151
|
Zhou J, Liu L, Wu P, Zhao L, Wu Y. Fusobacterium nucleatum Accelerates Atherosclerosis via Macrophage-Driven Aberrant Proinflammatory Response and Lipid Metabolism. Front Microbiol 2022; 13:798685. [PMID: 35359716 PMCID: PMC8963492 DOI: 10.3389/fmicb.2022.798685] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/03/2022] [Indexed: 12/18/2022] Open
Abstract
Periodontitis, an oral chronic inflammatory disease, is reported to show an association with atherosclerotic vascular disease. Fusobacterium nucleatum is an oral commensal bacterium that is abundantly implicated in various forms of periodontal diseases; however, its role in the pathogenesis of atherosclerosis is unclear. This study aimed to elucidate the underlying pathogenic mechanisms of atherosclerosis induced by F. nucleatum to provide new insight on the prevention and treatment of atherosclerosis. We used an animal model, that is, ApoE–/– mice were infected with F. nucleatum by oral gavage, and in vitro co-culture models to assess the pathogenicity of F. nucleatum. The results indicate that F. nucleatum ATCC 25586 invaded aortic tissues and substantially increased the progression of atherosclerotic lesions. In addition, F. nucleatum changed plaque composition into a less-stable phenotype, characterized with increased subcutaneous macrophage infiltration, M1 polarization, lipid deposition, cell apoptosis, and reduced extracellular matrix and collagen content. The serum levels of pro-atherosclerotic factors, such as interleukin (IL)-6, IL-1β, tumor necrosis factor (TNF)-α, monocyte chemoattractant protein-1 (MCP-1), c-reactive protein, and oxidized low-density lipoprotein (ox-LDL) and microRNAs (miR-146a, miR-155, and miR-23b) were considerably increased after F. nucleatum stimulation, whereas HDL-c level was reduced. F. nucleatum induced in vitro macrophage apoptosis in a time- and dose-dependent manner. F. nucleatum facilitated ox-LDL–induced cholesterol phagocytosis and accumulation by regulating the expression of lipid metabolism-related genes (AR-A1, ACAT1, ABCA1, and ABCG1). F. nucleatum further worsened the atherosclerotic plaque microenvironment by considerably increasing the levels of IL-6; IL-1β; TNF-α; MCP-1; and MMP-2, 8, and 9 and by suppressing fibronectin (FN) 1 levels during foam cell formation. This study shows that F. nucleatum ATCC 25586 is implicated in atherosclerosis by causing aberrant activation and lipid metabolism in macrophage.
Collapse
Affiliation(s)
- Jieyu Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lin Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Peiyao Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lei Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yafei Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
152
|
Li S, Wang J, Yan Y, Zhang Z, Gong W, Nie S. Clinical Characterization and Possible Pathological Mechanism of Acute Myocardial Injury in COVID-19. Front Cardiovasc Med 2022; 9:862571. [PMID: 35387441 PMCID: PMC8979292 DOI: 10.3389/fcvm.2022.862571] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 02/14/2022] [Indexed: 01/08/2023] Open
Abstract
COVID-19 is a respiratory disease that can cause damage to multiple organs throughout the body. Cardiovascular complications related to COVID-19 mainly include acute myocardial injury, heart failure, acute coronary syndrome, arrhythmia, myocarditis. Among them, myocardial injury is the most common complication in COVID-19 hospitalized patients, and is associated with poor prognosis such as death and arrhythmias. There is a continuous relationship between myocardial injury and the severity of COVID-19. The incidence of myocardial injury is higher in critically ill patients and dead patients, and myocardial injury is more likely to occur in the elderly critically ill patients with comorbidities. Myocardial injury is usually accompanied by more electrocardiogram abnormalities, higher inflammation markers and more obvious echocardiographic abnormalities. According to reports, COVID-19 patients with a history of cardiovascular disease have a higher in-hospital mortality, especially in the elder patients. At present, the mechanism of myocardial injury in COVID-19 is still unclear. There may be direct injury of myocardial cells, systemic inflammatory response, hypoxia, prethrombotic and procoagulant state, myocardial interstitial fibrosis, interferon-mediated immune response and coronary artery plaque instability and other related factors, and angiotensin-converting enzyme-2 receptor may play a key role in the myocardial injury in COVID-19.
Collapse
Affiliation(s)
- Siyi Li
- Coronary Heart Disease Center, Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Jinan Wang
- The Affiliated Rehabilitation Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Yan
- Coronary Heart Disease Center, Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Zekun Zhang
- Coronary Heart Disease Center, Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Wei Gong
- Coronary Heart Disease Center, Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Shaoping Nie
- Coronary Heart Disease Center, Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| |
Collapse
|
153
|
Baigent C, Windecker S, Andreini D, Arbelo E, Barbato E, Bartorelli AL, Baumbach A, Behr ER, Berti S, Bueno H, Capodanno D, Cappato R, Chieffo A, Collet JP, Cuisset T, de Simone G, Delgado V, Dendale P, Dudek D, Edvardsen T, Elvan A, González-Juanatey JR, Gori M, Grobbee D, Guzik TJ, Halvorsen S, Haude M, Heidbuchel H, Hindricks G, Ibanez B, Karam N, Katus H, Klok FA, Konstantinides SV, Landmesser U, Leclercq C, Leonardi S, Lettino M, Marenzi G, Mauri J, Metra M, Morici N, Mueller C, Petronio AS, Polovina MM, Potpara T, Praz F, Prendergast B, Prescott E, Price S, Pruszczyk P, Rodríguez-Leor O, Roffi M, Romaguera R, Rosenkranz S, Sarkozy A, Scherrenberg M, Seferovic P, Senni M, Spera FR, Stefanini G, Thiele H, Tomasoni D, Torracca L, Touyz RM, Wilde AA, Williams B. European Society of Cardiology guidance for the diagnosis and management of cardiovascular disease during the COVID-19 pandemic: part 1-epidemiology, pathophysiology, and diagnosis. Eur Heart J 2022; 43:1033-1058. [PMID: 34791157 PMCID: PMC8690026 DOI: 10.1093/eurheartj/ehab696] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/08/2021] [Accepted: 09/13/2021] [Indexed: 01/08/2023] Open
Abstract
AIMS Since its emergence in early 2020, the novel severe acute respiratory syndrome coronavirus 2 causing coronavirus disease 2019 (COVID-19) has reached pandemic levels, and there have been repeated outbreaks across the globe. The aim of this two-part series is to provide practical knowledge and guidance to aid clinicians in the diagnosis and management of cardiovascular disease (CVD) in association with COVID-19. METHODS AND RESULTS A narrative literature review of the available evidence has been performed, and the resulting information has been organized into two parts. The first, reported here, focuses on the epidemiology, pathophysiology, and diagnosis of cardiovascular (CV) conditions that may be manifest in patients with COVID-19. The second part, which will follow in a later edition of the journal, addresses the topics of care pathways, treatment, and follow-up of CV conditions in patients with COVID-19. CONCLUSION This comprehensive review is not a formal guideline but rather a document that provides a summary of current knowledge and guidance to practicing clinicians managing patients with CVD and COVID-19. The recommendations are mainly the result of observations and personal experience from healthcare providers. Therefore, the information provided here may be subject to change with increasing knowledge, evidence from prospective studies, and changes in the pandemic. Likewise, the guidance provided in the document should not interfere with recommendations provided by local and national healthcare authorities.
Collapse
|
154
|
Wen D, An R, Lin S, Yang W, Jia Y, Zheng M. Influence of Different Segmentations on the Diagnostic Performance of Pericoronary Adipose Tissue. Front Cardiovasc Med 2022; 9:773524. [PMID: 35310984 PMCID: PMC8929663 DOI: 10.3389/fcvm.2022.773524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 02/10/2022] [Indexed: 12/01/2022] Open
Abstract
Objective To investigate the influence of different segmentations on the diagnostic performance of pericoronary adipose tissue (PCAT) CT attenuation and radiomics features for the prediction of ischemic coronary artery stenosis. Methods From June 2016 to December 2018, 108 patients with 135 vessels were retrospectively analyzed in the present study. Vessel-based PCAT was segmented along the 40 mm-long proximal segments of three major epicardial coronary arteries, while lesion-based PCAT was defined around coronary lesions. CT attenuation and radiomics features derived from two segmentations were calculated and extracted. The diagnostic performance of PCAT CT attenuation or radiomics models in predicting ischemic coronary stenosis were also compared between vessel-based and lesion-based segmentations. Results The mean PCAT CT attenuation was −75.7 ± 9.1 HU and −76.1 ± 8.1 HU (p = 0.395) for lesion-based and vessel-based segmentations, respectively. A strong correlation was found between vessel-based and lesion-based PCAT CT attenuation for all cohort and subgroup analyses (all p < 0.01). A good agreement for all cohort and subgroup analyses was also detected between two segmentations. The diagnostic performance was comparable between vessel-based and lesion based PCAT CT attenuation in predicting ischemic stenosis. The radiomics features of PCAT based on vessel or lesion segmentation can both adequately identify the ischemic stenosis. However, no significant difference was detected between the two segmentations. Conclusions The quantitative evaluation of PCAT can be reliably measured both from vessel-based and lesion-based segmentation. Furthermore, the radiomics analysis of PCAT may potentially help predict hemodynamically significant coronary artery stenosis.
Collapse
Affiliation(s)
- Didi Wen
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Rui An
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | | | - Wangwei Yang
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yuyang Jia
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Minwen Zheng
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
- *Correspondence: Minwen Zheng
| |
Collapse
|
155
|
Tai YH, Chang ML, Chu PH, Yeh CC, Cherng YG, Chen TL, Liao CC. Risk of Acute Myocardial Infarction in Patients with Gastroenteritis: A Nationwide Case-Control Study. J Clin Med 2022; 11:jcm11051341. [PMID: 35268431 PMCID: PMC8911228 DOI: 10.3390/jcm11051341] [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: 01/18/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 01/27/2023] Open
Abstract
Gastroenteritis promotes the development of systemic inflammation and a hypercoagulable state. There are limited data regarding the association between gastroenteritis and acute myocardial infarction (AMI). We aimed to evaluate the risk of AMI after an episode of gastroenteritis. In this nested case-control study, we selected patients who were hospitalized for AMI (N = 103,584) as a case group during 2010-2017 and performed propensity score matching (case-control ratio 1:1) to select eligible controls from insurance research data in Taiwan. We applied multivariable logistic regressions to calculate adjusted odds ratios (ORs) with 95% confidence intervals (CIs) for the risk of AMI associated with recent gastroenteritis within 14 days before AMI. We also compared the outcomes after AMI in patients with or without gastroenteritis. A total of 1381 patients (1.3%) with AMI had a prior episode of gastroenteritis compared to 829 (0.8%) among the controls. Gastroenteritis was significantly associated with a subsequent risk of AMI (adjusted OR: 1.68, 95% CI: 1.54-1.83), which was augmented in hospitalizations for gastroenteritis (adjusted OR: 2.50, 95% CI: 1.20-5.21). The outcomes after AMI were worse in patients with gastroenteritis than in those without gastroenteritis, including increased 30-day in-hospital mortality (adjusted OR: 1.28, 95% CI: 1.08-1.52), medical expenditure, and length of hospital stay. Gastroenteritis may act as a trigger for AMI and correlates with worse post-AMI outcomes. Strategies of aggressive hydration and/or increased antithrombotic therapies for this susceptible population should be further developed.
Collapse
Affiliation(s)
- Ying-Hsuan Tai
- Department of Anesthesiology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan; (Y.-H.T.); (Y.-G.C.)
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Ming-Long Chang
- Department of Emergency Medicine, Taipei Medical University Hospital, Taipei 110, Taiwan;
- Department of Emergency Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Pao-Hsien Chu
- Division of Cardiology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan;
| | - Chun-Chieh Yeh
- Department of Surgery, China Medical University Hospital, Taichung 404, Taiwan;
- Department of Surgery, University of Illinois, Chicago, IL 60637, USA
| | - Yih-Giun Cherng
- Department of Anesthesiology, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235, Taiwan; (Y.-H.T.); (Y.-G.C.)
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Ta-Liang Chen
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Department of Anesthesiology, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
- Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Chien-Chang Liao
- Department of Anesthesiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Anesthesiology and Health Policy Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
- Department of Anesthesiology, Taipei Medical University Hospital, Taipei 110, Taiwan
- Research Center of Big Data and Meta-Analysis, Wan Fang Hospital, Taipei Medical University, Taipei 116, Taiwan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan
- Correspondence: or
| |
Collapse
|
156
|
Zhang J, Xie M, Huang X, Chen G, Yin Y, Lu X, Feng G, Yu R, Chen L. The Effects of Porphyromonas gingivalis on Atherosclerosis-Related Cells. Front Immunol 2022; 12:766560. [PMID: 35003080 PMCID: PMC8734595 DOI: 10.3389/fimmu.2021.766560] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/30/2021] [Indexed: 12/21/2022] Open
Abstract
Atherosclerosis (AS), one of the most common types of cardiovascular disease, has initially been attributed to the accumulation of fats and fibrous materials. However, more and more researchers regarded it as a chronic inflammatory disease nowadays. Infective disease, such as periodontitis, is related to the risk of atherosclerosis. Porphyromonas gingivalis (P. gingivalis), one of the most common bacteria in stomatology, is usually discovered in atherosclerotic plaque in patients. Furthermore, it was reported that P. gingivalis can promote the progression of atherosclerosis. Elucidating the underlying mechanisms of P. gingivalis in atherosclerosis attracted attention, which is thought to be crucial to the therapy of atherosclerosis. Nevertheless, the pathogenesis of atherosclerosis is much complicated, and many kinds of cells participate in it. By summarizing existing studies, we find that P. gingivalis can influence the function of many cells in atherosclerosis. It can induce the dysfunction of endothelium, promote the formation of foam cells as well as the proliferation and calcification of vascular smooth muscle cells, and lead to the imbalance of regulatory T cells (Tregs) and T helper (Th) cells, ultimately promoting the occurrence and development of atherosclerosis. This article summarizes the specific mechanism of atherosclerosis caused by P. gingivalis. It sorts out the interaction between P. gingivalis and AS-related cells, which provides a new perspective for us to prevent or slow down the occurrence and development of AS by inhibiting periodontal pathogens.
Collapse
Affiliation(s)
- Jiaqi Zhang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Mengru Xie
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Xiaofei Huang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Guangjin Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Ying Yin
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Xiaofeng Lu
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Guangxia Feng
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Ran Yu
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| |
Collapse
|
157
|
Yao Y, Li F, Zhang M, Jin L, Xie P, Liu D, Zhang J, Hu X, Lv F, Shang H, Zheng W, Sun X, Duanmu J, Wu F, Lan F, Xiao RP, Zhang Y. Targeting CaMKII-δ9 Ameliorates Cardiac Ischemia/Reperfusion Injury by Inhibiting Myocardial Inflammation. Circ Res 2022; 130:887-903. [PMID: 35152717 DOI: 10.1161/circresaha.121.319478] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND CaMKII (Ca2+/calmodulin-dependent kinase II) plays a central role in cardiac ischemia/reperfusion (I/R) injury-an important therapeutic target for ischemic heart disease. In the heart, CaMKII-δ is the predominant isoform and further alternatively spliced into 11 variants. In humans, CaMKII-δ9 and CaMKII-δ3, the major cardiac splice variants, inversely regulate cardiomyocyte viability with the former pro-death and the latter pro-survival. However, it is unknown whether specific inhibition of the detrimental CaMKII-δ9 prevents cardiac I/R injury and, if so, what is the underlying mechanism. Here, we aim to investigate the cardioprotective effect of specific CaMKII-δ9 inhibition against myocardial I/R damage and determine the underlying mechanisms. METHODS The role and mechanism of CaMKII-δ9 in cardiac I/R injury were investigated in mice in vivo, neonatal rat ventricular myocytes, and human embryonic stem cell-derived cardiomyocytes. RESULTS We demonstrate that CaMKII-δ9 inhibition with knockdown or knockout of its feature exon, exon 16, protects the heart against I/R-elicited injury and subsequent heart failure. I/R-induced cardiac inflammation was also ameliorated by CaMKII-δ9 inhibition, and compared with the previously well-studied CaMKII-δ2, CaMKII-δ9 overexpression caused more profound cardiac inflammation. Mechanistically, in addition to IKKβ (inhibitor of NF-κB [nuclear factor-κB] kinase subunit β), CaMKII-δ9, but not δ2, directly interacted with IκBα (NF-κB inhibitor α) with its feature exon 13-16-17 combination and increased IκBα phosphorylation and consequently elicited more pronounced activation of NF-κB signaling and inflammatory response. Furthermore, the essential role of CaMKII-δ9 in myocardial inflammation and damage was confirmed in human cardiomyocytes. CONCLUSIONS We not only identified CaMKII-δ9-IKK/IκB-NF-κB signaling as a new regulator of human cardiomyocyte inflammation but also demonstrated that specifically targeting CaMKII-δ9, the most abundant CaMKII-δ splice variant in human heart, markedly suppresses I/R-induced cardiac NF-κB activation, inflammation, and injury and subsequently ameliorates myocardial remodeling and heart failure, providing a novel therapeutic strategy for various ischemic heart diseases.
Collapse
Affiliation(s)
- Yuan Yao
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China. (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.)
| | - Fan Li
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China. (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.)
| | - Mao Zhang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China. (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.)
| | - Li Jin
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China. (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.)
| | - Peng Xie
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China. (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.)
| | - Dairu Liu
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China. (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.)
| | - Junxia Zhang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China. (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.)
| | - Xinli Hu
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China. (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.)
| | - Fengxiang Lv
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China. (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.)
| | - Haibao Shang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China. (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.)
| | - Wen Zheng
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China. (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.)
| | - Xueting Sun
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China. (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.)
| | - Jiaxin Duanmu
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, School of Basic Medical Sciences, Ministry of Education, Peking University Health Science Center, Beijing, China (J.D., Y.Z.)
| | - Fujian Wu
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (F.W., F. Lan)
| | - Feng Lan
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (F.W., F. Lan)
| | - Rui-Ping Xiao
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China. (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.).,Beijing City Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing, China. (R.-P.X.).,Peking-Tsinghua Center for Life Sciences, Beijing, China (R.-P.X.).,PKU-Nanjing Institute of Translational Medicine, China (R.-P.X.)
| | - Yan Zhang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China. (Y.Y., F. Li, M.Z., L.J., P.X., D.L., J.Z., X.H., F. Lv, H.S., W.Z., X.S., R.-P.X., Y.Z.).,Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, School of Basic Medical Sciences, Ministry of Education, Peking University Health Science Center, Beijing, China (J.D., Y.Z.)
| |
Collapse
|
158
|
Lin PC, Chen CY, Wu C, Su TC. Synergistic Effects of Inflammation and Atherogenic Dyslipidemia on Subclinical Carotid Atherosclerosis Assessed by Ultrasound in Patients with Familial Hypercholesterolemia and Their Family Members. Biomedicines 2022; 10:biomedicines10020367. [PMID: 35203576 PMCID: PMC8962410 DOI: 10.3390/biomedicines10020367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 11/23/2022] Open
Abstract
Low-density lipoprotein cholesterol (LDL-C) and total to high-density lipoprotein cholesterol (TC/HDL-C) ratio are both common risk factors for atherosclerotic cardiovascular diseases (ASCVDs). However, whether high-sensitivity C-reactive protein (hsCRP) has synergistic or attenuated effects on atherogenic dyslipidemia remains unclear. We investigated subclinical carotid atherosclerosis in patients with familial hypercholesterolemia (FH) and their family members. A total of 100 families with 761 participants were prospectively studied. Participants were categorized into four groups according to atherogenic dyslipidemia and inflammatory biomarkers. The group with LDL-C ≥ 160 mg/dL (or TC/HDL-C ratio ≥ 5) combined with hsCRP ≥ 2 mg/L have a thicker carotid intima-media thickness (CIMT) in different common carotid artery (CCA) areas and a higher percentage of high plaque scores compared with other subgroups. Multivariate logistic regression analysis revealed a significantly higher adjusted odds ratio (aOR) for thicker CIMT of 3.56 (95% CI: 1.56–8.16) was noted in those with concurrent LDL-C ≥ 160 mg/dL and hsCRP ≥ 2 mg/L compared with the group with concurrent LDL-C < 160 mg/dL and hsCRP < 2 mg/L. Our results demonstrated that systemic inflammation, in terms of higher hsCRP levels ≥ 2 mg/L, synergistically contributed to atherogenic dyslipidemia of higher LDL-C or a higher TC/HDL-C ratio on subclinical atherosclerosis.
Collapse
Affiliation(s)
- Po-Chih Lin
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan;
| | - Chung-Yen Chen
- Department of Environmental and Occupational Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan;
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei 100225, Taiwan
| | - Charlene Wu
- Global Health Program, College of Public Health, National Taiwan University, Taipei 10055, Taiwan;
| | - Ta-Chen Su
- Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan;
- Department of Environmental and Occupational Medicine, National Taiwan University Hospital, Taipei 100225, Taiwan;
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, Taipei 100225, Taiwan
- The Experimental Forest, National Taiwan University, Nantou 557009, Taiwan
- Correspondence: ; Tel.: +886-2-2312-3456 (ext. 66719 or 67183); Fax: +886-2-2371-2361
| |
Collapse
|
159
|
Wei KC, Sy CL, Wang WH, Wu CL, Chang SH, Huang YT. Major acute cardiovascular events after dengue infection-A population-based observational study. PLoS Negl Trop Dis 2022; 16:e0010134. [PMID: 35130277 PMCID: PMC8853534 DOI: 10.1371/journal.pntd.0010134] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 02/17/2022] [Accepted: 01/03/2022] [Indexed: 11/19/2022] Open
Abstract
Background Dengue virus (DENV) infection may be associated with increased risks of major adverse cardiovascular effect (MACE), but a large-scale study evaluating the association between DENV infection and MACEs is still lacking. Methods and findings All laboratory confirmed dengue cases in Taiwan during 2009 and 2015 were included by CDC notifiable database. The self-controlled case-series design was used to evaluate the association between DENV infection and MACE (including acute myocardial infarction [AMI], heart failure and stroke). The "risk interval" was defined as the first 7 days after the diagnosis of DENV infection and the "control interval" as 1 year before and 1 year after the risk interval. The incidence rate ratio (IRR) and 95% confidence interval (CI) for MACE were estimated by conditional Poisson regression. Finally, the primary outcome of the incidence of MACEs within one year of dengue was observed in 1,247 patients. The IRR of MACEs was 17.9 (95% CI 15.80–20.37) during the first week after the onset of DENV infection observed from 1,244 eligible patients. IRR were significantly higher for hemorrhagic stroke (10.9, 95% CI 6.80–17.49), ischemic stroke (15.56, 95% CI 12.44–19.47), AMI (13.53, 95% CI 10.13–18.06), and heart failure (27.24, 95% CI 22.67–32.73). No increased IRR was observed after day 14. Conclusions The risks for MACEs are significantly higher in the immediate time period after dengue infection. Since dengue infection is potentially preventable by early recognition and vaccination, the dengue-associated MACE should be taken into consideration when making public health management policies. Dengue infection is the most rapidly spreading mosquito-borne viral illness worldwide and becomes a vivid threat to non-tropical countries. Previous research has documented that viral infection can increase risks of adverse cardiovascular events. There were sporadic reports about the association of cardiovascular events and dengue infection in the endemic tropical countries. Our study analyzed the risks for major adverse cardiovascular events, and found that acute myocardial infarction, stroke and heart failure were significantly higher in the immediate time period (within one week) after dengue infection, especially in patients with ≥60 years of age, female gender and severe admission dengue cases.
Collapse
Affiliation(s)
- Kai-Che Wei
- Department of Dermatology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Cheng-Len Sy
- Department of Internal Medicine, Division of Infectious Diseases, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Wen-Hwa Wang
- Department of Internal Medicine, Division of Cardiology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Health Management Center, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- College of Management, I-Shou University, Kaohsiung, Taiwan
| | - Chia-Ling Wu
- Department of Medical Research and Development, Center for Big Data Analytics and Statistics, Chang Gung Memorial Hospital Linkou Main Branch, Taoyuan, Taiwan
| | - Shang-Hung Chang
- Department of Medical Research and Development, Center for Big Data Analytics and Statistics, Chang Gung Memorial Hospital Linkou Main Branch, Taoyuan, Taiwan
- Department of Internal Medicine, Division of Cardiology, Chang Gung Memorial Hospital Linkou Main Branch, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- * E-mail: (SH-C); (YT-H)
| | - Yu-Tung Huang
- Department of Medical Research and Development, Center for Big Data Analytics and Statistics, Chang Gung Memorial Hospital Linkou Main Branch, Taoyuan, Taiwan
- * E-mail: (SH-C); (YT-H)
| |
Collapse
|
160
|
Bytyçi I, Bajraktari G, Penson PE, Henein MY, Banach M. Efficacy and safety of colchicine in patients with coronary artery disease: A systematic review and meta-analysis of randomized controlled trials. Br J Clin Pharmacol 2022; 88:1520-1528. [PMID: 34409634 DOI: 10.1111/bcp.15041] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 07/05/2021] [Accepted: 07/24/2021] [Indexed: 02/05/2023] Open
Abstract
AIMS Inflammation plays a central role in the pathogenesis and clinical manifestations of atherosclerosis. Randomized controlled trials have investigated the potential benefit of colchicine in reducing cardiovascular (CV) events in patients with coronary artery disease (CAD) but produced conflicting results. The aim of this meta-analysis was to evaluate the efficacy and safety of colchicine in patients with CAD. METHODS We systematically searched selected electronic databases from inception until 10 December 2020. Primary clinical endpoints were: major adverse cardiac events; all-cause mortality; CV mortality; recurrent myocardial infarction; stroke; hospitalization; and adverse medication effects. Secondary endpoints were short-term effect of colchicine on inflammatory markers. RESULTS Twelve randomized controlled trials with a total of 13 073 patients with CAD (colchicine n = 6351 and placebo n = 6722) were included in the meta-analysis. At mean follow-up of 22.5 months, the colchicine group had lower risk of major adverse cardiac events (6.20 vs. 8.87%; P < .001), recurrent myocardial infarction (3.41 vs. 4.41%; P = .005), stroke (0.40 vs. 0.90%; P = .002) and hospitalization due to CV events (0.90 vs. 2.87%; P = .02) compared to the control group. The 2 patient groups had similar risk for all-cause mortality (2.08 vs. 1.88%; P = .82) and CV mortality (0.71 vs. 1.01%; P = .38). Colchicine significantly reduced high-sensitivity C-reactive protein (-4.25, P = .001) compared to controls but did not significantly affect interleukin (IL)-β1 and IL-18 levels. CONCLUSION Colchicine reduced CV events and inflammatory markers, high-sensitivity C-reactive protein and IL-6, in patients with coronary disease compared to controls. Its impact on cardiovascular and all-cause mortality requires further investigation.
Collapse
Affiliation(s)
- Ibadete Bytyçi
- Clinic of Cardiology, University Clinical Centre of Kosovo, Prishtina, Kosovo
- Department of Public Health and Clinical Medicine, Umeå University and Heart Centre, Umeå, Sweden
| | - Gani Bajraktari
- Clinic of Cardiology, University Clinical Centre of Kosovo, Prishtina, Kosovo
- Department of Public Health and Clinical Medicine, Umeå University and Heart Centre, Umeå, Sweden
- Medical Faculty, University of Prishtina, Prishtina, Kosovo
| | - Peter E Penson
- Liverpool Centre for Cardiovascular Science, University of Liverpool & Liverpool Heart & Chest Hospital, Liverpool, UK
- School of Pharmacy & Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Michael Y Henein
- Department of Public Health and Clinical Medicine, Umeå University and Heart Centre, Umeå, Sweden
- Molecular & Clinical Sciences Research Institute, St George University London, UK
- Brunel University, Middlesex, UK
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz, Poland
- Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
- Cardiovascular Research Centre, University of Zielona Gora, Zielona Gora, Poland
| | | | | |
Collapse
|
161
|
Inflammation as a mechanism and therapeutic target in peripheral artery disease. Can J Cardiol 2022; 38:588-600. [PMID: 35114347 DOI: 10.1016/j.cjca.2022.01.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 12/24/2022] Open
Abstract
Peripheral artery disease is one of three major clinical manifestations of atherosclerosis, the other two being coronary artery and cerebrovascular disease. Despite progress in surgery, antithrombotic therapy and therapies that modify conventional risk factors (lipid-, blood pressure-, and glucose-lowering interventions), patients with peripheral artery disease have unacceptably high risk of vascular complications. Additional strategies to reduce this residual risk are needed. The accumulated evidence that inflammation plays an important role in the pathogenesis of atherosclerosis has spurred recent efforts to evaluate anti-inflammatory agents as an additional therapeutic approach for atherothrombosis prevention and treatment. In this review, we examine the evidence supporting the role of inflammation in atherosclerosis, review recent trials evaluating anti-inflammatory approaches to reduce cardiovascular complications, and offer insights into the opportunities for novel anti-inflammatory strategies to reduce the burden of cardiovascular and limb complications in patients with peripheral artery disease.
Collapse
|
162
|
The Role of the VEGF Family in Atherosclerosis Development and Its Potential as Treatment Targets. Int J Mol Sci 2022; 23:ijms23020931. [PMID: 35055117 PMCID: PMC8781560 DOI: 10.3390/ijms23020931] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/09/2022] [Accepted: 01/14/2022] [Indexed: 02/07/2023] Open
Abstract
The vascular endothelial growth factor (VEGF) family, the crucial regulator of angiogenesis, lymphangiogenesis, lipid metabolism and inflammation, is involved in the development of atherosclerosis and further CVDs (cardiovascular diseases). This review discusses the general regulation and functions of VEGFs, their role in lipid metabolism and atherosclerosis development and progression. These functions present the great potential of applying the VEGF family as a target in the treatment of atherosclerosis and related CVDs. In addition, we discuss several modern anti-atherosclerosis VEGFs-targeted experimental procedures, drugs and natural compounds, which could significantly improve the efficiency of atherosclerosis and related CVDs' treatment.
Collapse
|
163
|
Kurian C, Mayer S, Kaur G, Sahni R, Feldstein E, Samaan M, Viswanathan D, Sami T, Ali S, Al-Shammari H, Bloomfield J, Bravo M, Nuoman R, Gulko E, Gandhi C, Al-Mufti F. Bihemispheric ischemic strokes in patients with COVID-19. Brain Circ 2022; 8:10-16. [PMID: 35372732 PMCID: PMC8973449 DOI: 10.4103/bc.bc_65_21] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/29/2021] [Accepted: 02/06/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND: There is emerging evidence that COVID-19 can trigger thrombosis because of a hypercoagulable state, including large-vessel occlusion ischemic strokes. Bihemispheric ischemic stroke is uncommon and is thought to indicate an embolic source. Here, we examine the findings and outcomes of patients with bihemispheric stroke in the setting of COVID-19. METHODS: We performed a retrospective cohort study at a quaternary academic medical center between March 1, 2020, and April 30, 2020. We identified all patients with laboratory-confirmed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection who presented with simultaneous bihemispheric ischemic strokes. RESULTS: Of 637 COVID-19 admissions during the 2-month period, 13 had a diagnosis of acute ischemic stroke, including 5 who developed bihemispheric cerebral infarction. Three of those 5 (60%) were female, median age was 54 (range 41–67), and all five were being managed for severe COVID-19-related pneumonia complicated by acute kidney injury and liver failure before the diagnosis of cerebral infarction was established. Five presented with elevated ferritin, lactate dehydrogenase, and interleukin-6 (IL-6) levels, and four had lymphopenia and elevated D-dimer levels. All patients underwent neuroimaging with computed tomography for persistent depressed mentation, with or without a focal neurologic deficit, demonstrating multifocal ischemic strokes with bihemispheric involvement. Outcome was poor in all patients: two were discharged to a rehabilitation facility with moderate-to-severe disability and three (60%) patients died. CONCLUSIONS: Stroke is implicated in SARS-CoV-2 infection. Although causality cannot be established, we present the imaging and clinical findings of patients with COVID-19 and simultaneous bihemispheric ischemic strokes. Multifocal ischemic strokes with bihemispheric involvement should be considered in COVID-19 patients with severe infection and poor neurologic status and may be associated with poor outcomes.
Collapse
|
164
|
Finegood ED, Miller GE. Childhood Violence Exposure, Inflammation, and Cardiometabolic Health. Curr Top Behav Neurosci 2022; 54:439-459. [PMID: 34935115 DOI: 10.1007/7854_2021_283] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Exposure to interpersonal violence during childhood, a severe and often traumatic form of social stress, is an enduring problem that an emerging body of work suggests may be relevant to cardiometabolic health and the progression of cardiovascular disease (CVD) across the life course. Less is known about this association causally, and consequently, the biological mechanisms that may confer risk for, and resilience to, poor health outcomes in the aftermath of violence are not well understood. Drawing on recent theoretical insights and empirical research in both humans and non-human animal models, the current paper articulates a hypothesis for one way that childhood violence could get "under the skin" to influence CVD. Based on this emerging literature, one plausible way that childhood violence exposure could increase susceptibility to CVD in later life is by sensitizing stress-response neurobiology and immune processes that regulate and promote inflammation, which is a key pathogenic mechanism in CVD. This is inherently a developmental process that begins in early life and that unfolds across the life course, although less is known about the specific mechanisms through which this occurs. The goal of this paper is to articulate some of these plausible mechanisms and to suggest areas for future research that aims to reduce the burden of disease among individuals who are exposed to violence.
Collapse
Affiliation(s)
- Eric D Finegood
- Department of Psychology, Institute for Policy Research, Northwestern University, Evanston, IL, USA.
| | - Gregory E Miller
- Department of Psychology, Institute for Policy Research, Northwestern University, Evanston, IL, USA
| |
Collapse
|
165
|
Shinge SAU, Zhang D, Din AU, Yu F, Nie Y. Emerging Piezo1 signaling in inflammation and atherosclerosis; a potential therapeutic target. Int J Biol Sci 2022; 18:923-941. [PMID: 35173527 PMCID: PMC8771847 DOI: 10.7150/ijbs.63819] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 11/08/2021] [Indexed: 11/24/2022] Open
Abstract
Purpose of Review: Atherosclerosis is the principal cause of cardiovascular diseases (CVDs) which are the major cause of death worldwide. Mechanical force plays an essential role in cardiovascular health and disease. To bring the awareness of mechanosensitive Piezo1 role in atherosclerosis and its therapeutic potentials we review recent literature to highlight its involvement in various mechanisms of the disease. Recent Findings: Recent studies reported Piezo1 channel as a sensor, and transducer of various mechanical forces into biochemical signals, which affect various cellular activities such as proliferation, migration, apoptosis and vascular remodeling including immune/inflammatory mechanisms fundamental phenomenon in atherogenesis. Summary: Numerous evidences suggest Piezo1 as a player in different mechanisms of cell biology, including immune/inflammatory and other cellular mechanisms correlated with atherosclerosis. This review discusses mechanistic insight about this matter and highlights the drugability and therapeutic potentials consistent with emerging functions Piezo1 in various mechanisms of atherosclerosis. Based on the recent works, we suggest Piezo1 as potential therapeutic target and a valid candidate for future research. Therefore, a deeper exploration of Piezo1 biology and translation towards the clinic will be a novel strategy for treating atherosclerosis and other CVDs.
Collapse
Affiliation(s)
- Shafiu A. Umar Shinge
- Cardiovascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan PRC
| | - Daifang Zhang
- Cardiovascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan PRC
- Clinical Research Center, Southwest Medical University, Luzhou, Sichuan PRC
| | - Ahmad Ud Din
- Drug Discovery Research Center, Southwest Medical University, Luzhou, Sichuan PRC
| | - FengXu Yu
- Cardiovascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan PRC
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, Sichuan PRC
| | - YongMei Nie
- Cardiovascular Surgery Department, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan PRC
- Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Southwest Medical University, Luzhou, Sichuan PRC
| |
Collapse
|
166
|
Yu X, Xu X, Wu T, Huang W, Xu C, Xie W, Long X. APOA1 Level is Negatively Correlated with the Severity of COVID-19. Int J Gen Med 2022; 15:689-698. [PMID: 35082518 PMCID: PMC8785137 DOI: 10.2147/ijgm.s332956] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 11/01/2021] [Indexed: 01/08/2023] Open
Abstract
Objective We used public data to analyze the proteomics, metabolomics and transcriptomics characteristics of COVID-19 patients to identify potential therapeutic targets. More importantly, we also collected clinical data for verification to make the analysis results more reliable. Methods Download the serum proteomics and metabolomics data of COVID-19 patients and describe their changes in serum proteins and metabolites, and use bioinformatics analysis methods to identify potential biomarkers and therapeutic targets. Finally, clinical data and experimental data of cell infection were combined for verification. Results It was found that the serum apolipoprotein A1 (APOA1) protein level in COVID-19 patients was down-regulated (log2FC = −0.39, false discovery rate (FDR) < 0.001), and the degree of reduction in the severe group was more significant (kruskal-test p = 2.5e-05). What is more, APOA1 was not only expressed lower in male patients (Wilcox-test p = 0.012), but also negatively correlated with C-reactive protein (CRP, r = −0.37, p = 0.019). The experiment data from SARS-CoV-2 infected cells further showed that the protein and transcript level of APOA1 gradually decreased as the infection time increased, and the transcription level (log2FC = −8.3, FDR = 0.0015) was more down-regulated than protein level (log2FC = −0.95, FDR = 0.0014). More importantly, the collected clinical data also confirmed that APOA1 was down-regulated in COVID-19 patients (kruskal-test p = 0.001), and APOA1 levels are negatively correlated with IL6 (r = −0.396, p = 2.22e-07), D-dimers (DD, r = −0.262, p = 8.19e-04), prothrombin time (PT, r = −0.464, p = 6.68e-10) and thrombin time (TT, r = −0.279, p = 3.46e-04). Conclusion The degree of down-regulation of APOA1 is positively correlated with the severity of COVID-19, and the expression level of APOA1 is negatively correlated with CRP, IL6, DD, PT, TT, and positively correlated with HD and LDL. This indicates that APOA1 may be a key molecule in tandem acute inflammatory response, coagulation abnormalities and cholesterol metabolism disorder in COVID-19, and could be a potential therapeutic target.
Collapse
Affiliation(s)
- Xiaosi Yu
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
- Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xianqun Xu
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
| | - Tianpeng Wu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, People’s Republic of China
| | - Wenjie Huang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
| | - Chen Xu
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
| | - Wen Xie
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
| | - Xinghua Long
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, People’s Republic of China
- Correspondence: Xinghua Long; Wen Xie Email ;
| |
Collapse
|
167
|
Bian Q, Chen J, Weng Y, Li S. Endothelialization strategy of implant materials surface: The newest research in recent 5 years. J Appl Biomater Funct Mater 2022; 20:22808000221105332. [PMID: 35666145 DOI: 10.1177/22808000221105332] [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: 11/16/2022] Open
Abstract
In recent years, more and more metal or non-metal materials have been used in the treatment of cardiovascular diseases, but the vascular complications after transplantation are still the main factors restricting the clinical application of most grafts, such as acute thrombosis and graft restenosis. Implant materials have been extensively designed and surface optimized by researchers, but it is still too difficult to avoid complications. Natural vascular endodermis has excellent function, anti-coagulant and anti-intimal hyperplasia, and it is also the key to maintaining the homeostasis of normal vascular microenvironment. Therefore, how to promote the adhesion of endothelial cells (ECs) on the surface of cardiovascular materials to achieve endothelialization of the surface is the key to overcoming the complications after implant materialization. At present, the surface endothelialization design of materials based on materials surface science, bioactive molecules, and biological function intervention and feedback has attracted much attention. In this review, we summarize the related research on the surface modification of materials by endothelialization in recent years, and analyze the advantages and challenges of current endothelialization design ideas, explain the relationship between materials, cells, and vascular remodeling in order to find a more ideal endothelialization surface modification strategy for future researchers to meet the requirements of clinical biocompatibility of cardiovascular materials.
Collapse
Affiliation(s)
- Qihao Bian
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, China.,School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Junying Chen
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, China.,School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Yajun Weng
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, China.,School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Suiyan Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| |
Collapse
|
168
|
Mannarino MR, Bianconi V, Gigante B, Strawbridge RJ, Savonen K, Kurl S, Giral P, Smit A, Eriksson P, Tremoli E, Veglia F, Baldassarre D, Pirro M. Neutrophil to lymphocyte ratio is not related to carotid atherosclerosis progression and cardiovascular events in the primary prevention of cardiovascular disease: Results from the IMPROVE study. Biofactors 2022; 48:100-110. [PMID: 34761838 PMCID: PMC9299016 DOI: 10.1002/biof.1801] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 10/21/2021] [Indexed: 12/11/2022]
Abstract
Inflammation is a component of the pathogenesis of atherosclerosis and is associated with an increased risk of atherosclerotic cardiovascular disease (ASCVD). The neutrophil to lymphocyte ratio (NLR) is a possible inflammation metric for the detection of ASCVD risk, although results of prospective studies are highly inconsistent on this topic. We investigated the cross-sectional relationship between NLR and carotid intima-media thickness (cIMT) in subjects at moderate-to-high ASCVD risk. The prospective association between NLR, cIMT progression, and incident vascular events (VEs) was also explored. In 3341 subjects from the IMT-Progression as Predictors of VEs (IMPROVE) study, we analyzed the association between NLR, cIMT, and its 15-month progression. The association between NLR and incident VEs was also investigated. NLR was positively associated with cross-sectional measures of cIMT, but not with cIMT progression. The association between NLR and cross-sectional cIMT measures was abolished when adjusted for confounders. No association was found between NRL and incident VEs. Similarly, there were no significant differences in the hazard ratios (HRs) of VEs across NLR quartiles. NLR was neither associated with the presence and progression of carotid atherosclerosis, nor with the risk of VEs. Our findings do not support the role of NLR as a predictor of the risk of atherosclerosis progression and ASCVD events in subjects at moderate-to-high ASCVD risk, in primary prevention. However, the usefulness of NLR for patients at a different level of ASCVD risk cannot be inferred from this study.
Collapse
Affiliation(s)
- Massimo R. Mannarino
- Unit of Internal Medicine, Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | - Vanessa Bianconi
- Unit of Internal Medicine, Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | - Bruna Gigante
- Division of Cardiovascular Medicine, Department of Medicine, SolnaKarolinska InstitutetStockholmSweden
| | - Rona J. Strawbridge
- Division of Cardiovascular Medicine, Department of Medicine, SolnaKarolinska InstitutetStockholmSweden
- Institute of Health and WellbeingUniversity of GlasgowGlasgowUK
- Health Data Research UKGlasgowUK
| | - Kai Savonen
- Foundation for Research in Health Exercise and NutritionKuopio & Research Institute of Exercise MedicineKuopioFinland
- Department of Clinical Physiology and Nuclear MedicineKuopio University HospitalKuopioFinland
| | - Sudhir Kurl
- Institute of Public Health and Clinical NutritionUniversity of Eastern FinlandKuopioFinland
| | - Philippe Giral
- Assistance Publique ‐ Hopitaux de ParisParisFrance
- Service Endocrinologie‐Metabolisme, Groupe Hôspitalier Pitie‐SalpetriereUnités de Prévention CardiovasculaireParisFrance
| | - Andries Smit
- Department of MedicineUniversity Medical Center GroningenGroningenthe Netherlands
- Department of MedicineIsala Clinics ZwolleZwolleThe Netherlands
| | - Per Eriksson
- Division of Cardiovascular Medicine, Department of Medicine, SolnaKarolinska InstitutetStockholmSweden
| | | | | | - Damiano Baldassarre
- Centro Cardiologico Monzino, IRCCSMilanItaly
- Department of Medical Biotechnology and Translational MedicineUniversità degli Studi di MilanoMilanItaly
| | - Matteo Pirro
- Unit of Internal Medicine, Department of Medicine and SurgeryUniversity of PerugiaPerugiaItaly
| | | |
Collapse
|
169
|
Maluf CB, Brito FDA, Simões TC, Peixoto SV, Vidigal PG. Perfil laboratorial após rompimento de barragem de mineração: resultados do Projeto Saúde Brumadinho. REVISTA BRASILEIRA DE EPIDEMIOLOGIA 2022. [DOI: 10.1590/1980-549720220013.supl.2.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
RESUMO Objetivo: Avaliar alterações em parâmetros laboratoriais na população do Projeto Saúde Brumadinho, segundo exposição ao rompimento da barragem. Métodos: Estudo transversal realizado em amostra representativa de residentes (≥12 anos) em Brumadinho, Minas Gerais, incluindo: não expostos (grupo referência); diretamente atingidos pela lama de rejeitos; e residentes em área de mineração. Foram estimadas as prevalências de resultados alterados de hemograma, colesterol total, colesterol lipoproteína de alta densidade (HDL), colesterol lipoproteína de baixa densidade (LDL), triglicérides, aspartato aminotransferase, alanina aminotransferase, creatinina, ureia, estimativa da taxa de filtração glomerular (TFGe) e proteína C-reativa ultrassensível (PCRus). As razões de prevalência (RP) e os intervalos de confiança de 95% (IC95%) de ter o exame alterado foram estimados por meio de modelos lineares generalizados com distribuição de probabilidade Poisson. Estimaram-se modelos brutos e ajustados por faixa etária, sexo, diabetes, índice de massa corporal, tabagismo, hipertensão. Resultados: Após ajustes, não se observou diferença nas RP entre as populações estudadas para a maioria dos testes, com exceção da população residente em área com atividade de mineração e não diretamente atingida pela lama, com menor chance de ter colesterol total alterado (RP=0,84; IC95% 0,74–0,95) e maior chance de ter colesterol HDL (RP=1,26; IC95% 1,07–1,50) e PCRus (RP=1,19; IC95% 1,04–1,37) alterado e TFGe<60 mL/min/1,73 m2 (RP=1,51; IC95% 1,05–2,19). Conclusão: Não foram encontradas diferenças significativas na prevalência de alterações bioquímicas e hematológicas entre a população diretamente exposta aos rejeitos e a população não exposta. Apenas o grupo residente em área de mineração apresentou maior prevalência de alterações relacionadas com dislipidemia, disfunção renal e inflamação.
Collapse
|
170
|
Bhat RA, Maqbool S, Rathi A, Ali SM, Hussenbocus YAAM, Wentao X, Qu Y, Zhang Y, Sun Y, Fu HX, Wang LY, Dwivedi A, Bhat JA, Iqbal RS, Islam MM, Tibrewal A, Gao C. The Effects of the SARS-CoV-2 Virus on the Cardiovascular System and Coagulation State Leading to Cardiovascular Diseases: A Narrative Review. INQUIRY : A JOURNAL OF MEDICAL CARE ORGANIZATION, PROVISION AND FINANCING 2022; 59:469580221093442. [PMID: 35613600 PMCID: PMC9149622 DOI: 10.1177/00469580221093442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 03/15/2022] [Accepted: 03/21/2022] [Indexed: 01/08/2023]
Abstract
The novel coronavirus pandemic has led to morbidity and mortality throughout the world. Until now, it is a highly virulent contagion attacking the respiratory system in humans, especially people with chronic diseases and the elderly who are most vulnerable. A majority of afflicted are those suffering from cardiovascular and coronary diseases. In this review article, an attempt has been made to discuss and thoroughly review the mode of therapies that alleviate cardiac complications and complications due to hypercoagulation in patients infected with the SARS-CoV-2 virus. Presently a host of thrombolytic drugs are in use like Prourokinase, Retelapse, RhTNK-tPA and Urokinase. However, thrombolytic therapy, especially if given intravenously, is associated with a serious risk of intracranial haemorrhage, systemic haemorrhage, immunologic complications, hypotension and myocardial rupture. The effects of the SARS-CoV-2 virus upon the cardiovascular system and coagulation state of the body are being closely studied. In connection to the same, clinical prognosis and complications of thrombolytic therapy are being scrutinized. It is noteworthy to mention that myocardial oxygen supply/demand mismatch, direct myocardial cells injury and acute plaque rupture are the multiple mechanisms responsible for acute coronary syndrome and cardiac complications in Covid-19 infection. However, this review has limitations as data available in this context is limited, scattered and heterogenous that questions the reliability of the same. So, more multi-centric studies involving representative populations, carried out meticulously, could further assist in responding better to cardiac complications among Covid-19 patients.
Collapse
Affiliation(s)
- Rafiq A. Bhat
- Department of Interventional
Cardiology, Fuwai Central China Cardiovascular Hospital, Zhengzhou University, Zhengzhou, People’s Republic of China; Henan Provincial
People’s Hospital, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Syed Maqbool
- Department of Cardiology, Government Superspeciality
Hospital, Srinagar, India
| | - Akanksha Rathi
- Department of Community Medicine, Vedanta Institute of Medical
Sciences, Palghar, India
| | - Syed Manzoor Ali
- Department of Cardiology, Super
Speciality Division, Government Medical
College, Srinagar, India
| | | | - Xiao Wentao
- Department of Interventional
Cardiology, Fuwai Central China Cardiovascular Hospital, Zhengzhou University, Zhengzhou, People’s Republic of China; Henan Provincial
People’s Hospital, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Yongsheng Qu
- Department of Interventional
Cardiology, Fuwai Central China Cardiovascular Hospital, Zhengzhou University, Zhengzhou, People’s Republic of China; Henan Provincial
People’s Hospital, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - You Zhang
- Department of Interventional
Cardiology, Fuwai Central China Cardiovascular Hospital, Zhengzhou University, Zhengzhou, People’s Republic of China; Henan Provincial
People’s Hospital, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Yuxiao Sun
- Department of Interventional
Cardiology, Fuwai Central China Cardiovascular Hospital, Zhengzhou University, Zhengzhou, People’s Republic of China; Henan Provincial
People’s Hospital, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Hai-Xia Fu
- Department of Interventional
Cardiology, Fuwai Central China Cardiovascular Hospital, Zhengzhou University, Zhengzhou, People’s Republic of China; Henan Provincial
People’s Hospital, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Ling Yun Wang
- Department of Cardiology, The First Affiliated Hospital of
Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Atul Dwivedi
- Department of Clinical and Basic
Sciences, Medical School of Hubei Polytechnic
University, Hubei, China
| | - Javaid Akhter Bhat
- State Key Laboratory of Crop
Genetics and Germplasm Enhancement, Nanjing Agricultural
University, Nanjing, People’s Republic of China
| | - Raja saqib Iqbal
- Department of Paediatrics, Batra Hospital and Medical Research
Centre, New Delhi, India
| | - Md Monowarul Islam
- Department of Cardiology, The First Affiliated Hospital of
Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Abhishek Tibrewal
- Department of Community Medicine, Institute of Biostatistics and
Epidemiology, Gurgaon, India
| | - Chuanyu Gao
- Department of Interventional
Cardiology, Fuwai Central China Cardiovascular Hospital, Zhengzhou University, Zhengzhou, People’s Republic of China; Henan Provincial
People’s Hospital, Zhengzhou University, Zhengzhou, People’s Republic of China
- Henan Provincial Key Laboratory for
Control of Coronary Heart Disease, Zhengzhou, People’s Republic of China
| |
Collapse
|
171
|
Targeting Inflammation in the Diagnosis, Management, and Prevention of Cardiovascular Diseases. Glob Heart 2022; 17:80. [PMID: 36382160 PMCID: PMC9635324 DOI: 10.5334/gh.1156] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 09/01/2022] [Indexed: 01/25/2023] Open
Abstract
Inflammation plays an important role in the development and progression of cardiovascular diseases (CVDs). Hypertension and hyperlipidemia are the key risk factors of CVDs and induce inflammation in the heart and vessels. Unhealthy diet, infection, and smoking coupled with genetic factors lead to the development of CVDs as well as induce inflammation. Risk factors of CVDs such as hypertension and hyperlipidemia along with diabetes activate nuclear factor kappa B, which regulates the transcription of immunoglobulin free light chain (FLC) κ in B cells and the production of multiple inflammatory molecules, leading to inflammation. FLCs are novel biomarkers of inflammation, and their levels have been associated with overall mortality in a general population and with cardiovascular outcomes. In this review, the role of inflammation in the pathogenesis of CVDs, new biomarkers of inflammation, and dietary options counterbalancing inflammatory processes, such as the polyphenol-rich French maritime pine bark extract Pycnogenol, are discussed.
Collapse
|
172
|
Seki A, Fishbein MC. Age-related cardiovascular changes and diseases. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00004-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
173
|
De Novo Sphingolipid Biosynthesis in Atherosclerosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1372:31-46. [DOI: 10.1007/978-981-19-0394-6_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
174
|
Jafari-Oori M, Dehi M, Ebadi A, Moradian ST, jafari M. Incidence of cardiac complications following COVID-19 infection: An umbrella meta-analysis study. Heart Lung 2022; 52:136-145. [PMID: 35074740 PMCID: PMC8743575 DOI: 10.1016/j.hrtlng.2022.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 01/01/2022] [Accepted: 01/03/2022] [Indexed: 01/08/2023]
Abstract
Background COVID-19 causes fatal cardiac damages. Despite many overwhelming meta-analysis related to cardiac complications following COVID-19 disease, no umbrella meta-analysis study has been conducted. Objectives We aimed to report the summarized pooled incidences of cardiac complications in the overall, critically ill, and deceased patients, compare the cardiac complications between the severe/non-severe or deceased/non-deceased patients, and also compare poor outcomes between patients with/without acute myocardial injury (AMI). Methods PubMed, Scopus, web of science, Cochrane, ProQuest, Springer, Sage journals were searched before April 2021. After assessing the quality and duplicate data, data were run by the random/fixed-effect models, I2 heterogeneity index, Egger's test, and sensitivity analysis. Results After removing duplicate data, in the overall COVID-19 patients, the pooled incidence of AMI, heart failure, arrhythmia, cardiac arrest, and acute coronary syndrome (ACS) were 21%, 14%, 16%, 3.46%, and 1.3%, respectively. In the patients with severe disease, the pooled incidence of AMI and shock were 33 and 35%, respectively. Similarly, in the deceased COVID-19 patients, the pooled incidence rate of AMI and arrhythmia were 56% and 47.5%, respectively. The patients with severe disease were at higher risk of AMI (RR = 5.27) and shock (OR = 20.18) compared with the non-severe cases. Incidence of AMI was associated with transfer to the intensive care units (ICU) (RR = 2.92) and mortality (RR = 2.57, OR = 8.36), significantly. Conclusion Cardiac complications were found to be increased alarmingly in COVID-19 patients. Baseline and during hospitalization checking with electrocardiography, echocardiography, and measuring of cardiac biomarkers should be applied.
Collapse
|
175
|
El Hadri K, Smith R, Duplus E, El Amri C. Inflammation, Oxidative Stress, Senescence in Atherosclerosis: Thioredoxine-1 as an Emerging Therapeutic Target. Int J Mol Sci 2021; 23:ijms23010077. [PMID: 35008500 PMCID: PMC8744732 DOI: 10.3390/ijms23010077] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/19/2021] [Accepted: 12/19/2021] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is a leading cause of cardiovascular diseases (CVD) worldwide and intimately linked to aging. This pathology is characterized by chronic inflammation, oxidative stress, gradual accumulation of low-density lipoproteins (LDL) particles and fibrous elements in focal areas of large and medium arteries. These fibrofatty lesions in the artery wall become progressively unstable and thrombogenic leading to heart attack, stroke or other severe heart ischemic syndromes. Elevated blood levels of LDL are major triggering events for atherosclerosis. A cascade of molecular and cellular events results in the atherosclerotic plaque formation, evolution, and rupture. Moreover, the senescence of multiple cell types present in the vasculature were reported to contribute to atherosclerotic plaque progression and destabilization. Classical therapeutic interventions consist of lipid-lowering drugs, anti-inflammatory and life style dispositions. Moreover, targeting oxidative stress by developing innovative antioxidant agents or boosting antioxidant systems is also a well-established strategy. Accumulation of senescent cells (SC) is also another important feature of atherosclerosis and was detected in various models. Hence, targeting SCs appears as an emerging therapeutic option, since senolytic agents favorably disturb atherosclerotic plaques. In this review, we propose a survey of the impact of inflammation, oxidative stress, and senescence in atherosclerosis; and the emerging therapeutic options, including thioredoxin-based approaches such as anti-oxidant, anti-inflammatory, and anti-atherogenic strategy with promising potential of senomodulation.
Collapse
|
176
|
Cwajda-Białasik J, Mościcka P, Jawień A, Szewczyk MT. Microbiological Status of Venous Leg Ulcers and Its Predictors: A Single-Center Cross-Sectional Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182412965. [PMID: 34948575 PMCID: PMC8700924 DOI: 10.3390/ijerph182412965] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/04/2021] [Accepted: 12/06/2021] [Indexed: 11/16/2022]
Abstract
Venous leg ulcers are frequently colonized by microbes. This can be particularly devastating if the ulcer is infected with alert pathogens, i.e., highly virulent microorganisms with well-developed mechanisms of antibiotic resistance. We analyzed the microbiological status of venous leg ulcers and identified the clinicodemographic predictors of culture-positive ulcers, especially in ulcers with colonization by alert pathogens. METHODS This study included 754 patients with chronic venous leg ulcers. Material for microbiological analysis was collected by swabbing only from patients who did not receive any antibiotic treatment. RESULTS A total of 636 (84.3%) patients presented with culture-positive ulcers. Alert pathogens, primarily Pseudomonas aeruginosa, were detected in 28.6% of the positive cultures. In a logistic regression model, culture-positive ulcers were predicted independently by age > 65 years, current ulcer duration > 12 months, and ulceration area greater than 8.25 cm2. Two of these factors, duration of current ulcer > 12 months and ulceration area > 8.25 cm2, were also identified as the independent predictors of colonization by alert pathogens. CONCLUSIONS Colonization/infection is particularly likely in older persons with chronic and/or large ulcers. Concomitant atherosclerosis was an independent predictor of culture-negative ulcers.
Collapse
Affiliation(s)
- Justyna Cwajda-Białasik
- Department of Perioperative Nursing, Department of Surgical Nursing and Chronic Wound Care, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-821 Bydgoszcz, Poland; (P.M.); (M.T.S.)
- Outpatient Department for Chronic Wound Management, Antoni Jurasz University Hospital No. 1, 85-094 Bydgoszcz, Poland
- Correspondence:
| | - Paulina Mościcka
- Department of Perioperative Nursing, Department of Surgical Nursing and Chronic Wound Care, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-821 Bydgoszcz, Poland; (P.M.); (M.T.S.)
- Outpatient Department for Chronic Wound Management, Antoni Jurasz University Hospital No. 1, 85-094 Bydgoszcz, Poland
| | - Arkadiusz Jawień
- Department of Vascular Surgery and Angiology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-094 Bydgoszcz, Poland;
| | - Maria Teresa Szewczyk
- Department of Perioperative Nursing, Department of Surgical Nursing and Chronic Wound Care, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-821 Bydgoszcz, Poland; (P.M.); (M.T.S.)
- Outpatient Department for Chronic Wound Management, Antoni Jurasz University Hospital No. 1, 85-094 Bydgoszcz, Poland
| |
Collapse
|
177
|
Zhang X, Zhang H, Yang X, Qin Q, Sun X, Hou Y, Chen D, Jia M, Su X, Chen Y. Angiotensin II upregulates endothelin receptors through the adenosine monophosphate-activated protein kinase/sirtuin 1 pathway in vascular smooth muscle cells. J Pharm Pharmacol 2021; 73:1652-1662. [PMID: 34570873 DOI: 10.1093/jpp/rgab137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 08/26/2021] [Indexed: 11/13/2022]
Abstract
OBJECTIVES This study was designed to test our hypothesis that angiotensin II (Ang II) upregulates endothelin (ET) receptors in vascular smooth muscle cells (VSMCs). METHODS Rat superior mesenteric artery (SMA) without endothelium was cultured in serum-free medium for 24 h in the presence of Ang II with or without metformin or nicotinamide. In vivo, rats were implanted subcutaneously with a mini-osmotic pump infusing AngII (500 ng/kg/min) for 4 weeks. The level of protein expression was determined using Western blotting. The contractile response to ET receptor agonists was studied using sensitive myography. Caudal artery blood pressure (BP) was measured using non-invasive tail-cuff plethysmography. KEY FINDINGS The results showed that Ang II significantly increased ET receptors and decreased phosphorylated-adenosine monophosphate-activated protein kinase α (p-AMPKα) in SMA. Furthermore, metformin significantly inhibited Ang II-upregulated ET receptors and upregulated Ang II-decreased sirtuin 1 (Sirt1). However, this effect was reversed by nicotinamide. Moreover, the in-vivo results showed that metformin not only inhibited Ang II-induced upregulation of ET receptors but also recovered Ang II-decreased p-AMPKα and Sirt1. In addition, metformin significantly inhibited Ang II-elevated BP. However, the effect was reversed by nicotinamide, except for p-AMPKα. CONCLUSIONS Ang II upregulated ET receptors in VSMCs to elevate BP by inhibiting AMPK, thereby inhibiting Sirt1.
Collapse
Affiliation(s)
- Xin Zhang
- Institute of Basic and Translational Medicine, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Hongmei Zhang
- The First Affiliated Hospital of Xi'an Medical University, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Xinpu Yang
- Institute of Basic and Translational Medicine, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Qiaohong Qin
- Institute of Basic and Translational Medicine, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Xia Sun
- School of Basic and Medical Sciences, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Ying Hou
- Institute of Basic and Translational Medicine, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Di Chen
- School of Basic and Medical Sciences, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Min Jia
- Institute of Basic and Translational Medicine, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Xingli Su
- Institute of Basic and Translational Medicine, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi, China
- School of Basic and Medical Sciences, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Yulong Chen
- Institute of Basic and Translational Medicine, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
178
|
Vilela EM, Fontes-Carvalho R. Inflammation and ischemic heart disease: The next therapeutic target? Rev Port Cardiol 2021; 40:785-796. [PMID: 34857118 DOI: 10.1016/j.repce.2021.10.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 02/16/2021] [Indexed: 12/13/2022] Open
Abstract
Inflammation plays an important role in several stages of the cardiovascular continuum. In recent decades a plethora of studies have provided new data highlighting the role of inflammation in atherogenesis and atherothrombosis in two-way interactions with various cardiovascular risk factors and further influencing these dynamic processes. The concept of targeting residual inflammatory risk among individuals with ischemic heart disease (IHD) is therefore gaining increasing attention. Recently, several landmark randomized controlled trials have assessed different pharmacological approaches that may mitigate this residual risk. The results of some of these studies, such as CANTOS with canakinumab and COLCOT and LoDoCo2 with colchicine, are promising and have provided data to support this concept. Moreover, though several aspects remain to be clarified, these trials have shown the potential of modulating inflammation as a new target to reduce the risk of cardiovascular events in secondary prevention patients. In the present review, we aim to present a pragmatic overview of the complex interplay between inflammation and IHD, and to critically appraise the current evidence on this issue while presenting future perspectives on this topic of pivotal contemporary interest.
Collapse
Affiliation(s)
- Eduardo M Vilela
- Cardiology Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal.
| | - Ricardo Fontes-Carvalho
- Cardiology Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal; Cardiovascular Research Center (UniC), Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
179
|
Currò CT, Cotroneo M, Ciacciarelli A, Dell'Aera C, Grillo F, La Spina P, Fazio MC, Laganà A, De Caro J, Trimarchi G, Toscano A, Musolino RF, Casella C. Ischemic Stroke and Asymptomatic Pulmonary Opacities. J Stroke Cerebrovasc Dis 2021; 31:106230. [PMID: 34864609 DOI: 10.1016/j.jstrokecerebrovasdis.2021.106230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/13/2021] [Indexed: 11/18/2022] Open
Abstract
PURPOSE Pulmonary opacities are described in many chest radiograph reports in stroke patients and are often asymptomatic. The aim of this study is to investigate the clinical features associated with asymptomatic pulmonary opacities (APO) and evaluate APO prognostic value in ischemic stroke. METHODS We prospectively analyzed patients with acute ischemic stroke without pneumonia symptoms referred to our stroke center. All patients underwent a chest x-ray within 48 hours after admission. We divided patients with APO (defined as asymptomatic area of increased pulmonary attenuation) and patients without APO. Firstly, we compared clinical, laboratory, and echocardiographic parameters between groups; secondly, APO were evaluated as a possible predictor of the neurological severity at discharge and functional outcome at 90 days. RESULTS We included 162 patients, 78 of whom had APO. On univariate analysis, we observed a significant difference between patients with APO and patients without APO in terms of neutrophils (73,56±10,10 vs 69,72±12,29; p=0,031), atrial fibrillation (AF, paroxysmal 27,4% vs 25,6%; permanent 19% vs 2,6%; p<0,002), cardioembolic etiology (37,2% vs 22,7%, p <0,033), thrombectomy (58,3% vs 42,9%, p <0,05), dysphagia/vomit (34,5% vs 14,10%, p<0,005), admission NIHSS (14,20±5,98 vs 10,29±5,82, p=0,001), discharge NIHSS (8,68±6,73 vs 5,64±6,20; p=0,003), and 90 days mRS (3,47±2,12 vs 2,31±1,93; p=0,001). On multivariate analysis, APO were significantly associated with admission NIHSS and AF. APO were a predictor of outcome on univariate analysis, but not on multivariate. CONCLUSION APO in acute ischemic stroke are associated with AF. APO were not a predictor of neurological and functional outcome.
Collapse
Affiliation(s)
- Carmelo Tiberio Currò
- Stroke Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy.
| | - Masina Cotroneo
- Stroke Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Antonio Ciacciarelli
- Stroke Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Cristina Dell'Aera
- Stroke Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Francesco Grillo
- Stroke Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Paolino La Spina
- Stroke Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Maria Carolina Fazio
- Stroke Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Angelina Laganà
- Stroke Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Jolanda De Caro
- Stroke Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | | | - Antonio Toscano
- Stroke Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Rosa Fortunata Musolino
- Stroke Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Carmela Casella
- Stroke Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| |
Collapse
|
180
|
Tóth E, Beinrohr L, Gubucz I, Szabó L, Tenekedjiev K, Nikolova N, Nagy AI, Hidi L, Sótonyi P, Szikora I, Merkely B, Kolev K. Fibrin to von Willebrand factor ratio in arterial thrombi is associated with plasma levels of inflammatory biomarkers and local abundance of extracellular DNA. Thromb Res 2021; 209:8-15. [PMID: 34844046 DOI: 10.1016/j.thromres.2021.11.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/02/2021] [Accepted: 11/13/2021] [Indexed: 01/02/2023]
Abstract
INTRODUCTION The composition of thrombi determines their structure, mechanical stability, susceptibility to lysis, and consequently, the clinical outcome in coronary artery disease (CAD), acute ischemic stroke (AIS), and peripheral artery disease (PAD). Fibrin forms the primary matrix of thrombi intertwined with DNA, derived from neutrophil extracellular traps (NETs), and von Willebrand factor (VWF) bridging DNA and platelets. Here we examined the relative content of fibrin, DNA and VWF in thrombi and analyzed their interrelations and quantitative associations with systemic biomarkers of inflammation and clinical characteristics of the patients. PATIENTS, METHODS Thrombi extracted from AIS (n = 17), CAD (n = 18) or PAD (n = 19) patients were processed for scanning electron microscopy, (immune)stained for fibrin, VWF and extracellular DNA. Fibrin fiber diameter, cellular components, fibrin/DNA and fibrin/VWF ratios were measured. RESULTS Patients' age presented as a strong explanatory factor for a linear decline trend of the VWF content relative to fibrin in thrombi from CAD (adjusted-R2 = 0.43) and male AIS (adjusted-R2 = 0.66) patients. In a subgroup of CAD and PAD patients with dyslipidemia and high (above 80%) prevalence of atherothrombosis a significant correlation was observed between the VWF and DNA content in thrombi (adjusted-R2 = 0.40), whereas a 3.7-fold lower linear regression coefficient was seen in AIS patients, in whom the fraction of thrombi of atherosclerotic origin was 57%. Independently of anatomical location, in patients with atherosclerosis the VWF in thrombi correlated with the plasma C-reactive protein levels. CONCLUSIONS The observed interrelations between thrombus constituents and systemic inflammatory biomarkers suggest an intricate interplay along the VWF/NET/fibrin axis in arterial thrombosis.
Collapse
Affiliation(s)
- Erzsébet Tóth
- Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - László Beinrohr
- Department of Biochemistry, Semmelweis University, Budapest, Hungary
| | - István Gubucz
- National Institute of Clinical Neurosciences, Budapest, Hungary
| | - László Szabó
- Department of Biochemistry, Semmelweis University, Budapest, Hungary; Department of Functional and Structural Materials, Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Kiril Tenekedjiev
- Australian Maritime College, University of Tasmania, Launceston, Australia; Department of Information Technology, Nikola Vaptsarov Naval Academy, Varna, Bulgaria
| | - Natalia Nikolova
- Australian Maritime College, University of Tasmania, Launceston, Australia; Department of Information Technology, Nikola Vaptsarov Naval Academy, Varna, Bulgaria
| | - Anikó I Nagy
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary; Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - László Hidi
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Péter Sótonyi
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - István Szikora
- National Institute of Clinical Neurosciences, Budapest, Hungary
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Krasimir Kolev
- Department of Biochemistry, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
181
|
Prigent K, Vigne J. Advances in Radiopharmaceutical Sciences for Vascular Inflammation Imaging: Focus on Clinical Applications. Molecules 2021; 26:molecules26237111. [PMID: 34885690 PMCID: PMC8659223 DOI: 10.3390/molecules26237111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/09/2021] [Accepted: 11/19/2021] [Indexed: 01/18/2023] Open
Abstract
Biomedical imaging technologies offer identification of several anatomic and molecular features of disease pathogenesis. Molecular imaging techniques to assess cellular processes in vivo have been useful in advancing our understanding of several vascular inflammatory diseases. For the non-invasive molecular imaging of vascular inflammation, nuclear medicine constitutes one of the best imaging modalities, thanks to its high sensitivity for the detection of probes in tissues. 2-[18F]fluoro-2-deoxy-d-glucose ([18F]FDG) is currently the most widely used radiopharmaceutical for molecular imaging of vascular inflammatory diseases such as atherosclerosis and large-vessel vasculitis. The combination of [18F]FDG and positron emission tomography (PET) imaging has become a powerful tool to identify and monitor non-invasively inflammatory activities over time but suffers from several limitations including a lack of specificity and avid background in different localizations. The use of novel radiotracers may help to better understand the underlying pathophysiological processes and overcome some limitations of [18F]FDG PET for the imaging of vascular inflammation. This review examines how [18F]FDG PET has given us deeper insight into the role of inflammation in different vascular pathologies progression and discusses perspectives for alternative radiopharmaceuticals that could provide a more specific and simple identification of pathologies where vascular inflammation is implicated. Use of these novel PET tracers could lead to a better understanding of underlying disease mechanisms and help inform the identification and stratification of patients for newly emerging immune-modulatory therapies. Future research is needed to realize the true clinical translational value of PET imaging in vascular inflammatory diseases.
Collapse
Affiliation(s)
- Kevin Prigent
- CHU de Caen Normandie, Department of Nuclear Medicine, Normandie Université, UNICAEN, 14000 Caen, France;
| | - Jonathan Vigne
- CHU de Caen Normandie, Department of Nuclear Medicine, Normandie Université, UNICAEN, 14000 Caen, France;
- CHU de Caen Normandie, Department of Pharmacy, Normandie Université, UNICAEN, 14000 Caen, France
- UNICAEN, INSERM U1237, Etablissement Français du Sang, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Normandie University, 14000 Caen, France
- Correspondence:
| |
Collapse
|
182
|
Cao G, Xuan X, Zhang R, Hu J, Dong H. Gene Therapy for Cardiovascular Disease: Basic Research and Clinical Prospects. Front Cardiovasc Med 2021; 8:760140. [PMID: 34805315 PMCID: PMC8602679 DOI: 10.3389/fcvm.2021.760140] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/11/2021] [Indexed: 12/16/2022] Open
Abstract
In recent years, the vital role of genetic factors in human diseases have been widely recognized by scholars with the deepening of life science research, accompanied by the rapid development of gene-editing technology. In early years, scientists used homologous recombination technology to establish gene knock-out and gene knock-in animal models, and then appeared the second-generation gene-editing technology zinc-finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs) that relied on nucleic acid binding proteins and endonucleases and the third-generation gene-editing technology that functioned through protein-nucleic acids complexes-CRISPR/Cas9 system. This holds another promise for refractory diseases and genetic diseases. Cardiovascular disease (CVD) has always been the focus of clinical and basic research because of its high incidence and high disability rate, which seriously affects the long-term survival and quality of life of patients. Because some inherited cardiovascular diseases do not respond well to drug and surgical treatment, researchers are trying to use rapidly developing genetic techniques to develop initial attempts. However, significant obstacles to clinical application of gene therapy still exists, such as insufficient understanding of the nature of cardiovascular disease, limitations of genetic technology, or ethical concerns. This review mainly introduces the types and mechanisms of gene-editing techniques, ethical concerns of gene therapy, the application of gene therapy in atherosclerosis and inheritable cardiovascular diseases, in-stent restenosis, and delivering systems.
Collapse
Affiliation(s)
- Genmao Cao
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xuezhen Xuan
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Ruijing Zhang
- Department of Nephrology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jie Hu
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Honglin Dong
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
183
|
Libby P. Inflammation during the life cycle of the atherosclerotic plaque. Cardiovasc Res 2021; 117:2525-2536. [PMID: 34550337 PMCID: PMC8783385 DOI: 10.1093/cvr/cvab303] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 09/20/2021] [Indexed: 12/12/2022] Open
Abstract
Inflammation orchestrates each stage of the life cycle of atherosclerotic plaques. Indeed, inflammatory mediators likely link many traditional and emerging risk factors with atherogenesis. Atheroma initiation involves endothelial activation with recruitment of leucocytes to the arterial intima, where they interact with lipoproteins or their derivatives that have accumulated in this layer. The prolonged and usually clinically silent progression of atherosclerosis involves periods of smouldering inflammation, punctuated by episodes of acute activation that may arise from inflammatory mediators released from sites of extravascular injury or infection or from subclinical disruptions of the plaque. Smooth muscle cells and infiltrating leucocytes can proliferate but also undergo various forms of cell death that typically lead to formation of a lipid-rich 'necrotic' core within the evolving intimal lesion. Extracellular matrix synthesized by smooth muscle cells can form a fibrous cap that overlies the lesion's core. Thus, during progression of atheroma, cells not only procreate but perish. Inflammatory mediators participate in both processes. The ultimate clinical complication of atherosclerotic plaques involves disruption that provokes thrombosis, either by fracture of the plaque's fibrous cap or superficial erosion. The consequent clots can cause acute ischaemic syndromes if they embarrass perfusion. Incorporation of the thrombi can promote plaque healing and progressive intimal thickening that can aggravate stenosis and further limit downstream blood flow. Inflammatory mediators regulate many aspects of both plaque disruption and healing process. Thus, inflammatory processes contribute to all phases of the life cycle of atherosclerotic plaques, and represent ripe targets for mitigating the disease.
Collapse
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, 77 Avenue Louis Pasteur, Boston, MA, USA
| |
Collapse
|
184
|
Fröbert O, Götberg M, Erlinge D, Akhtar Z, Christiansen EH, MacIntyre CR, Oldroyd KG, Motovska Z, Erglis A, Moer R, Hlinomaz O, Jakobsen L, Engstrøm T, Jensen LO, Fallesen CO, Jensen SE, Angerås O, Calais F, Kåregren A, Lauermann J, Mokhtari A, Nilsson J, Persson J, Stalby P, Islam AKMM, Rahman A, Malik F, Choudhury S, Collier T, Pocock SJ, Pernow J. Influenza Vaccination After Myocardial Infarction: A Randomized, Double-Blind, Placebo-Controlled, Multicenter Trial. Circulation 2021; 144:1476-1484. [PMID: 34459211 DOI: 10.1161/circulationaha.121.057042] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Observational and small, randomized studies suggest that influenza vaccine may reduce future cardiovascular events in patients with cardiovascular disease. METHODS We conducted an investigator-initiated, randomized, double-blind trial to compare inactivated influenza vaccine with saline placebo administered shortly after myocardial infarction (MI; 99.7% of patients) or high-risk stable coronary heart disease (0.3%). The primary end point was the composite of all-cause death, MI, or stent thrombosis at 12 months. A hierarchical testing strategy was used for the key secondary end points: all-cause death, cardiovascular death, MI, and stent thrombosis. RESULTS Because of the COVID-19 pandemic, the data safety and monitoring board recommended to halt the trial before attaining the prespecified sample size. Between October 1, 2016, and March 1, 2020, 2571 participants were randomized at 30 centers across 8 countries. Participants assigned to influenza vaccine totaled 1290 and individuals assigned to placebo equaled 1281; of these, 2532 received the study treatment (1272 influenza vaccine and 1260 placebo) and were included in the modified intention to treat analysis. Over the 12-month follow-up, the primary outcome occurred in 67 participants (5.3%) assigned influenza vaccine and 91 participants (7.2%) assigned placebo (hazard ratio, 0.72 [95% CI, 0.52-0.99]; P=0.040). Rates of all-cause death were 2.9% and 4.9% (hazard ratio, 0.59 [95% CI, 0.39-0.89]; P=0.010), rates of cardiovascular death were 2.7% and 4.5%, (hazard ratio, 0.59 [95% CI, 0.39-0.90]; P=0.014), and rates of MI were 2.0% and 2.4% (hazard ratio, 0.86 [95% CI, 0.50-1.46]; P=0.57) in the influenza vaccine and placebo groups, respectively. CONCLUSIONS Influenza vaccination early after an MI or in high-risk coronary heart disease resulted in a lower risk of a composite of all-cause death, MI, or stent thrombosis, and a lower risk of all-cause death and cardiovascular death, as well, at 12 months compared with placebo. Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT02831608.
Collapse
Affiliation(s)
- Ole Fröbert
- Örebro University, Faculty of Health, Department of Cardiology, Sweden (O.F., F.C.)
| | - Matthias Götberg
- Department of Cardiology, Skane University Hospital, Clinical Sciences, Lund University, Sweden (M.G., D.E., A.M.)
| | - David Erlinge
- Department of Cardiology, Skane University Hospital, Clinical Sciences, Lund University, Sweden (M.G., D.E., A.M.)
| | - Zubair Akhtar
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka (Z.A.)
| | | | - Chandini R MacIntyre
- The Kirby Institute, UNSW Medicine, University of New South Wales, Sydney, Australia (C.R.M.)
| | - Keith G Oldroyd
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, United Kingdom, and West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, Glasgow, United Kingdom (K.G.O.)
| | - Zuzana Motovska
- Cardiocenter, Third Faculty of Medicine, Charles University, and University Hospital Kralovske Vinohrady, Prague, Czech Republic (Z.M.)
| | - Andrejs Erglis
- Pauls Stradins Clinical University Hospital, University of Latvia, Riga (A.E.)
| | - Rasmus Moer
- LHL-sykehuset Gardermoen, Oslo, Norway (R.M.)
| | - Ota Hlinomaz
- International clinical research center, St. Anne University Hospital and Masaryk University, Brno, Czech Republic (O.H.)
| | - Lars Jakobsen
- Department of Cardiology, Aarhus University Hospital, Denmark (E.H.C., L.J.)
| | | | - Lisette O Jensen
- Department of Cardiology, Odense University Hospital, Denmark (L.O.J., C.O.F.)
| | | | - Svend E Jensen
- Department of Cardiology, Aalborg University Hospital, and Department of Clinical Medicine, Aalborg University, Denmark (S.E.J.)
| | - Oskar Angerås
- Sahlgrenska University Hospital and Institute of Medicine, Department of molecular and clinical medicine, Gothenburg University, Sweden (O.A.)
| | - Fredrik Calais
- Örebro University, Faculty of Health, Department of Cardiology, Sweden (O.F., F.C.)
| | | | - Jörg Lauermann
- Department of Cardiology, Jönköping, Region Jönköping County, and Department of Health, Medicine and Caring, Linköping University, Sweden (J.L.)
| | - Arash Mokhtari
- Department of Cardiology, Skane University Hospital, Clinical Sciences, Lund University, Sweden (M.G., D.E., A.M.)
| | - Johan Nilsson
- Cardiology, Heart Centre, Department of Public Health and Clinical Medicine, Umeå University, Sweden (J.N.)
| | - Jonas Persson
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Karolinska Institutet, Danderyd University Hospital, Stockholm, Sweden (J. Persson)
| | - Per Stalby
- Department of Cardiology, Karlstad Central Hospital, Sweden (P.S.)
| | - Abu K M M Islam
- National Institute of Cardiovascular Diseases, Sher-e-Bangla Nagar, Dhaka, Bangladesh (A.K.K.M.I., A.R.)
| | - Afzalur Rahman
- National Institute of Cardiovascular Diseases, Sher-e-Bangla Nagar, Dhaka, Bangladesh (A.K.K.M.I., A.R.)
| | - Fazila Malik
- National Heart Foundation Hospital and Research Institute, Dhaka, Bangladesh (F.M., S.C.)
| | - Sohel Choudhury
- National Heart Foundation Hospital and Research Institute, Dhaka, Bangladesh (F.M., S.C.)
| | - Timothy Collier
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (T.C., S.J.P.)
| | - Stuart J Pocock
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, United Kingdom (T.C., S.J.P.)
| | - John Pernow
- Cardiology Unit, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden (J. Pernow)
| |
Collapse
|
185
|
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
|
186
|
Maffia P, Antoniades C, Ahluwalia A, Cirino G. Molecular imaging-The first visual themed issue published in the British Journal of Pharmacology. Br J Pharmacol 2021; 178:4213-4215. [PMID: 34664273 DOI: 10.1111/bph.15632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Affiliation(s)
- Pasquale Maffia
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Amrita Ahluwalia
- The William Harvey Research Institute, Queen Mary University of London, Barts and The London School of Medicine & Dentistry, London, UK
| | - Giuseppe Cirino
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| |
Collapse
|
187
|
Li XQ, Tang XR, Li LL. Antipsychotics cardiotoxicity: What's known and what's next. World J Psychiatry 2021; 11:736-753. [PMID: 34733639 PMCID: PMC8546771 DOI: 10.5498/wjp.v11.i10.736] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 07/08/2021] [Accepted: 09/02/2021] [Indexed: 02/06/2023] Open
Abstract
Chronic use of antipsychotic medications entails a dilemma between the benefit of alleviating psychotic symptoms and the risk of troubling, sometimes life-shortening adverse effects. Antipsychotic-induced cardiotoxicity is one of the most life-threatening adverse effects that raises widespread concerns. These cardiotoxic effects range from arrhythmia to heart failure in the clinic, with myocarditis/cardiomyopathy, ischemic injuries, and unexplained cardiac lesions as the pathological bases. Multiple mechanisms have been proposed to underlie antipsychotic cardiotoxicity. This review aims to summarize the clinical signs and pathological changes of antipsychotic cardiotoxicity and introduce recent progress in understanding the underlying mechanisms at both the subcellular organelle level and the molecular level. We also provide an up-to-date perspective on future clinical monitoring and therapeutic strategies for antipsychotic cardiotoxicity. We propose that third-generation antipsychotics or drug adjuvant therapy, such as cannabinoid receptor modulators that confer dual benefits — i.e., alleviating cardiotoxicity and improving metabolic disorders — deserve further clinical evaluation and marketing.
Collapse
Affiliation(s)
- Xiao-Qing Li
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xin-Ru Tang
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Li-Liang Li
- Department of Forensic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
188
|
Radkhah N, Shabbidar S, Zarezadeh M, Safaeiyan A, Barzegar A. Effects of vitamin D supplementation on apolipoprotein A1 and B100 levels in adults: Systematic review and meta-analysis of controlled clinical trials. J Cardiovasc Thorac Res 2021; 13:190-197. [PMID: 34630965 PMCID: PMC8493225 DOI: 10.34172/jcvtr.2021.21] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 02/10/2021] [Indexed: 11/09/2022] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of death around the world. According to the studies, apolipoproteins A1 and B100 play crucial role in CVD development and progression. Also, findings have indicated the positive role of vitamin D on these factors. Thus, we conducted the present meta-analysis of randomized controlled trials (RCTs) to demonstrate the impact of vitamin D supplementation on apolipoproteins A1 and B100 levels in adults. PubMed and Scopus databases and Google Scholar were searched up to 21 December 2020. Relevant articles were screened, extracted, and assessed for quality based on the Cochrane collaboration's risk of bias tool. Data analysis conducted by random-effect model and expressed by standardized mean difference (SMD). The heterogeneity between studies was assessed by I-squared (I2) test. Subgroups and sensitivity Analyses were also conducted. Seven RCTs were identified investigating the impact of vitamin D on Apo A1 levels and six on Apo B100 levels. The findings showed the insignificant effect of vitamin D supplementation on Apo A1 (SMD=0.26 mg/dL; 95% confidence interval (CI), -0.10, 0.61; P = 0.155) and Apo B100 (standardized mean difference (SMD)=-0.06 mg/dL; 95% CI, -0.24, 0.12; P = 0.530) in adults. There was a significant between-study heterogeneity in Apo A1 (I2=89.3%, P < 0.001) and Apo B100 (I2 = 57.1%, P = 0.030). However, significant increase in Apo A1 in daily dosage of vitamin D (SMD=0.56 mg/dL; 95% CI, 0.02, 1.11; P = 0.044) and ≤12 weeks of supplementation duration (SMD=0.71 mg/dL; 95% CI, 0.08, 1.34; P = 0.028) was observed. No significant effects of vitamin D on Apo A1 and Apo B100 levels after subgroup analysis by mean age, gender, study population, dosage and duration of study. Overall, daily vitamin D supplementation and ≤12 weeks of supplementation might have beneficial effects in increasing Apo A1 levels, however, future high-quality trials considering these a primary outcome are required.
Collapse
Affiliation(s)
- Nima Radkhah
- Department of Community Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sakineh Shabbidar
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Meysam Zarezadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abdolrasoul Safaeiyan
- Department of Vital Statistics and Epidemiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Barzegar
- Department of Community Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
189
|
Gianos E, Jackson EA, Tejpal A, Aspry K, O'Keefe J, Aggarwal M, Jain A, Itchhaporia D, Williams K, Batts T, Allen KE, Yarber C, Ostfeld RJ, Miller M, Reddy K, Freeman AM, Fleisher KE. Oral health and atherosclerotic cardiovascular disease: A review. Am J Prev Cardiol 2021; 7:100179. [PMID: 34611631 PMCID: PMC8387275 DOI: 10.1016/j.ajpc.2021.100179] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Periodontal disease (PD) is common in the US and globally. Evidence suggests that poor oral health is associated with atherosclerotic cardiovascular disease (ASCVD); however, this relationship has not been a major focus in clinical cardiology. This manuscript will review the growing evidence linking PD to ASCVD, including pathophysiologic mechanisms and coexistent risk factors. Public health considerations with a focus on disparities, social determinants, preventive strategies, and a call to action to reduce the burden of coincident ASCVD and PD are also reviewed.
Collapse
Affiliation(s)
- Eugenia Gianos
- Division of Cardiology, Lenox Hill Hospital, Northwell Health, New York, NY, United States
| | - Elizabeth A Jackson
- Division of Cardiovascular Disease, Department of Internal Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Astha Tejpal
- Division of Cardiology, Lenox Hill Hospital, Northwell Health, New York, NY, United States
| | - Karen Aspry
- Lifespan Cardiovascular Institute, and Division of Cardiology, Brown University, Alpert Medical School, Providence, RI, United States
| | - James O'Keefe
- Saint Luke's Mid America Heart Institute and University of Missouri-Kansas City School of Medicine, Kansas City, MI, United States
| | - Monica Aggarwal
- Division of Cardiology, University of Florida, Gainesville, FL, United States
| | - Ankur Jain
- Division of Cardiology, University of Florida, Gainesville, FL, United States
| | - Dipti Itchhaporia
- Jeffrey M. Carlton Heart & Vascular Institute, Hoag Memorial Hospital, Newport Beach, CA, United States
| | - Kim Williams
- Department of Medicine, Division of Cardiology, Rush University Medical Center, Chicago, IL, United States
| | - Travis Batts
- Division of Cardiology, Department of Medicine, Wilford Hall Ambulatory Surgical Center, San Antonio, TX, United States
| | - Kathleen E Allen
- Geisel School of Medicine at Dartmouth, Hanover, NY, United States
| | - Clark Yarber
- Department of Internal Medicine, Montefiore Health System, Bronx, NY, United States
| | - Robert J Ostfeld
- Division of Cardiology, Department of Medicine, Montefiore Health System, Bronx, NY, United States
| | - Michael Miller
- Department of Cardiovascular Medicine, Epidemiology & Public Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Koushik Reddy
- Division of Cardiology, James A. Haley VA Medical Center, University of South Florida, Tampa, FL, United States
| | - Andrew M Freeman
- Division of Cardiology, Department of Medicine, National Jewish Health, Denver, CO, United States
| | - Kenneth E Fleisher
- Department of Oral and Maxillofacial Surgery, NYU College of Dentistry, New York, NY, United States
| |
Collapse
|
190
|
Arévalos V, Ortega-Paz L, Rodríguez-Arias JJ, Calvo López M, Castrillo-Golvano L, Salazar-Rodríguez A, Sabaté-Tormos M, Spione F, Sabaté M, Brugaletta S. Acute and Chronic Effects of COVID-19 on the Cardiovascular System. J Cardiovasc Dev Dis 2021; 8:128. [PMID: 34677197 PMCID: PMC8541609 DOI: 10.3390/jcdd8100128] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/24/2021] [Accepted: 09/29/2021] [Indexed: 01/08/2023] Open
Abstract
COVID-19 has shown significant morbidity with the involvement of multiple systems, including the cardiovascular system. Cardiovascular manifestations in the acute phase can include myocardial injury itself, myocardial infarction, venous thromboembolic events, myocarditis, Takotsubo syndrome, and different arrhythmic events. Myocardial injury defined by the rise of cardiac biomarkers in blood has been found in multiple studies with a prevalence of about 20%. Its presence is related to worse clinical outcomes and in-hospital mortality. The mechanisms of myocardial injury have been the subject of intense research but still need to be clarified. The characterization of the cardiac affectation with echocardiography and cardiac magnetic resonance has found mixed results in different studies, with a striking incidence of imaging criteria for myocarditis. Regarding post-acute and chronic follow-up results, the persistence of symptoms and imaging changes in recovered COVID-19 patients has raised concerns about the duration and the possible significance of these findings. Even though the knowledge about this disease has increased incredibly in the last year, many aspects are still unclear and warrant further research.
Collapse
Affiliation(s)
- Victor Arévalos
- Department of Cardiology, Clinic Cardiovascular Institute, Hospital Clinic, 08036 Barcelona, Spain; (V.A.); (L.O.-P.); (J.J.R.-A.); (M.C.L.); (L.C.-G.); (A.S.-R.); (F.S.); (M.S.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain
| | - Luis Ortega-Paz
- Department of Cardiology, Clinic Cardiovascular Institute, Hospital Clinic, 08036 Barcelona, Spain; (V.A.); (L.O.-P.); (J.J.R.-A.); (M.C.L.); (L.C.-G.); (A.S.-R.); (F.S.); (M.S.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain
| | - Juan José Rodríguez-Arias
- Department of Cardiology, Clinic Cardiovascular Institute, Hospital Clinic, 08036 Barcelona, Spain; (V.A.); (L.O.-P.); (J.J.R.-A.); (M.C.L.); (L.C.-G.); (A.S.-R.); (F.S.); (M.S.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain
| | - Margarita Calvo López
- Department of Cardiology, Clinic Cardiovascular Institute, Hospital Clinic, 08036 Barcelona, Spain; (V.A.); (L.O.-P.); (J.J.R.-A.); (M.C.L.); (L.C.-G.); (A.S.-R.); (F.S.); (M.S.)
| | - Leticia Castrillo-Golvano
- Department of Cardiology, Clinic Cardiovascular Institute, Hospital Clinic, 08036 Barcelona, Spain; (V.A.); (L.O.-P.); (J.J.R.-A.); (M.C.L.); (L.C.-G.); (A.S.-R.); (F.S.); (M.S.)
| | - Anthony Salazar-Rodríguez
- Department of Cardiology, Clinic Cardiovascular Institute, Hospital Clinic, 08036 Barcelona, Spain; (V.A.); (L.O.-P.); (J.J.R.-A.); (M.C.L.); (L.C.-G.); (A.S.-R.); (F.S.); (M.S.)
| | - Marta Sabaté-Tormos
- Department of Medicine, Medical School, Universitat de Barcelona, 08036 Barcelona, Spain;
| | - Francesco Spione
- Department of Cardiology, Clinic Cardiovascular Institute, Hospital Clinic, 08036 Barcelona, Spain; (V.A.); (L.O.-P.); (J.J.R.-A.); (M.C.L.); (L.C.-G.); (A.S.-R.); (F.S.); (M.S.)
| | - Manel Sabaté
- Department of Cardiology, Clinic Cardiovascular Institute, Hospital Clinic, 08036 Barcelona, Spain; (V.A.); (L.O.-P.); (J.J.R.-A.); (M.C.L.); (L.C.-G.); (A.S.-R.); (F.S.); (M.S.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain
| | - Salvatore Brugaletta
- Department of Cardiology, Clinic Cardiovascular Institute, Hospital Clinic, 08036 Barcelona, Spain; (V.A.); (L.O.-P.); (J.J.R.-A.); (M.C.L.); (L.C.-G.); (A.S.-R.); (F.S.); (M.S.)
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain
| |
Collapse
|
191
|
Collado A, Domingo E, Piqueras L, Sanz MJ. Primary hypercholesterolemia and development of cardiovascular disorders: Cellular and molecular mechanisms involved in low-grade systemic inflammation and endothelial dysfunction. Int J Biochem Cell Biol 2021; 139:106066. [PMID: 34438057 DOI: 10.1016/j.biocel.2021.106066] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 12/28/2022]
Abstract
Primary hypercholesterolemia, a metabolic disorder characterized by elevated circulating levels of cholesterol products, mainly low-density lipoproteins, is associated with arteriosclerosis development. Cardiovascular disease, predominantly myocardial infarction and stroke, remains the main cause of death worldwide, with atherosclerosis considered to be the most common underlying pathology. In addition to elevated plasma levels of low-density lipoproteins, low-grade systemic inflammation and endothelial dysfunction seem to be the main drivers of premature atherosclerosis. Here we review current knowledge related to cellular and molecular mechanisms involved in low-grade systemic inflammation and endothelial dysfunction associated with primary hypercholesterolemia. We also discuss the contribution of different inflammatory mediators, immune players and signaling pathways implicated in leukocyte adhesion to the dysfunctional endothelium, a key feature of atherogenesis development. A better understanding of these processes linked to primary hypercholesterolemia should shed new light on cardiovascular disease development and might guide novel and effective therapeutic strategies to impair its progression.
Collapse
Affiliation(s)
- Aida Collado
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain; Institute of Health Research INCLIVA, University Clinic Hospital of Valencia, Valencia, Spain.
| | - Elena Domingo
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain; Institute of Health Research INCLIVA, University Clinic Hospital of Valencia, Valencia, Spain
| | - Laura Piqueras
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain; Institute of Health Research INCLIVA, University Clinic Hospital of Valencia, Valencia, Spain; CIBERDEM-Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders, Carlos III Health Institute, Spanish Ministry of Health, Madrid, Spain
| | - Maria-Jesus Sanz
- Department of Pharmacology, Faculty of Medicine, University of Valencia, Valencia, Spain; Institute of Health Research INCLIVA, University Clinic Hospital of Valencia, Valencia, Spain; CIBERDEM-Spanish Biomedical Research Center in Diabetes and Associated Metabolic Disorders, Carlos III Health Institute, Spanish Ministry of Health, Madrid, Spain.
| |
Collapse
|
192
|
The Role and Molecular Mechanism of P2Y12 Receptors in the Pathogenesis of Atherosclerotic Cardiovascular Diseases. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11199078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The P2Y receptor family is a class of G protein-coupled receptors activated primarily by adenosine triphosphate (ATP), adenosine diphosphate (ADP), uridine triphosphate (UTP) and uridine diphosphate (UDP). The P2Y12 receptor is expressed on platelets which mediates platelet aggregation and morphological changes. At the same time, during the process of vascular remodeling and atherosclerosis, ADP can also promote the migration and proliferation of vascular smooth muscle and endothelial cells through P2Y12 receptor activating. Furthermore, P2Y12 is involved in many signal transductions processes, such as intimal hyperplasia, monocyte infiltration and so on, which play an important role in immune inflammation and brain injury. In order to solve the diseases induced by P2Y12 receptor, inhibitors such as ticagrelor, clopidogrel were widely used for cardiovascular diseases. However, there were some problems, such as limited antithrombotic effect, remain unsolved. This article summarizes the role and molecular mechanism of P2Y12 receptors in the pathogenesis of cardiovascular-related diseases, providing in-depth expounding on the molecular mechanism of P2Y12 receptor inhibitors and contributing to the treatment of diseases based on P2Y12 receptors.
Collapse
|
193
|
Association Between Tooth Loss and Longitudinal Changes in B-Type Natriuretic Peptide Over 5 Years in Postmenopausal Women: The Nagahama Study. Curr Probl Cardiol 2021; 47:100997. [PMID: 34582901 DOI: 10.1016/j.cpcardiol.2021.100997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/14/2021] [Indexed: 11/20/2022]
Abstract
There is disparity between the sexes in cardiovascular diseases including heart failure (HF). This study aimed to investigate the effect of periodontal disease (PD) on plasma B-type natriuretic peptide (BNP) concentration across sex, age, and menopausal status, as well as the interaction effect of PD and diabetes mellitus (DM) on BNP. This large-scale prospective cohort study enrolled 7539 individuals with no myocardial infarctions or angina pectoris at baseline from the general Japanese population. The association between baseline number of missing teeth (MT) and the longitudinal changes in BNP over 5 years (ΔBNP) was evaluated according to sex and menopausal status. Among 7539 participants, 3190 were postmenopausal women with a mean age ± standard deviation of 61.1 ± 7.6 at baseline. Multivariate analysis revealed a positive association between MT and ΔBNP among postmenopausal women even after adjusting for covariates, including traditional HF risk factors (coefficient, 0.210; 95% confidence interval [CI], 0.107 to 0.312; P < 0.001), but not in men aged > 50. Including an interaction term (MT × DM) in the multivariate model revealed a positive interaction between MT and DM in ΔBNP among postmenopausal women (coefficient for interaction, 1.365; 95% CI, 0.902 to 1.827; P for interaction < 0.001). In conclusion, our study showed a positive association between MT and ΔBNP, as well as a positive effect of the interactive association between MT and DM, among postmenopausal women. Our results suggest a sex difference of an adverse effect of PD on initial myocardial wall stress in the ventricles.
Collapse
|
194
|
Samoilova EM, Yusubalieva GM, Belopasov VV, Ekusheva EV, Baklaushev VP. [Infections and inflammation in the development of stroke]. Zh Nevrol Psikhiatr Im S S Korsakova 2021; 121:11-21. [PMID: 34553576 DOI: 10.17116/jnevro202112108211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The review systematizes data on the role of infectious diseases and systemic inflammation in the pathogenesis of stroke. Various risk factors for stroke associated with pro-inflammatory reactions and their contribution to the pathogenesis of cerebrovascular pathology are analyzed. The interaction of systemic inflammation with hemostasis disturbances and clots formation, activation of autoreactive clones of cytotoxic lymphocytes, the progression of endothelial damage, and other processes is shown. Along with infection, these factors increase the risk of stroke. The key mechanisms of the pathogenesis from the development of acute or chronic inflammation to the preconditions of stroke are presented. The mechanisms of the acting of the infectious process as a trigger factor and/or medium-term or long-term risk factors of stroke are described. A separate section is devoted to the mechanisms of developing cerebrovascular diseases after COVID-19. Identifying an increased risk of stroke due to infection can be of great preventive value. Understanding of this risk by specialists followed by correction of drug therapy and rehabilitation measures can reduce the incidence of cerebrovascular complications in infectious patients.
Collapse
Affiliation(s)
- E M Samoilova
- Federal Scientific and Clinical Center of Specialized Types of Medical Care and Medical Technologies of the Federal Medical and Biological Agency of Russia, Moscow, Russia
| | - G M Yusubalieva
- Federal Scientific and Clinical Center of Specialized Types of Medical Care and Medical Technologies of the Federal Medical and Biological Agency of Russia, Moscow, Russia
| | - V V Belopasov
- Astrakhan State Medical University, Astrakhan, Russia
| | - E V Ekusheva
- Academy of Postgraduate Education of the Federal Scientific and Clinical Center for Specialized Types of Medical Care and Medical Technologies FMBA of Russia, Moscow, Russia.,Belgorod State National Research University, Belgorod, Russia
| | - V P Baklaushev
- Federal Scientific and Clinical Center of Specialized Types of Medical Care and Medical Technologies of the Federal Medical and Biological Agency of Russia, Moscow, Russia
| |
Collapse
|
195
|
Yuan C, Li Y, Liu L, Tayier B, Yang L, Guan L, Mu Y. Experimental Study on the Compatibility and Characteristics of a Dual-Target Microbubble Loaded with Anti-miR-33. Int J Nanomedicine 2021; 16:6265-6280. [PMID: 34539179 PMCID: PMC8445104 DOI: 10.2147/ijn.s324514] [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: 06/12/2021] [Accepted: 08/22/2021] [Indexed: 12/27/2022] Open
Abstract
Objective To prepare a new type of dual-target microbubble loaded with anti-miR-33 (ANM33). Methods Carrier core nanobubbles (NBs) were prepared by thin film hydration, and microbubbles loaded with PM1 (PCNBs) were prepared by grafting DSPE-PEG2000-maleimide-PM1 onto the NB surface. ANM33 was connected via electrostatic adsorption and covalent bonding, and hyaluronic acid (HA) was covalently connected. PM1 and HA were the targets, and ANM33 was the intervention drug. To evaluate the general physical and chemical properties of the prepared dual-target microbubbles loaded with ANM33 (HA-PANBs), we observed their morphology, particle size and surface potential while monitoring their stability and in vitro imaging ability, evaluated their toxic effect on cells and verified their ability to target cells. Results HA-PANBs had a regular morphology and good stability. The average particle size measured by a Malvern potentiometer was 1421.75±163.23 nm, and the average surface potential was −5.51±1.87 mV. PM1 and ANM33 were effectively connected to the NBs. The PM1, ANM33, and HA binding reached 89.0±1.1%, 65.02±5.0%, and 61.4±3.5%, respectively, and the maximum binding reached 2 µg, 5 µg, and 7 µg/108 microbubbles, respectively. HA-PANBs had no obvious toxic effects on cells, and their ability to continuously enhance imaging in vitro persisted for more than 15 minutes, obviously targeting foam cells in the early stage of AS. Conclusion HA-PANBs are ideal ultrasound contrast agents. The successful, firm connection of PM1 and HA to the NBs significantly increased the amount of carried ANM33. When microbubbles prepared with 2:4:7 PM1:ANM33:HA were used as a contrast agent, they had a high ANM33 carrying capacity, stable physical properties, and significantly enhanced imaging and targeting of foam cells in the early stage of AS.
Collapse
Affiliation(s)
- Chen Yuan
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, People's Republic of China
| | - Yanhong Li
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, People's Republic of China
| | - Liyun Liu
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, People's Republic of China
| | - Baihetiya Tayier
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, People's Republic of China
| | - Lingjie Yang
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, People's Republic of China
| | - Lina Guan
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, People's Republic of China.,State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, First Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China.,Xinjiang Key Laboratory of Medical Animal Model Research, Clinical Medical Research Institute of First Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China
| | - Yuming Mu
- Department of Echocardiography, First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, People's Republic of China.,State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, First Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China.,Xinjiang Key Laboratory of Medical Animal Model Research, Clinical Medical Research Institute of First Affiliated Hospital of Xinjiang Medical University, Urumqi, People's Republic of China
| |
Collapse
|
196
|
da Cruz BO, Cardozo LFMD, Coutinho-Wolino KS, Mesquita CT, Leal VO, Mafra D, Stockler-Pinto MB. Brazil Nut Supplementation Does Not Regulate PPARβ/δ Signaling Pathway in Peripheral Blood Mononuclear Cells from Coronary Artery Disease Patients. J Am Coll Nutr 2021; 41:780-787. [PMID: 34516363 DOI: 10.1080/07315724.2021.1963882] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Peroxisome proliferator-activated receptor (PPAR)β/δ activation is a potential target for modulation of inflammation in cardiovascular disease. PPARβ/δ activation depends on the presence of a ligand, which may be pharmacological or natural, such as bioactive compounds and nutrients. Due to its composition, rich in selenium and unsaturated fatty acids, Brazil nuts have been related to reduced oxidative stress and inflammation in chronic non-communicable diseases and could regulate PPARβ/δ. This study aimed to evaluate the effects of Brazil nut supplementation on PPARβ/δ mRNA expression in patients with Coronary Artery Disease (CAD). METHODS A secondary analysis of a randomized controlled clinical trial was performed with 36 CAD patients. Patients were randomly assigned to either the Supplementation group or the control group and followed up for three months. The Supplementation group consumed 1 Brazil nut/day; the control group did not receive any intervention. At the baseline and after three months, analysis of gene expression and biochemical parameters linked to inflammatory biomarkers and oxidative stress was carried out. RESULTS In the supplementation group, no significant change was observed in PPARβ/δ (0.9 ± 0.5 vs 1.2 ± 0.6; p = 0.178) and NF-κB (1.6 ± 1.5 vs 0.8 ± 0.30, p = 0.554) mRNA expression. There were no significant changes in both groups concerning all the other biochemical parameters. CONCLUSION One Brazil nut per day for three months was not able to increase the PPARβ/δ expression in CAD patients.
Collapse
Affiliation(s)
- Beatriz Oliveira da Cruz
- Graduate Program in Cardiovascular Sciences, Universidade Federal Fluminense, Niterói, Brazil.,Clinical Research Unit, Hospital Universitário Antônio Pedro, Universidade Federal Fluminense, Niterói, Brazil
| | - Ludmila Ferreira Medeiros de Cardozo
- Graduate Program in Cardiovascular Sciences, Universidade Federal Fluminense, Niterói, Brazil.,Clinical Research Unit, Hospital Universitário Antônio Pedro, Universidade Federal Fluminense, Niterói, Brazil
| | - Karen Salve Coutinho-Wolino
- Clinical Research Unit, Hospital Universitário Antônio Pedro, Universidade Federal Fluminense, Niterói, Brazil.,Graduate Program in Nutrition Sciences, Universidade Federal Fluminense, Niterói, Brazil
| | - Claudio Tinoco Mesquita
- Graduate Program in Cardiovascular Sciences, Universidade Federal Fluminense, Niterói, Brazil.,Hospital Universitário Antonio Pedro - EBSERH-UFF, Niterói, Brazil
| | - Viviane Oliveira Leal
- Hospital Universitário Pedro Ernesto, Universidade Estadual do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Denise Mafra
- Graduate Program in Cardiovascular Sciences, Universidade Federal Fluminense, Niterói, Brazil.,Clinical Research Unit, Hospital Universitário Antônio Pedro, Universidade Federal Fluminense, Niterói, Brazil.,Graduate Program in Nutrition Sciences, Universidade Federal Fluminense, Niterói, Brazil.,Graduate Program in Medical Sciences, Universidade Federal Fluminense, Niterói, Brazil
| | - Milena Barcza Stockler-Pinto
- Graduate Program in Cardiovascular Sciences, Universidade Federal Fluminense, Niterói, Brazil.,Clinical Research Unit, Hospital Universitário Antônio Pedro, Universidade Federal Fluminense, Niterói, Brazil.,Graduate Program in Nutrition Sciences, Universidade Federal Fluminense, Niterói, Brazil
| |
Collapse
|
197
|
Martí-Carvajal AJ, De Sanctis JB, Dayer M, Martí-Amarista CE, Alegría E, Monge Martín D, Abd El Aziz M, Correa-Pérez A, Nicola S, Parise Vasco JM. Interleukin-receptor antagonist and tumor necrosis factor inhibitors for the primary and secondary prevention of atherosclerotic cardiovascular diseases. Hippokratia 2021. [DOI: 10.1002/14651858.cd014741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Arturo J Martí-Carvajal
- Facultad de Ciencias de la Salud Eugenio Espejo (Centro Cochrane Ecuador); Universidad UTE; Quito Ecuador
- Facultad de Medicina (Centro Cochrane Madrid); Universidad Francisco de Vitoria; Madrid Spain
- Cátedra Rectoral de Medicina Basada en la Evidencia; Universidad de Carabobo; Valencia Venezuela
| | - Juan Bautista De Sanctis
- The Institute of Molecular and Translational Medicine; Palacky University Olomouc, Faculty of Medicine and Dentistry; Olomouc Czech Republic
| | - Mark Dayer
- Department of Cardiology; Somerset NHS Foundation Trust; Taunton UK
| | | | - Eduardo Alegría
- Faculty of Medicine; Universidad Francisco de Vitoria; Madrid Spain
| | | | - Mohamed Abd El Aziz
- Internal medicine; Texas Tech University Health Sciences Center El PasoPaul L. Foster School of Medicine; El Paso, Texas USA
| | - Andrea Correa-Pérez
- Faculty of Medicine; Universidad Francisco de Vitoria; Madrid Spain
- Clinical Biostatistics Unit; Hospital Universitario Ramón y Cajal (IRYCIS); Madrid Spain
| | - Susana Nicola
- Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC); Universidad UTE; Quito Ecuador
| | - Juan Marcos Parise Vasco
- Centro Asociado Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC); Universidad UTE; Quito Ecuador
| |
Collapse
|
198
|
Silvis MJM, Fiolet ATL, Opstal TSJ, Dekker M, Suquilanda D, Zivkovic M, Duyvendak M, The SHK, Timmers L, Bax WA, Mosterd A, Cornel JH, de Kleijn DPV. Colchicine reduces extracellular vesicle NLRP3 inflammasome protein levels in chronic coronary disease: A LoDoCo2 biomarker substudy. Atherosclerosis 2021; 334:93-100. [PMID: 34492522 DOI: 10.1016/j.atherosclerosis.2021.08.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 06/27/2021] [Accepted: 08/04/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND AIMS Colchicine reduces the risk of cardiovascular events in patients with coronary disease. Colchicine has broad anti-inflammatory effects and part of the atheroprotective effects have been suggested to be the result of NLRP3 inflammasome inhibition. We studied the effect of colchicine on extracellular vesicle (EV) NLRP3 protein levels and inflammatory markers, high sensitivity-CRP (hs-CRP) and interleukin (IL)-6, in patients with chronic coronary disease. METHODS In vitro, the NLRP3 inflammasome was stimulated in PMA-differentiated- and undifferentiated THP-1 cells. In vivo, measurements were performed in serum obtained from 278 participants of the LoDoCo2 trial, one year after randomization to colchicine 0.5 mg once daily or placebo. EVs were isolated using precipitation. NLRP3 protein presence in EVs was confirmed using iodixanol density gradient centrifugation. Levels of NLRP3 protein, hs-CRP and IL-6 were measured using ELISA. RESULTS In vitro, NLRP3 inflammasome stimulation showed an increase of EV NLRP3 protein levels. EV NLRP3 protein levels were lower in patients treated with colchicine (median 1.38 ng/mL), compared to placebo (median 1.58 ng/mL) (p = 0.025). No difference was observed in serum NLRP3 protein levels. Serum hs-CRP levels were lower in patients treated with colchicine (median 0.80 mg/L) compared to placebo (median 1.34 mg/L) (p < 0.005). IL-6 levels were lower in patients treated with colchicine (median 2.07 ng/L) compared to placebo (median 2.59 ng/L), although this was not statistically significant (p = 0.076). CONCLUSIONS Colchicine leads to a reduction of EV NLRP3 protein levels. This indicates that inhibitory effects on the NLRP3 inflammasome might contribute to the atheroprotective effects of colchicine in coronary disease.
Collapse
Affiliation(s)
- Max J M Silvis
- Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Aernoud T L Fiolet
- Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands; Dutch Network for Cardiovascular Research (WCN), Utrecht, the Netherlands; Department of Cardiology, Meander Medical Center, Amersfoort, the Netherlands
| | - Tjerk S J Opstal
- Department of Cardiology, Northwest Clinics, Alkmaar, the Netherlands; Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mirthe Dekker
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, the Netherlands; Department of Cardiology, Amsterdam University Medical Centre, Amsterdam, the Netherlands
| | - Daniel Suquilanda
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Minka Zivkovic
- Van Creveldkliniek, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Michiel Duyvendak
- Department of Clinical Pharmacy, Antonius Hospital Sneek, Sneek, the Netherlands
| | - Salem H K The
- Department of Cardiology, Treant Hospital, Emmen, the Netherlands
| | - Leo Timmers
- Department of Cardiology, St. Antonius Hospital Nieuwegein, the Netherlands
| | - Willem A Bax
- Department of Internal Medicine, Northwest Clinics, Alkmaar, the Netherlands
| | - Arend Mosterd
- Dutch Network for Cardiovascular Research (WCN), Utrecht, the Netherlands; Department of Cardiology, Meander Medical Center, Amersfoort, the Netherlands
| | - Jan H Cornel
- Dutch Network for Cardiovascular Research (WCN), Utrecht, the Netherlands; Department of Cardiology, Northwest Clinics, Alkmaar, the Netherlands; Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Dominique P V de Kleijn
- Department of Vascular Surgery, University Medical Centre Utrecht, Utrecht, the Netherlands; Netherlands Heart Institute, Utrecht, the Netherlands.
| |
Collapse
|
199
|
Abstract
PURPOSE OF REVIEW After coronary artery bypass grafting (CABG), patients remain at increased risk of cardiovascular events and death. Cardiac surgeons have the opportunity to reduce this risk by optimizing post-CABG patients' medical therapy. RECENT FINDINGS Recent developments in lipid-lowering, diabetes management, antithrombotic therapy, and anti-inflammatory therapy can significantly improve prognosis in patients with chronic coronary artery disease. PCSK-9 inhibitors should be used in patients with elevated LDL cholesterol despite maximally tolerated statin therapy. Icosapent ethyl should be considered in patients with elevated triglycerides despite maximally tolerated statin therapy. Long-acting GLP-1 receptor agonists or SLGT-2 inhibitors should be used in all post-CABG patients with type 2 diabetes. Intensified antithrombotic therapy with DAPT or DPI reduces MACE (and DPI reduces mortality) in patients with high atherosclerotic burden. Colchicine has not yet been incorporated into guidelines on OMT for stable CAD but it is reasonable to consider using it in high-risk patients. SUMMARY We review the foundations of optimal medical therapy after CABG, and summarize recent advances with a focus on practical application for the busy cardiac surgeon.
Collapse
|
200
|
Doi T, Langsted A, Nordestgaard BG. A possible explanation for the contrasting results of REDUCE-IT vs. STRENGTH: cohort study mimicking trial designs. Eur Heart J 2021; 42:4807-4817. [PMID: 34455435 DOI: 10.1093/eurheartj/ehab555] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/23/2021] [Accepted: 08/09/2021] [Indexed: 01/11/2023] Open
Abstract
AIMS We tested the hypothesis that the contrasting results for the effect of high-dose, purified omega-3 fatty acids on the prevention of atherosclerotic cardiovascular disease (ASCVD) in two randomized trials, Reduction of Cardiovascular Events with Icosapent Ethyl-Intervention Trial (REDUCE-IT) vs. Long-Term Outcomes Study to Assess Statin Residual Risk with Epanova in High Cardiovascular Risk Patients with Hypertriglyceridaemia (STRENGTH), can be explained by differences in the effect of active and comparator oils on lipid traits and C-reactive protein. METHODS AND RESULTS In the Copenhagen General Population Study (CGPS) with 106 088 individuals, to mimic trial designs we analysed those who met key inclusion criteria in REDUCE-IT (n = 5684; ASCVD = 852) and STRENGTH (n = 6862; ASCVD = 697). Atherosclerotic cardiovascular disease incidence was followed for the median durations of REDUCE-IT and STRENGTH (4.9 and 3.5 years), respectively. When combining changes in plasma triglycerides, low-density lipoprotein cholesterol, and C-reactive protein observed in the active oil groups of the original studies, estimated hazard ratios for ASCVD in the CGPS were 0.96 [95% confidence interval 0.93-0.99] mimicking REDUCE-IT and 0.94 (0.91-0.98) mimicking STRENGTH. In the comparator oil groups, corresponding hazard ratios were 1.07 (1.04-1.10) and 0.99 (0.98-0.99). Combining these results, the active oil vs. comparator oil hazard ratio was 0.88 (0.84-0.93) in the CGPS mimicking REDUCE-IT compared to 0.75 (0.68-0.83) in the REDUCE-IT. The corresponding hazard ratio was 0.96 (0.93-0.99) in the CGPS mimicking STRENGTH compared to 0.99 (0.90-1.09) in STRENGTH. CONCLUSION The contrasting results of REDUCE-IT vs. STRENGTH can partly be explained by a difference in the effect of comparator oils (mineral vs. corn), but not of active oils [eicosapentaenoic acid (EPA) vs. EPA + docosahexaenoic acid], on lipid traits and C-reactive protein. The unexplained additional 13% risk reduction in REDUCE-IT likely is through other effects of EPA or mineral oil.
Collapse
Affiliation(s)
- Takahito Doi
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Herlev 2730, Denmark.,The Copenhagen General Population Study, Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Herlev 2730, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark
| | - Anne Langsted
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Herlev 2730, Denmark.,The Copenhagen General Population Study, Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Herlev 2730, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Herlev 2730, Denmark.,The Copenhagen General Population Study, Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Borgmester Ib Juuls Vej 73, Herlev 2730, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, Copenhagen 2200, Denmark
| |
Collapse
|