University, Gwangju, Republic of Korea
| | - Mahn-Won Park
- Division of Cardiology, Daejeon St. Mary's hospital, College of Medicine, The Catholic University of Korea, Daejeon, Republic of Korea.
| |
Collapse
|
35
|
Hoel H, Pettersen EM, Høiseth LØ, Mathiesen I, Seternes A, Seljeflot I, Hisdal J. Effects of intermittent negative pressure treatment on circulating vascular biomarkers in patients with intermittent claudication. Vasc Med 2021; 26:489-496. [PMID: 33985385 PMCID: PMC8493410 DOI: 10.1177/1358863x211007933] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The aim of this study was to investigate the effects of lower extremity intermittent negative pressure (INP) treatment for 1 hour twice daily for 12 weeks, on circulating vascular biomarkers in patients with intermittent claudication. Patients were randomized to treatment with -40 mmHg INP (treatment group), or -10 mmHg INP (sham control group). Venous blood samples were collected at baseline and after 12 weeks, and concentrations of vascular adhesion molecule-1 (VCAM-1), intracellular adhesion molecule-1 (ICAM-1), E-selectin, P-selectin, von Willebrand factor (vWF), l-arginine, asymmetric dimethylarginine (ADMA), and symmetric dimethylarginine (SDMA) were analyzed. A larger proportion of the patients in the treatment group (25/31) had a reduction in vWF levels after 12 weeks, compared to the sham control group (17/30) (p = 0.043). Within the treatment group there was a significant mean (SEM) reduction in the concentration of vWF of -11% (4) (p = 0.019), whereas there was no significant change in the levels of vWF in the sham control group (1% (6); p = 0.85). There were no significant differences in the change of any of the biomarker levels between the groups after 12 weeks of treatment. In conclusion, there were no differences in the change of the circulating levels of the measured biomarkers between the treatment group and the sham control group after 12 weeks of INP treatment. However, the observed changes in vWF might indicate a beneficial effect of INP treatment on endothelial activation and endothelial injury. Clinicaltrials.gov Identifier: NCT03640676.
Collapse
Affiliation(s)
- Henrik Hoel
- Institute of Clinical Medicine, Faculty
of Medicine, University of Oslo, Oslo, Norway
- Department of Vascular Surgery, Oslo
University Hospital, Oslo, Norway
- Otivio AS, Oslo, Norway
| | - Erik Mulder Pettersen
- Department of Surgery, Sørlandet
Hospital, Kristiansand, Norway
- Department of Circulation and Medical
Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science
and Technology, Trondheim, Norway
| | | | | | - Arne Seternes
- Department of Circulation and Medical
Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science
and Technology, Trondheim, Norway
- Department of Surgery, Section for
Vascular Surgery, St Olavs Hospital, Trondheim, Norway
| | - Ingebjørg Seljeflot
- Institute of Clinical Medicine, Faculty
of Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Center for
Clinical Heart Research, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Jonny Hisdal
- Institute of Clinical Medicine, Faculty
of Medicine, University of Oslo, Oslo, Norway
- Department of Vascular Surgery, Oslo
University Hospital, Oslo, Norway
| |
Collapse
|
36
|
Kamtchum-Tatuene J, Nomani AZ, Falcione S, Munsterman D, Sykes G, Joy T, Spronk E, Vargas MI, Jickling GC. Non-stenotic Carotid Plaques in Embolic Stroke of Unknown Source. Front Neurol 2021; 12:719329. [PMID: 34630291 PMCID: PMC8492999 DOI: 10.3389/fneur.2021.719329] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/30/2021] [Indexed: 01/01/2023] Open
Abstract
Embolic stroke of unknown source (ESUS) represents one in five ischemic strokes. Ipsilateral non-stenotic carotid plaques are identified in 40% of all ESUS. In this narrative review, we summarize the evidence supporting the potential causal relationship between ESUS and non-stenotic carotid plaques; discuss the remaining challenges in establishing the causal link between non-stenotic plaques and ESUS and describe biomarkers of potential interest for future research. In support of the causal relationship between ESUS and non-stenotic carotid plaques, studies have shown that plaques with high-risk features are five times more prevalent in the ipsilateral vs. the contralateral carotid and there is a lower incidence of atrial fibrillation during follow-up in patients with ipsilateral non-stenotic carotid plaques. However, non-stenotic carotid plaques with or without high-risk features often coexist with other potential etiologies of stroke, notably atrial fibrillation (8.5%), intracranial atherosclerosis (8.4%), patent foramen ovale (5-9%), and atrial cardiopathy (2.4%). Such puzzling clinical associations make it challenging to confirm the causal link between non-stenotic plaques and ESUS. There are several ongoing studies exploring whether select protein and RNA biomarkers of plaque progression or vulnerability could facilitate the reclassification of some ESUS as large vessel strokes or help to optimize secondary prevention strategies.
Collapse
Affiliation(s)
- Joseph Kamtchum-Tatuene
- Faculty of Medicine and Dentistry, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Ali Z. Nomani
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Sarina Falcione
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Danielle Munsterman
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Gina Sykes
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Twinkle Joy
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Elena Spronk
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Maria Isabel Vargas
- Division of Neuroradiology, Department of Radiology and Medical Imaging, Geneva University Hospital, Geneva, Switzerland
| | - Glen C. Jickling
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
37
|
Ma HY, Mao Q, Zhu YB, Cong CL, Zheng SY, Zhang Q, Chen CC, Li LQ. Time-resolved Fluorescence Immunoassay (TRFIA) for the Simultaneous Detection of MMP-9 and Lp-PLA2 in Serum. J Fluoresc 2021; 31:1771-1777. [PMID: 34495467 DOI: 10.1007/s10895-021-02811-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 08/27/2021] [Indexed: 11/28/2022]
Abstract
Currently, atherosclerosis accounts for the majority of cardiovascular morbidity and mortality worldwide, and predicting the stability of atherosclerotic plaque is the main method to prevent atherosclerotic death. This study aims to establish a dual-label time-resolved fluorescence immunoassay (TRFIA) of matrix metalloprotein-9 (MMP-9) and lipoprotein-associated phospholipaseA2 (Lp-PLA2) to predict atherosclerotic plaque stability. A dual-label TRFIA was introduced for the simultaneous quantification of MMP-9 and Lp-PLA2 using fluorescent lanthanide (Eu3+ and Sm3+) chelates. The performance (sensitivity, specificity, accuracy, precision and reference intervals in different subjects) of this TRFIA was evaluated and compared with commercial kit. The sensitivity of the TRFIA for MMP-9 was 0.85 ng/mL and for Lp-PLA2 was 0.68 ng/mL with high affinity and specificity. The average recoveries were 94.58% to 109.82%, and 104.32% to 109.26%, respectively. All intra- and inter-assay CVs ranged from 3.10% to 5.46%. For the normal subjects, the cutoff value was 160.70 ng/mL for MMP-9 and 183.73 ng/mL for LP-PLA2; for the subjects with stable plaque, the cutoff value was 181.98~309.22 ng/mL for MMP-9 and 194.73~337.89 ng/mL for LP-PLA2; for the subjects with unstable plaque, the cutoff value was 330.43 ng/mL for MMP-9 and 343.23 ng/mL for LP-PLA2. This TRFIA detection results agreed well with the results of commercial kit (R2=0.9567 and R2=0.9771, respectively) in clinical serum samples. The TRFIA developed has a wide detection range and good sensitivity for the high-throughput simultaneous detection of MMP-9 and Lp-PLA2 in serum, which provides a new method for predicting the stability of atherosclerotic plaque.
Collapse
Affiliation(s)
- Hong-Yan Ma
- Department of Cardiology, Beihua University Affiliated Hospital, Jilin, 132000, China
| | - Qian Mao
- Department of Cardiology, Beihua University Affiliated Hospital, Jilin, 132000, China
| | - Yan-Bin Zhu
- Department of Cardiology, Beihua University Affiliated Hospital, Jilin, 132000, China
| | - Chun-Li Cong
- Department of Cardiology, Beihua University Affiliated Hospital, Jilin, 132000, China
| | - Shi-Yu Zheng
- Department of Cardiology, Beihua University Affiliated Hospital, Jilin, 132000, China
| | - Qi Zhang
- Department of Cardiology, Beihua University Affiliated Hospital, Jilin, 132000, China
| | - Cui-Cui Chen
- Guangzhou Youdi Biotechnology Co., Ltd, Guangzhou, 510663, China
| | - Lai-Qing Li
- Guangzhou Youdi Biotechnology Co., Ltd, Guangzhou, 510663, China.
| |
Collapse
|
38
|
Takahashi L, Ishigami T, Tomiyama H, Kato Y, Kikuchi H, Tasaki K, Yamashita J, Inoue S, Taguri M, Nagao T, Chikamori T, Ishikawa Y, Yokoyama U. Increased Plasma Levels of Myosin Heavy Chain 11 Is Associated with Atherosclerosis. J Clin Med 2021; 10:jcm10143155. [PMID: 34300321 PMCID: PMC8304775 DOI: 10.3390/jcm10143155] [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: 06/10/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 11/16/2022] Open
Abstract
Many studies have revealed numerous potential biomarkers for atherosclerosis, but tissue-specific biomarkers are still needed. Recent lineage-tracing studies revealed that smooth muscle cells (SMCs) contribute substantially to plaque formation, and the loss of SMCs causes plaque vulnerability. We investigated the association of SMC-specific myosin heavy chain 11 (myosin-11) with atherosclerosis. Forty-five patients with atherosclerosis and 34 control subjects were recruited into our study. In the atherosclerosis patients, 35 patients had either coronary artery disease (CAD) or peripheral artery disease (PAD), and 10 had both CAD and PAD. Coronary arteries isolated from five patients were subjected to histological study. Circulating myosin-11 levels were higher in the CAD or PAD group than in controls. The area under the receiver operating characteristic curve of myosin-11 was 0.954. Circulating myosin-11 levels in the CAD and PAD group were higher than in the CAD or PAD group, while high-sensitivity C-reactive protein concentrations did not differ between these groups. Multinomial logistic regression analyses showed a significant association of myosin-11 levels with the presence of multiple atherosclerotic regions. Myosin-11 was expressed in the medial layer of human atherosclerotic lesions where apoptosis elevated. Circulating myosin-11 levels may be useful for detecting spatial expansion of atherosclerotic regions.
Collapse
Affiliation(s)
- Lisa Takahashi
- Department of Cardiology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan; (L.T.); (H.T.); (J.Y.); (T.C.)
- Department of Physiology, Tokyo Medical University, 6-6-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan;
| | - Tomoaki Ishigami
- Department of Cardio-Renal Medicine and Medical Science, Yokohama City University, 3-9 Fukuura, Yokohama 236-0004, Japan;
| | - Hirofumi Tomiyama
- Department of Cardiology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan; (L.T.); (H.T.); (J.Y.); (T.C.)
| | - Yuko Kato
- Department of Physiology, Tokyo Medical University, 6-6-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan;
| | - Hiroyuki Kikuchi
- Department of Preventive Medicine and Public Health, Tokyo Medical University, 6-6-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan; (H.K.); (S.I.)
| | - Koichiro Tasaki
- Department of Pathology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan; (K.T.); (T.N.)
| | - Jun Yamashita
- Department of Cardiology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan; (L.T.); (H.T.); (J.Y.); (T.C.)
| | - Shigeru Inoue
- Department of Preventive Medicine and Public Health, Tokyo Medical University, 6-6-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan; (H.K.); (S.I.)
| | - Masataka Taguri
- Department of Data Science, Yokohama City University, 22-2 Seto, Kanazawa-ku, Yokohama 236-0027, Japan;
| | - Toshitaka Nagao
- Department of Pathology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan; (K.T.); (T.N.)
| | - Taishiro Chikamori
- Department of Cardiology, Tokyo Medical University, 6-7-1 Nishi-shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan; (L.T.); (H.T.); (J.Y.); (T.C.)
| | - Yoshihiro Ishikawa
- Cardiovascular Research Institute, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan;
| | - Utako Yokoyama
- Department of Physiology, Tokyo Medical University, 6-6-1 Shinjuku, Shinjuku-ku, Tokyo 160-8402, Japan;
- Cardiovascular Research Institute, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan;
- Correspondence: ; Tel.: +81-03-351-6141
| |
Collapse
|
39
|
Keskin M, Öcal L, Cerşit S, Yılmaz C, Küp A, Çelik M, Doğan S, Koyuncu A, Kaya A, Turkmen MM. The Predictive Role of a Novel Risk Index in Patients Undergoing Carotid Artery Stenting: Systemic Immune-Inflammation Index. J Stroke Cerebrovasc Dis 2021; 30:105955. [PMID: 34242858 DOI: 10.1016/j.jstrokecerebrovasdis.2021.105955] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/28/2021] [Accepted: 06/13/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Inflammatory mechanisms play an important role in both atherosclerosis and stroke. There are several inflammatory peripheral blood count markers associated with carotid artery stenosis degree, symptomatic carotid artery lesions and carotid artery stent restenosis that reported in previous studies. However, the prognostic role of the blood cell counts and their ratios in predicting in-hospital and long-term outcomes in patients undergoing carotid artery stenting (CAS) has not been comprehensively investigated. Systemic immune-inflammation index (SII) proved its' efficiency in patients with solid tumors and its' role was rarely examined in cardiovascular disorders and stroke. The current study evaluated the effect of this novel risk index on in-hospital and long-term outcomes in a large patient population who underwent CAS. METHOD A total of 732 patients with carotid artery stenosis who underwent CAS were enrolled to the study. SII was calculated using the following formula: neutrophil-to-lymphocyte ratio × total platelet count in the peripheral blood (per mm3) and the patients were stratified accordingly: T1, T2 and T3. In-hospital and 5-year outcomes were compared between the tertiles of SII. RESULTS During the hospitalization, major stroke, ipsilateral stoke, myocardial infarction, death and major adverse cardiovascular events (MACE) rates were significantly higher in high SII level (T3) compared to SII levels (T1 and 2). In long-term outcomes, ipsilateral stroke, major stroke, transient ischemic attack, death, and MACE were significantly higher in the patients with higher SII level (T3). The 5-year Kaplan-Meier overall survival for T1, T2, and T3 were 97.5%, 96.7% and 86.0% respectively. In-hospital and 5-year regression analyses demonstrated that high SII was independently associated with MACE and mortality. CONCLUSION SII was independently associated with in-hospital and long-term clinical outcomes in patients undergoing CAS. Immune and inflammation status, as assessed easily and quickly using SII, has a good discriminative value in these patients.
Collapse
Affiliation(s)
- Muhammed Keskin
- Cardiology, Bahcesehir University, Faculty of Medicine, Istanbul, Turkey.
| | - Lütfi Öcal
- Cardiology, Health Sciences University, Kartal Koşuyolu Heart Training and Research Hospital, Istanbul, Turkey
| | - Sinan Cerşit
- Cardiology, Health Sciences University, Kartal Koşuyolu Heart Training and Research Hospital, Istanbul, Turkey
| | - Cemalettin Yılmaz
- Cardiology, Health Sciences University, Kartal Koşuyolu Heart Training and Research Hospital, Istanbul, Turkey
| | - Ayhan Küp
- Cardiology, Health Sciences University, Kartal Koşuyolu Heart Training and Research Hospital, Istanbul, Turkey
| | - Mehmet Çelik
- Cardiology, Health Sciences University, Kartal Koşuyolu Heart Training and Research Hospital, Istanbul, Turkey
| | - Selami Doğan
- Cardiology, Bahcesehir University, Faculty of Medicine, Istanbul, Turkey
| | | | - Adnan Kaya
- Cardiology, Duzce University, Duzce, Turkey
| | - Mehmet Muhsin Turkmen
- Cardiology, Health Sciences University, Kartal Koşuyolu Heart Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
40
|
Elevation of CD40/CD40L Inflammatory Pathway Molecules in Carotid Plaques from Moderate-and-Severe Obstructive Sleep Apnea Patients. Diagnostics (Basel) 2021; 11:diagnostics11060935. [PMID: 34067481 PMCID: PMC8224789 DOI: 10.3390/diagnostics11060935] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 11/30/2022] Open
Abstract
A chronic inflammatory process characteristic of obstructive sleep apnea promotes vascular endothelial dysfunction and atherogenesis. This process can lead to destabilization and rupture of cardiovascular plaques, which clinically manifests as an acute coronary syndrome or stroke. The aim of this study was to investigate the inflammatory pathway leading to plaque destabilization in non-to-mild and moderate-to-severe groups of OSA patients. This prospective study involved enrollment of patients scheduled for endarterectomy. A sleep study was performed prior to surgery. Immunohistochemistry was performed on atherosclerotic plaques from carotid arteries obtained during standard open endarterectomy to determine levels of CD40, CD40L receptors, MCP-1, and MMP-9. The 46 patients included 14 controls, 13 with mild, 11 with moderate, and 8 with severe OSA. Increased expression of CD40, CD40L receptors, MCP-1, and MMP-9 were found to be proportionate with OSA severity. However, significant differences among groups were observed only for MCP-1 (p = 0.014). Increased expression of inflammatory markers (CD40, CD40L, MCP-1, MMP-9) is associated with increasing OSA severity. This suggests the CD40-CD4-L inflammatory pathway may contribute to plaque instability and rupture in OSA patients.
Collapse
|
41
|
Wang X, Fu Y, Xie Z, Cao M, Qu W, Xi X, Zhong S, Piao M, Peng X, Jia Y, Meng L, Tian J. Establishment of a Novel Mouse Model for Atherosclerotic Vulnerable Plaque. Front Cardiovasc Med 2021; 8:642751. [PMID: 33796572 PMCID: PMC8007762 DOI: 10.3389/fcvm.2021.642751] [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: 12/16/2020] [Accepted: 02/23/2021] [Indexed: 12/23/2022] Open
Abstract
Background and Aims: Acute coronary syndrome (ACS) is a group of clinical syndromes characterized by rupture or erosion of atherosclerotic unstable plaques. Effective intervention for vulnerable plaques (VP) is of great significance to reduce adverse cardiovascular events. Methods: Fbn1C1039G+/− mice were crossbred with LDLR−/− mice to obtain a novel model for atherosclerotic VP. After the mice were fed with a high-fat diet (HFD) for 12 or 24 weeks, pathological staining and immunohistochemistry analyses were employed to evaluate atherosclerotic lesions. Results: Compared to control mice, Fbn1C1039G+/−LDLR−/− mice developed more severe atherosclerotic lesions, and the positive area of oil red O staining in the aortic sinus was significantly increased after 12 weeks (21.7 ± 2.0 vs. 6.3 ± 2.1) and 24 weeks (32.6 ± 2.5 vs. 18.7 ± 2.6) on a HFD. Additional vulnerable plaque characteristics, including significantly larger necrotic cores (280 ± 19 vs. 105 ± 7), thinner fiber caps (14.0 ± 2.8 vs. 32.6 ± 2.7), apparent elastin fiber fragmentation and vessel dilation (3,010 ± 67 vs. 1,465 ± 49), a 2-fold increase in macrophage number (8.5 ± 1.0 vs. 5.0 ± 0.6), obviously decreased smooth muscle cell number (0.6 ± 0.1 vs. 2.1 ± 0.2) and an ~25% decrease in total collagen content (33.6 ± 0.3 vs. 44.9 ± 9.1) were observed in Fbn1C1039G+/−LDLR−/− mice compared with control mice after 24 weeks. Furthermore, spontaneous plaque rupture, neovascularization, and intraplaque hemorrhage were detected in the model mouse plaque regions but not in those of the control mice. Conclusions: Plaques in Fbn1C1039G+/−LDLR−/− mice fed a HFD show many features of human advanced atherosclerotic unstable plaques. These results suggest that the Fbn1C1039G+/−LDLR−/− mouse is a novel model for investigating the pathological and physiological mechanisms of advanced atherosclerotic unstable plaques.
Collapse
Affiliation(s)
- Xueyu Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Yahong Fu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Zulong Xie
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Muhua Cao
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Wenbo Qu
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Xiangwen Xi
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Shan Zhong
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Minghui Piao
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Xiang Peng
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Ying Jia
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, China
| | - Lingbo Meng
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinwei Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
42
|
Cao J, Yan P, Zhou Y, Zhou X, Sun Z, Zhu XQ. Clinical Utility of the Serum Level of Lipoprotein-Related Phospholipase A2 in Acute Ischemic Stroke With Cerebral Artery Stenosis. Front Neurol 2021; 12:642483. [PMID: 33746893 PMCID: PMC7969974 DOI: 10.3389/fneur.2021.642483] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/02/2021] [Indexed: 12/01/2022] Open
Abstract
We aimed to study the clinical utility of serum lipoprotein-associated phospholipase A2 (Lp-PLA2) in acute ischemic stroke (AIS) with cerebral artery stenosis (CAS). We included 200 AIS patients and 90 healthy controls in this study. AIS patients were classified into three subgroups depending on the severity of CAS. They were also classified based on the stability of the carotid plaques. Spearman correlation analysis was performed to determine the correlation relationship between the level of Lp-PLA2 and neurologic injury. Binary logistic regression analysis was performed to determine the independent risk factors for AIS. Receiver operating characteristic (ROC) analysis was performed to assess the diagnostic value of Lp-PLA2 for AIS and for the degree of CAS. We found that the serum level of Lp-PLA2 in AIS patients was significantly higher than that in the control group. Lp-PLA2 was further identified as an independent risk factor for AIS (p = 0.001, OR = 1.057). In addition, serum Lp-PLA2 level was the highest in AIS patients with severe CAS or occlusion. Lp-PLA2 level was higher in AIS patients with unstable plaques and in AIS patients with moderate to severe neurological injury. Lp-PLA2 level was positively correlated with National Institutes of Health Stroke Scale (NIHSS) score (r = 0.335, p = 0.001). We found that the optimal cut-off value for Lp-PLA2 level was 123.365 ng/ml, at which the sensitivity and specificity for the diagnosis of ACI were 74.5 and 86.7%, respectively, and the area under ROC curve (AUC) was 0.892. Similarly, the optimal value for Lp-PLA2 level was 136.46 ng/ml, at which the sensitivity and specificity for the diagnosis of the presence of moderate to severe artery stenosis or occlusion were 79.6 and 95.2%, respectively, and the AUC was 0.938. The ROC curve indicated that serum Lp-PLA2 level has an excellent diagnostic value for AIS and severe stenosis. Based on these results we conclude that Lp-PLA2 could be a potential biomarker to complement the current imaging methods in the prediction and diagnosis of AIS. An elevated Lp-PLA2 level is also correlated with carotid plaque instability, severe neurological injury and cerebrovascular stenosis. Future longitudinal studies are needed to determine whether there is a causative relationship between Lp-PLA2 and AIS.
Collapse
Affiliation(s)
- Jing Cao
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ping Yan
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yajun Zhou
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xia Zhou
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhongwu Sun
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiao-Qun Zhu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
43
|
Detering L, Abdilla A, Luehmann HP, Williams JW, Huang LH, Sultan D, Elvington A, Heo GS, Woodard PK, Gropler RJ, Randolph GJ, Hawker CJ, Liu Y. CC Chemokine Receptor 5 Targeted Nanoparticles Imaging the Progression and Regression of Atherosclerosis Using Positron Emission Tomography/Computed Tomography. Mol Pharm 2021; 18:1386-1396. [PMID: 33591187 PMCID: PMC8737066 DOI: 10.1021/acs.molpharmaceut.0c01183] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Chemokines and chemokine receptors play an important role in the initiation and progression of atherosclerosis by mediating the trafficking of inflammatory cells. Chemokine receptor 5 (CCR5) has major implications in promoting the development of plaques to advanced stage and related vulnerability. CCR5 antagonist has demonstrated the effective inhibition of atherosclerotic progression in mice, making it a potential biomarker for atherosclerosis management. To accurately determine CCR5 in vivo, we synthesized CCR5 targeted Comb nanoparticles through a modular design and construction strategy with control over the physiochemical properties and functionalization of CCR5 targeting peptide d-Ala-peptide T-amide (DAPTA-Comb). In vivo pharmacokinetic evaluation through 64Cu radiolabeling showed extended blood circulation of 64Cu-DAPTA-Combs conjugated with 10%, 25%, and 40% DAPTA. The different organ distribution profiles of the three nanoparticles demonstrated the effect of DAPTA on not only physicochemical properties but also targeting efficiency. In vivo positron emission tomography/computed tomography (PET/CT) imaging in an apolipoprotein E knockout mouse atherosclerosis model (ApoE-/-) showed that the three 64Cu-DAPTA-Combs could sensitively and specifically detect CCR5 along the progression of atherosclerotic lesions. In an ApoE-encoding adenoviral vector (AAV) induced plaque regression ApoE-/- mouse model, decreased monocyte recruitment, CD68+ macrophages, CCR5 expression, and plaque size were all associated with reduced PET signals, which not only further confirmed the targeting efficiency of 64Cu-DAPTA-Combs but also highlighted the potential of these targeted nanoparticles for atherosclerosis imaging. Moreover, the up-regulation of CCR5 and colocalization with CD68+ macrophages in the necrotic core of ex vivo human plaque specimens warrant further investigation for atherosclerosis prognosis.
Collapse
Affiliation(s)
- Lisa Detering
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri 63110, United States
| | - Allison Abdilla
- Materials Department, University of California, Santa Barbara, California 93106, United States
| | - Hannah P Luehmann
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri 63110, United States
| | - Jesse W Williams
- Department of Pathology and Immunology, Washington University, St. Louis, Missouri 63110, United States
| | - Li-Hao Huang
- Department of Pathology and Immunology, Washington University, St. Louis, Missouri 63110, United States
| | - Deborah Sultan
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri 63110, United States
| | - Andrew Elvington
- Department of Pathology and Immunology, Washington University, St. Louis, Missouri 63110, United States
| | - Gyu Seong Heo
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri 63110, United States
| | - Pamela K Woodard
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri 63110, United States
| | - Robert J Gropler
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri 63110, United States
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University, St. Louis, Missouri 63110, United States
| | - Craig J Hawker
- Materials Department, University of California, Santa Barbara, California 93106, United States
| | - Yongjian Liu
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri 63110, United States
| |
Collapse
|
44
|
Shokr H, Gherghel D. European Society of Cardiology/European Society of Hypertension versus the American College of Cardiology/American Heart Association guidelines on the cut-off values for early hypertension: a microvascular perspective. Sci Rep 2021; 11:3473. [PMID: 33568754 PMCID: PMC7876123 DOI: 10.1038/s41598-021-83096-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 01/11/2021] [Indexed: 11/09/2022] Open
Abstract
The aim of this study was to investigate retinal and peripheral microvascular function in asymptomatic individuals that fall into different BP groups when using either the ESC/ESH or the ACC/AHA guidelines. Retinal and peripheral microvascular function was assessed in 358 participants by means of dynamic retinal vessel analysis and digital thermal monitoring, respectively. Blood pressure and lipid panel were also evaluated. Retinal vascular function measured in all groups belonging to the ACC/ASH classifications were within the normal values for age-matched normal population. Individuals classed as grade 1 hypertension according to the ESC/ESH guidelines, however, exhibited a significantly decreased artery baseline (p = 0.0004) and MC (p = 0.040), higher slopeAD (p = 0.0018) and decreased vein MC (p = 0.0446) compared to age matched normal individuals. In addition, they also had significant lower artery baseline, artery BDF, MD and MC than individuals classed as stage 1 hypertension based on the ACC/ASH guidelines (p = 0.00022, p = 0.0179, p = 0.0409 and p = 0.0329 respectively). Peripheral vascular reactivity (aTR) was lower in ESC /ESH grade I compared to those graded ACC/ASH stage I hypertension (p = 0.0122). The conclusion of this study is that microvascular dysfunctions is present at multiple levels only in individuals with ESC/ESH grade 1 hypertension. This observation could be important when deciding personalised care in individuals with early hypertensive changes.
Collapse
Affiliation(s)
- H Shokr
- Vascular Research Laboratory, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK
| | - D Gherghel
- Vascular Research Laboratory, College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK.
| |
Collapse
|
45
|
Prunicki M, Cauwenberghs N, Ataam JA, Movassagh H, Kim JB, Kuznetsova T, Wu JC, Maecker H, Haddad F, Nadeau K. Immune biomarkers link air pollution exposure to blood pressure in adolescents. Environ Health 2020; 19:108. [PMID: 33066786 PMCID: PMC7566149 DOI: 10.1186/s12940-020-00662-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 10/01/2020] [Indexed: 05/08/2023]
Abstract
BACKGROUND Childhood exposure to air pollution contributes to cardiovascular disease in adulthood. Immune and oxidative stress disturbances might mediate the effects of air pollution on the cardiovascular system, but the underlying mechanisms are poorly understood in adolescents. Therefore, we aimed to identify immune biomarkers linking air pollution exposure and blood pressure levels in adolescents. METHODS We randomly recruited 100 adolescents (mean age, 16 years) from Fresno, California. Using central-site data, spatial-temporal modeling, and distance weighting exposures to the participant's home, we estimated average pollutant levels [particulate matter (PM), polyaromatic hydrocarbons (PAH), ozone (O3), carbon monoxide (CO) and nitrogen oxides (NOx)]. We collected blood samples and vital signs on health visits. Using proteomic platforms, we quantitated markers of inflammation, oxidative stress, coagulation, and endothelial function. Immune cellular characterization was performed via mass cytometry (CyTOF). We investigated associations between pollutant levels, cytokines, immune cell types, and blood pressure (BP) using partial least squares (PLS) and linear regression, while adjusting for important confounders. RESULTS Using PLS, biomarkers explaining most of the variance in air pollution exposure included markers of oxidative stress (GDF-15 and myeloperoxidase), acute inflammation (C-reactive protein), hemostasis (ADAMTS, D-dimer) and immune cell types such as monocytes. Most of these biomarkers were independently associated with the air pollution levels in fully adjusted regression models. In CyTOF analyses, monocytes were enriched in participants with the highest versus the lowest PM2.5 exposure. In both PLS and linear regression, diastolic BP was independently associated with PM2.5, NO, NO2, CO and PAH456 pollution levels (P ≤ 0.009). Moreover, monocyte levels were independently related to both air pollution and diastolic BP levels (P ≤ 0.010). In in vitro cell assays, plasma of participants with high PM2.5 exposure induced endothelial dysfunction as evaluated by eNOS and ICAM-1 expression and tube formation. CONCLUSIONS For the first time in adolescents, we found that ambient air pollution levels were associated with oxidative stress, acute inflammation, altered hemostasis, endothelial dysfunction, monocyte enrichment and diastolic blood pressure. Our findings provide new insights on pollution-related immunological and cardiovascular disturbances and advocate preventative measures of air pollution exposure.
Collapse
Affiliation(s)
- Mary Prunicki
- Sean N Parker Center for Allergy and Asthma Research, Stanford University, Stanford, USA
| | - Nicholas Cauwenberghs
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Jennifer Arthur Ataam
- Research and Innovation Unit, INSERM U999, DHU TORINO, Paris Sud University, Marie Lannelongue Hospital, Le Plessis Robinson, France
- Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, USA
| | - Hesam Movassagh
- Sean N Parker Center for Allergy and Asthma Research, Stanford University, Stanford, USA
| | - Juyong Brian Kim
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Tatiana Kuznetsova
- Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Joseph C. Wu
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Holden Maecker
- Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, USA
| | - Francois Haddad
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University, Stanford, USA
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, USA
| | - Kari Nadeau
- Sean N Parker Center for Allergy and Asthma Research, Stanford University, Stanford, USA
| |
Collapse
|
46
|
Kashiwazaki D, Yamamoto S, Akioka N, Hori E, Shibata T, Kuwayama N, Noguchi K, Kuroda S. High-intensity vessel sign on fluid-attenuated inversion recovery imaging: a novel imaging marker of high-risk carotid stenosis-a MRI and SPECT study. Acta Neurochir (Wien) 2020; 162:2573-2581. [PMID: 32458404 DOI: 10.1007/s00701-020-04408-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/10/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND Measurement of luminal stenosis and determination of plaque instability using MR plaque imaging are effective strategies for evaluating high-risk carotid stenosis. Nevertheless, new methods are required to identify patients with carotid stenosis at risk of future stroke. We aimed to clarify the mechanisms and clinical implications of the hyperintense vessel sign (HVS) as a marker of high-risk carotid stenosis. METHODS We included 148 patients who underwent carotid stent (CAS) or carotid endarterectomy (CEA). MRI FLAIR was performed to detect HVS prior to and within 7 days after CAS/CEA. MR plaque imaging and 123I-iodoamphetamine SPECT was performed prior to CEA/CAS. Detailed characteristics of HVS were categorized in terms of symptomatic status, hemodynamic state, plaque composition, and HVS on time series. RESULTS Forty-six of 80 symptomatic hemispheres (57.5%) and 5 of 68 asymptomatic hemispheres (7.4%) presented HVS (P < 0.01). Of the 46 symptomatic hemispheres with HVS, 19 (41.3%) presented with hemodynamic impairment and 27 (58.7%) presented without hemodynamic impairment. Of 19 hemispheres with hemodynamic impairment, 12 subjects (63.2%) showed high intensity and 7 (36.8%) showed iso-intensity plaques on T1WI. All 27 hemispheres without hemodynamic impairment showed high-intensity plaques. Of the five asymptomatic and HVS-positive hemispheres, one showed hemodynamic impairment; MR plaque imaging revealed T1 iso-intensity. The other four hemispheres that did not show hemodynamic impairment showed T1WI high-intensity plaques. CONCLUSION There are two possible mechanisms of HVS, hemodynamic impairment due to severe carotid stenosis and micro-embolism from unstable plaques. HVS could be a radiological marker for high-risk carotid stenosis.
Collapse
Affiliation(s)
- Daina Kashiwazaki
- Department of Neurosurgery, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan.
| | - Shusuke Yamamoto
- Department of Neurosurgery, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Naoki Akioka
- Department of Neurosurgery, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Emiko Hori
- Department of Neurosurgery, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Takashi Shibata
- Department of Neurosurgery, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Naoya Kuwayama
- Department of Neurosurgery, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| | - Kyo Noguchi
- Department of Radiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Satoshi Kuroda
- Department of Neurosurgery, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, 930-0194, Japan
| |
Collapse
|
47
|
MicroRNAs as sentinels and protagonists of carotid artery thromboembolism. Clin Sci (Lond) 2020; 134:169-192. [PMID: 31971230 DOI: 10.1042/cs20190651] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/12/2019] [Accepted: 01/03/2020] [Indexed: 02/06/2023]
Abstract
Stroke is the leading cause of serious disability in the world and a large number of ischemic strokes are due to thromboembolism from unstable carotid artery atherosclerotic plaque. As it is difficult to predict plaque rupture and surgical treatment of asymptomatic disease carries a risk of stroke, carotid disease continues to present major challenges with regard to clinical decision-making and revascularization. There is therefore an imminent need to better understand the molecular mechanisms governing plaque instability and rupture, as this would allow for the development of biomarkers to identify at-risk asymptomatic carotid plaque prior to disease progression and stroke. Further, it would aid in creation of therapeutics to stabilize carotid plaque. MicroRNAs (miRNAs) have been implicated as key protagonists in various stages of atherosclerotic plaque initiation, development and rupture. Notably, they appear to play a crucial role in carotid artery thromboembolism. As the molecular pathways governing the role of miRNAs are being uncovered, we are learning that their involvement is complex, tissue- and stage-specific, and highly selective. Notably, miRNAs can be packaged and secreted in extracellular vesicles (EVs), where they participate in cell-cell communication. The measurement of EV-encapsulated miRNAs in the circulation may inform disease mechanisms occurring in the plaque itself, and therefore may serve as sentinels of unstable plaque as well as therapeutic targets.
Collapse
|
48
|
Li M, Chen Y, Zhang Y, Li D, Liu J. Correlation between monocyte chemoattractant protein-1/chemokine (C-C motif) ligand 2 and coronary plaque characteristics. Exp Biol Med (Maywood) 2020; 245:1335-1343. [PMID: 32640896 DOI: 10.1177/1535370220941424] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
IMPACT STATEMENT Vulnerable plaques are plaques which are susceptible to rupture or thrombosis and trigger a series of adverse events such as coronary disorders. CCL2 is a soluble basic protein belonging to the CC subfamily. Previous studies have been investigated on the correlation between inflammatory factors and clinical events, but there are few studies on the correlation between CCL2 and plaque characteristics. Our study found that the high expression of CCL2 is involved in multiple processes in the genesis and progression of coronary artery disease, and would be a potential clinical prognostic indicator. In addition, high expression of CCL2 may be related to gene pathways such as Nod-like receptor signaling pathway, suggesting that CCL2 is involved in the inflammatory response and immune process of coronary artery disease.
Collapse
Affiliation(s)
- Meng Li
- Department of Cardiology, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Zhengzhou University, Zhengzhou 450003, China
| | - Yan Chen
- Department of Cardiology, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Zhengzhou University, Zhengzhou 450003, China
| | - Yan Zhang
- Department of Cardiology, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Zhengzhou University, Zhengzhou 450003, China
| | - Danna Li
- Department of Ultrasound, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jun Liu
- Department of Cardiology, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Zhengzhou University, Zhengzhou 450003, China
| |
Collapse
|
49
|
Matrix Metalloproteinases as Biomarkers of Atherosclerotic Plaque Instability. Int J Mol Sci 2020; 21:ijms21113946. [PMID: 32486345 PMCID: PMC7313469 DOI: 10.3390/ijms21113946] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/27/2020] [Accepted: 05/29/2020] [Indexed: 02/07/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-dependent endopeptidases responsible for tissue remodeling and degradation of extracellular matrix (ECM) proteins. MMPs may modulate various cellular and signaling pathways in atherosclerosis responsible for progression and rupture of atherosclerotic plaques. The effect of MMPs polymorphisms and the expression of MMPs in both the atherosclerotic plaque and plasma was shown. They are independent predictors of atherosclerotic plaque instability in stable coronary heart disease (CHD) patients. Increased levels of MMPs in patients with advanced cardiovascular disease (CAD) and acute coronary syndrome (ACS) was associated with future risk of cardiovascular events. These data confirm that MMPs may be biomarkers in plaque instability as they target in potential drug therapies for atherosclerosis. They provide important prognostic information, independent of traditional risk factors, and may turn out to be useful in improving risk stratification.
Collapse
|
50
|
Yang YL, Wu CH, Hsu PF, Chen SC, Huang SS, Chan WL, Lin SJ, Chou CY, Chen JW, Pan JP, Charng MJ, Chen YH, Wu TC, Lu TM, Huang PH, Cheng HM, Huang CC, Sung SH, Lin YJ, Leu HB. Systemic immune-inflammation index (SII) predicted clinical outcome in patients with coronary artery disease. Eur J Clin Invest 2020; 50:e13230. [PMID: 32291748 DOI: 10.1111/eci.13230] [Citation(s) in RCA: 336] [Impact Index Per Article: 67.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 03/28/2020] [Accepted: 04/02/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND This study examines the predictive value of a novel systemic immune-inflammation index (SII, platelet × neutrophil/lymphocyte ratio) in coronary artery disease (CAD) patients. METHODS A total of 5602 CAD patients who had undergone a percutaneous coronary intervention (PCI) were enrolled. They were divided into two groups by baseline SII score (high SII vs low SII) to analyse the relationship between SII groups and the long-term outcome. The primary outcomes were major cardiovascular events (MACE) which includes nonfatal myocardial infarction (MI), nonfatal stroke and cardiac death. Secondary outcomes included a composite of MACE and hospitalization for congestive heart failure. RESULTS An optimal SII cut-off point of 694.3 × 109 was identified for MACE in the CAD training cohort (n = 373) and then verified in the second larger CAD cohort (n = 5602). Univariate and multivariate analyses showed that a higher SII score (≥694.3) was independently associated with increased risk of developing cardiac death (HR: 2.02; 95% CI: 1.43-2.86), nonfatal MI (HR: 1.42; 95% CI: 1.09-1.85), nonfatal stroke (HR: 1.96; 95% CI: 1.28-2.99), MACE (HR: 1.65; 95% CI: 1.36-2.01) and total major events (HR: 1.53; 95% CI: 1.32-1.77). In addition, the SII significantly improved risk stratification of MI, cardiac death, heart failure, MACE and total major events than conventional risk factors in CAD patients by the significant increase in the C-index (P < .001) and reclassification risk categories by significant NRI (P < .05) and IDI (P < .05). CONCLUSIONS SII had a better prediction of major cardiovascular events than traditional risk factors in CAD patients after coronary intervention.
Collapse
Affiliation(s)
- Ya-Ling Yang
- Division of Cardiology, Department of Medicine, Healthcare and Management Center, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine and Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan.,Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital Taoyuan Branch, Taoyuan, Taiwan
| | - Cheng-Hsueh Wu
- Division of Cardiology, Department of Medicine, Healthcare and Management Center, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Pai-Feng Hsu
- Division of Cardiology, Department of Medicine, Healthcare and Management Center, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine and Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan.,Healthcare and Management Center, Taipei, Taiwan
| | - Su-Chan Chen
- Division of Cardiology, Department of Medicine, Healthcare and Management Center, Taipei, Taiwan.,Institute of Clinical Medicine and Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Shao-Sung Huang
- Division of Cardiology, Department of Medicine, Healthcare and Management Center, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan.,Healthcare and Management Center, Taipei, Taiwan
| | - Wan Leong Chan
- Division of Cardiology, Department of Medicine, Healthcare and Management Center, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine and Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Shing-Jong Lin
- Division of Cardiology, Department of Medicine, Healthcare and Management Center, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine and Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Chia-Yu Chou
- Division of Cardiology, Department of Medicine, Healthcare and Management Center, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Jaw-Wen Chen
- Division of Cardiology, Department of Medicine, Healthcare and Management Center, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine and Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Ju-Pin Pan
- Division of Cardiology, Department of Medicine, Healthcare and Management Center, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Min-Ji Charng
- Division of Cardiology, Department of Medicine, Healthcare and Management Center, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Ying-Hwa Chen
- Division of Cardiology, Department of Medicine, Healthcare and Management Center, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Tao-Cheng Wu
- Division of Cardiology, Department of Medicine, Healthcare and Management Center, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Tse-Min Lu
- Division of Cardiology, Department of Medicine, Healthcare and Management Center, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Po-Hsun Huang
- Division of Cardiology, Department of Medicine, Healthcare and Management Center, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hao-Min Cheng
- Division of Cardiology, Department of Medicine, Healthcare and Management Center, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine and Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Chin-Chou Huang
- Division of Cardiology, Department of Medicine, Healthcare and Management Center, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Medical Education, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shih-Hsien Sung
- Division of Cardiology, Department of Medicine, Healthcare and Management Center, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yenn-Jiang Lin
- Division of Cardiology, Department of Medicine, Healthcare and Management Center, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hsin-Bang Leu
- Division of Cardiology, Department of Medicine, Healthcare and Management Center, Taipei, Taiwan.,Faculty of Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Clinical Medicine and Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan.,Healthcare and Management Center, Taipei, Taiwan
| |
Collapse
|