1
|
Rizwan S, Ayubcha C, Al-Daoud O, Al-Atout M, Amiruddin R, Werner TJ, Alavi A. PET imaging of atherosclerosis: artificial intelligence applications and recent advancements. Nucl Med Commun 2025; 46:503-514. [PMID: 40143664 DOI: 10.1097/mnm.0000000000001973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
PET imaging has become a valuable tool for assessing atherosclerosis by targeting key processes such as inflammation and microcalcification. Among available tracers, 18F-sodium fluoride has demonstrated superior performance compared to 18F-fluorodeoxyglucose, particularly in detecting coronary artery disease. However, the role of other tracers remains underexplored, requiring further validation. Emerging technologies such as artificial intelligence show potential in enhancing diagnostic speed and accuracy. Furthermore, the integration of the Alavi-Carlsen Calcification Score offers a novel approach to evaluating global disease burden, presenting a more clinically applicable method for predicting outcomes. Techniques such as total-body PET provide faster and more comprehensive imaging of the entire vascular system with reduced radiation exposure, representing a significant advancement in early detection and intervention. The combination of molecular imaging and advanced computational tools may revolutionize the management of atherosclerosis, facilitating earlier identification of at-risk individuals and improving long-term cardiovascular outcomes.
Collapse
Affiliation(s)
- Shaheer Rizwan
- Robert Wood Johnson Medical School, New Brunswick, New Jersey
| | - Cyrus Ayubcha
- Harvard Medical School, Boston, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Omar Al-Daoud
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mamdouh Al-Atout
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Raisa Amiruddin
- Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Thomas J Werner
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Abass Alavi
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
2
|
Buja LM, McDonald MM, Zhao B, Narula N, Narula J, Barth RF. Insights from autopsy-initiated pathological studies of the pathogenesis and clinical manifestations of atherosclerosis and ischemic heart disease: Part I. Atherosclerosis. Cardiovasc Pathol 2025; 76:107726. [PMID: 39971218 DOI: 10.1016/j.carpath.2025.107726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/21/2025] Open
Abstract
CONTEXT Ischemic heart disease (IHD) due to coronary atherosclerosis constitutes the leading cause of morbidity and mortality worldwide. This review was undertaken to document the historical basis for our contemporary understanding of atherosclerosis-based disease and to provide a rationale for continued support for autopsy-based research to make further progress in reducing the morbidity and mortality from atherosclerosis-related disease. OBJECTIVES To analyze the contributions of the autopsy-initiated pathological studies to complement and validate other lines of investigation in determining the pathology and pathogenesis of the leading worldwide cause of morbidity and mortality, namely, atherosclerosis and its major complications of coronary atherosclerosis, ischemic heart disease, coronary thrombosis, acute myocardial infarction, and sudden cardiac death. DATA SOURCES Systematic search on PubMed to gather relevant studies concerning autopsy studies and reviews of the pathology and pathogenesis of atherosclerosis, ischemic heart disease, coronary atherosclerosis, coronary thrombosis, myocardial infarction, and sudden cardiac death CONCLUSIONS: Extensive published reports have confirmed the continuing importance of the autopsy as a powerful tool to understand the pathogenesis, clinical features, and therapeutic options for major diseases. This specifically has been shown by the analysis of atherosclerosis and its major manifestation of ischemic heart disease, as presented in this (Part I) and its companion (Part II) review. Autopsy-initiated pathological studies have documented the prevalence and natural history of atherosclerosis in different human populations in relationship to the prevalence of risk factors and established that the clinically silent phase of the disease begins in the first decades of life. Insights from these studies have been essential in developing and evaluating strategies for continued progress in preventing and controlling the disability and death associated with atherosclerotic heart disease.
Collapse
Affiliation(s)
- L Maximilian Buja
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth-Houston), Houston, Texas, USA.
| | | | - Bihong Zhao
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth-Houston), Houston, Texas, USA
| | - Navneet Narula
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth-Houston), Houston, Texas, USA
| | - Jagat Narula
- Division of Cardiology, Department of Internal Medicine, The University of Texas Health Science Center at Houston (UTHealth-Houston), Houston, Texas, USA
| | - Rolf F Barth
- Department of Pathology, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
3
|
Yang S, Zhou Y, Xu D, Dong Y, Tang H, Jing P, Lu YN, Yuan M, Zhao Z, Chen L. The associations between the FAGR and all-cause and cardiovascular mortality in patients with STEMI. Sci Rep 2025; 15:9237. [PMID: 40102522 PMCID: PMC11920094 DOI: 10.1038/s41598-025-93951-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 03/11/2025] [Indexed: 03/20/2025] Open
Abstract
Although the fibrinogen-to-albumin-to-globulin ratio (FAGR) has been proven to be related to coronary artery disease (CAD), the association between the FAGR and acute ST-segment elevation myocardial infarction (STEMI) has not been adequately investigated. This study aimed to evaluate the prognostic potential of the FAGR for STEMI. A total of 1042 patients with STEMI after emergency PCI admitted to the First Affiliated Hospital of Kunming Medical University from June 2018 to January 2023 were enrolled in the study. Patients were divided into a low FAGR group and a high FAGR group according to the median FAGR (2.44). We used Kaplan-Meier plots, restricted cubic spline regression, Cox survival analyses and time-dependent ROC analyses to explore the predictive value of the FAGR for all-cause and cardiovascular mortality. Kaplan‒Meier analysis revealed that the cumulative incidence rates of all-cause and cardiovascular mortality in patients with STEMI were greater in the high FAGR group. Multivariate Cox proportional hazard analysis revealed that the FAGR was an independent predictor of both all-cause and cardiovascular death. In terms of the prediction of all-cause mortality, the FAGR had an area under the ROC curve of 0.720, which was better than that for fibrinogen (AUC = 0.687). In terms of the prediction of cardiovascular mortality, the area under the ROC curve for the FAGR was 0.726, which was also better than that for Fib (AUC = 0.698). The present results suggest that the FAGR may serve as a potential prognostic indicator in patients with STEMI after emergency PCI.
Collapse
Affiliation(s)
- Sirui Yang
- Department of Cardiology, Kunming Medical University First Affiliated Hospital, Kunming, Yunnan Province, China
| | - Yanji Zhou
- Department of Pediatrics, Kunming Medical University First Affiliated Hospital, Kunming, Yunnan Province, China
| | - Dan Xu
- Curtin Medical School, School of Public Health, Faculty of Health Sciences, Curtin University, Perth, Australia
| | - Yilong Dong
- School of Medicine, Yunnan University, Kunming, Yunnan Province, China
| | - Haoran Tang
- Kunming Medical University First Affiliated Hospital, The First Clinical College of Kunming Medical University, Kunming City, Yunnan Province, China
| | - Pan Jing
- Department of Cardiology, Kunming Medical University First Affiliated Hospital, Kunming, Yunnan Province, China
| | - Ya-Nan Lu
- Department of Cardiology, Kunming Medical University First Affiliated Hospital, Kunming, Yunnan Province, China
| | - Minjing Yuan
- Kunming Medical University First Affiliated Hospital, The First Clinical College of Kunming Medical University, Kunming City, Yunnan Province, China
| | - Zhensong Zhao
- Kunming Medical University First Affiliated Hospital, The First Clinical College of Kunming Medical University, Kunming City, Yunnan Province, China
| | - Lixing Chen
- Department of Cardiology, Kunming Medical University First Affiliated Hospital, Kunming, Yunnan Province, China.
| |
Collapse
|
4
|
Zhang C, Wang J, Kong P, Sun G, Sun H, Yu Y. Circ-Sirt6 promotes the stability and expression of m 6A modified Sirt6 mRNA by recruiting IGF2BP2. FASEB J 2025; 39:e70405. [PMID: 39982754 DOI: 10.1096/fj.202402619r] [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: 10/28/2024] [Revised: 01/15/2025] [Accepted: 02/10/2025] [Indexed: 02/22/2025]
Abstract
Coronary artery disease (CAD) is the leading cause of death worldwide. Even though numerous circRNAs have been linked to the formation and progression of CAD, more circular RNA regulators still need to be uncovered in order to enhance the knowledge of the CAD regulatory networks. Microarray analysis was employed to identify aberrantly expressed circRNAs in vascular pathology, and the level of circ-Sirt6 in abnormally proliferated vascular smooth muscle cells (VSMCs) was detected by qRT-PCR. The effects of circ-Sirt6, IGF2BP2, and Sirt6 on the proliferation and migration of VSMCs were examined by EdU incorporation assay and migration assays. The interaction between circ-Sirt6, IGF2BP2, and Sirt6 was verified by RNA pull-down assay, RIP, FISH, and immunofluorescent staining. Dot blot assay and m6A-methylated RNA immunoprecipitation-qPCR were performed to confirm that circ-Sirt6 promoted m6A modification and stabilization of Sirt6 mRNA. According to our research, circ-Sirt6 expression was markedly downregulated in VSMCs treated with PDGF-BB and carotid artery balloon injury. The in vitro and in vivo proliferation and migration of VSMCs were inhibited by overexpression of circ-Sirt6. Mechanistically, circ-Sirt6 specifically binds to IGF2BP2 and promotes the stability and expression of Sirt6 mRNA by enhancing its m6A modification. Our findings highlight the importance of circ-Sirt6-mediated m6A in VSMC phenotype switching. Circ-Sirt6 may be a novel biological target for CAD.
Collapse
MESH Headings
- Sirtuins/genetics
- Sirtuins/metabolism
- RNA, Circular/genetics
- RNA, Circular/metabolism
- RNA-Binding Proteins/metabolism
- RNA-Binding Proteins/genetics
- Animals
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/cytology
- Humans
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Cell Proliferation
- Myocytes, Smooth Muscle/metabolism
- Cell Movement
- Mice
- Male
- RNA Stability
- Adenosine/analogs & derivatives
- Adenosine/metabolism
- Cells, Cultured
- Coronary Artery Disease/metabolism
- Coronary Artery Disease/genetics
- Coronary Artery Disease/pathology
Collapse
Affiliation(s)
- Chuanfeng Zhang
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong, P.R. China
| | - Jianing Wang
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong, P.R. China
| | - Peng Kong
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Key Laboratory of Vascular Biology of Hebei Province, Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Guangbin Sun
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong, P.R. China
| | - Hongfang Sun
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong, P.R. China
| | - Yuan Yu
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong, P.R. China
| |
Collapse
|
5
|
Lin A, Miano JM, Fisher EA, Misra A. Chronic inflammation and vascular cell plasticity in atherosclerosis. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1408-1423. [PMID: 39653823 DOI: 10.1038/s44161-024-00569-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 10/10/2024] [Indexed: 12/13/2024]
Abstract
Vascular smooth muscle cells, endothelial cells and macrophages undergo phenotypic conversions throughout atherosclerosis progression, both as a consequence of chronic inflammation and as subsequent drivers of it. The inflammatory hypothesis of atherosclerosis has been catapulted to the forefront of cardiovascular research as clinical trials have shown that anti-inflammatory therapy reduces adverse cardiovascular events. However, no current therapies have been specifically designed to target the phenotype of plaque cells. Fate mapping has revealed that plaque cells convert to detrimental and beneficial cell phenotypes during atherosclerosis, with cumulative evidence highlighting that vascular cell plasticity is intimately linked with plaque inflammation, ultimately impacting lesion stability. Here we review vascular cell plasticity during atherosclerosis in the context of the chronic inflammatory plaque microenvironment. We highlight the need to better understand how plaque cells behave during therapeutic intervention. We then propose modulating plaque cell phenotype as an unexplored therapeutic paradigm in the clinical setting.
Collapse
Affiliation(s)
- Alexander Lin
- Atherosclerosis and Vascular Remodelling Group, Heart Research Institute, Sydney, New South Wales, Australia
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, New South Wales, Australia
| | - Joseph M Miano
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Edward A Fisher
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Ashish Misra
- Atherosclerosis and Vascular Remodelling Group, Heart Research Institute, Sydney, New South Wales, Australia.
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
6
|
Madaudo C, Coppola G, Parlati ALM, Corrado E. Discovering Inflammation in Atherosclerosis: Insights from Pathogenic Pathways to Clinical Practice. Int J Mol Sci 2024; 25:6016. [PMID: 38892201 PMCID: PMC11173271 DOI: 10.3390/ijms25116016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/18/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
This comprehensive review explores the various scenarios of atherosclerosis, a systemic and chronic arterial disease that underlies most cardiovascular disorders. Starting from an overview of its insidious development, often asymptomatic until it reaches advanced stages, the review delves into the pathophysiological evolution of atherosclerotic lesions, highlighting the central role of inflammation. Insights into clinical manifestations, including heart attacks and strokes, highlight the disease's significant burden on global health. Emphasis is placed on carotid atherosclerosis, clarifying its epidemiology, clinical implications, and association with cognitive decline. Prevention strategies, lifestyle modifications, risk factor management, and nuanced antithrombotic treatment considerations are critical to managing cardiovascular complications, thus addressing a crucial aspect of cardiovascular health.
Collapse
Affiliation(s)
- Cristina Madaudo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Cardiology Unit, University of Palermo, University Hospital P. Giaccone, 90127 Palermo, Italy; (C.M.)
| | - Giuseppe Coppola
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Cardiology Unit, University of Palermo, University Hospital P. Giaccone, 90127 Palermo, Italy; (C.M.)
| | | | - Egle Corrado
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Cardiology Unit, University of Palermo, University Hospital P. Giaccone, 90127 Palermo, Italy; (C.M.)
| |
Collapse
|
7
|
Hassab AH, Hur DJ, Vallabhajosyula P, Tellides G, Assi R. Intimomedial tears of the aorta heal by smooth muscle cell-mediated fibrosis without atherosclerosis. JCI Insight 2024; 9:e172437. [PMID: 38592807 PMCID: PMC11141924 DOI: 10.1172/jci.insight.172437] [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: 05/19/2023] [Accepted: 04/03/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUNDDisease of the aorta varies from atherosclerosis to aneurysms, with complications including rupture, dissection, and poorly characterized limited tears. We studied limited tears without any mural hematoma, termed intimomedial tears, to gain insight into aortic vulnerability to excessive wall stresses. Our premise is that minimal injuries in aortas with sufficient medial resilience to prevent tear progression correspond to initial mechanisms leading to complete structural failure in aortas with significantly compromised medial resilience.METHODSIntimomedial tears were macroscopically identified in 9 of 108 ascending aortas after surgery and analyzed by histology and immunofluorescence confocal microscopy.RESULTSNonhemorrhagic, nonatheromatous tears correlated with advanced aneurysmal disease and most lacked distinctive symptoms or radiological signs. Tears traversed the intima and part of the subjacent media, while the resultant defects were partially or completely filled with neointima characterized by differentiated smooth muscle cells, scattered leukocytes, dense fibrosis, and absent elastic laminae despite tropoelastin synthesis. Healed lesions contained organized fibrin at tear edges without evidence of plasma and erythrocyte extravasation or lipid accumulation.CONCLUSIONThese findings suggest a multiphasic model of aortic wall failure in which primary lesions of intimomedial tears either heal if the media is sufficiently resilient or progress as dissection or rupture by medial delamination and tear completion, respectively. Moreover, mural incorporation of thrombus and cellular responses to injury, two historically important concepts in atheroma pathogenesis, contribute to vessel wall repair with adequate conduit function, but even together are not sufficient to induce atherosclerosis.FUNDINGNIH (R01-HL146723, R01-HL168473) and Yale Department of Surgery.
Collapse
Affiliation(s)
| | - David J. Hur
- Department of Internal Medicine (Cardiovascular Medicine), Yale School of Medicine, New Haven, Connecticut, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
| | | | - George Tellides
- Department of Surgery (Cardiac), and
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
- Program in Vascular Biology and Therapeutics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Roland Assi
- Department of Surgery (Cardiac), and
- Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut, USA
- Program in Vascular Biology and Therapeutics, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
8
|
Yan YX, Lu YK, Liu YH, Zhang J, Wang S, Dong J, Xiao HB. Identification of circular RNA hsa_circ_0034621 as a novel biomarker for carotid atherosclerosis and the potential function as a regulator of NLRP3 inflammasome. Atherosclerosis 2024; 391:117491. [PMID: 38471264 DOI: 10.1016/j.atherosclerosis.2024.117491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 02/13/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024]
Abstract
BACKGROUND AND AIMS NLRP3 inflammasome plays a key role in vascular inflammation and atherosclerosis. Circular RNAs (circRNAs) are involved in disease development by regulating gene expression, and have emerged as promising novel disease biomarkers. This study aimed to identify the NLRP3 inflammasome-associated circRNA biomarkers of carotid atherosclerosis. METHODS Based on the differential expression profiles of circRNAs in patients with carotid artery plaque (CAP) and healthy controls, hsa_circ_0043621, hsa_circ_0051995, and hsa_circ_0123388 were screened and validated using real-time quantitative polymerase chain reaction (RT-qPCR). Potential circRNA-miRNA-mRNA interactions were explored using a luciferase assay. The biological roles of the validated circRNAs were investigated in human umbilical vein endothelial cells (HUVECs) using Western blotting, transwell, and CCK-8 assays. Clinical significance was assessed using receiver operating characteristic (ROC) curves and logistic regression analysis. RESULTS The expression levels of all candidate circRNAs were significantly higher in patients with CAP than in controls (p<0.05), which was consistent with the results of the microarray analysis. Overexpression of hsa_circ_0043621 significantly increased the expression of NLRP3, induced migration of HUVECs, and inhibited cell proliferation. hsa_circ_0043621 demonstrated reasonable diagnostic accuracy for CAP detection and increased intima-media thickness (IMT). hsa_circ_0043621 upregulation was an independent predictor of an increased risk of CAP and increased IMT. CONCLUSIONS hsa_circ_0043621 is a valuable circulating biomarker of carotid atherosclerosis and may contribute to its pathogenesis by regulating the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Yu-Xiang Yan
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China; Municipal Key Laboratory of Clinical Epidemiology, Beijing, China.
| | - Ya-Ke Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China
| | - Yu-Hong Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China
| | - Jie Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China; Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Shuo Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China
| | - Jing Dong
- Health Management Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Huan-Bo Xiao
- Department of Preventive Medicine, Yanjing Medical College, Capital Medical University, Beijing, China
| |
Collapse
|
9
|
Guo Q, Jin Y, Chen X, Ye X, Shen X, Lin M, Zeng C, Zhou T, Zhang J. NF-κB in biology and targeted therapy: new insights and translational implications. Signal Transduct Target Ther 2024; 9:53. [PMID: 38433280 PMCID: PMC10910037 DOI: 10.1038/s41392-024-01757-9] [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: 10/19/2023] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 03/05/2024] Open
Abstract
NF-κB signaling has been discovered for nearly 40 years. Initially, NF-κB signaling was identified as a pivotal pathway in mediating inflammatory responses. However, with extensive and in-depth investigations, researchers have discovered that its role can be expanded to a variety of signaling mechanisms, biological processes, human diseases, and treatment options. In this review, we first scrutinize the research process of NF-κB signaling, and summarize the composition, activation, and regulatory mechanism of NF-κB signaling. We investigate the interaction of NF-κB signaling with other important pathways, including PI3K/AKT, MAPK, JAK-STAT, TGF-β, Wnt, Notch, Hedgehog, and TLR signaling. The physiological and pathological states of NF-κB signaling, as well as its intricate involvement in inflammation, immune regulation, and tumor microenvironment, are also explicated. Additionally, we illustrate how NF-κB signaling is involved in a variety of human diseases, including cancers, inflammatory and autoimmune diseases, cardiovascular diseases, metabolic diseases, neurological diseases, and COVID-19. Further, we discuss the therapeutic approaches targeting NF-κB signaling, including IKK inhibitors, monoclonal antibodies, proteasome inhibitors, nuclear translocation inhibitors, DNA binding inhibitors, TKIs, non-coding RNAs, immunotherapy, and CAR-T. Finally, we provide an outlook for research in the field of NF-κB signaling. We hope to present a stereoscopic, comprehensive NF-κB signaling that will inform future research and clinical practice.
Collapse
Affiliation(s)
- Qing Guo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yizi Jin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinyu Chen
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Stem Cell Research Center, Shanghai Cancer Institute & Department of Urology, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, PR China
| | - Xiaomin Ye
- Department of Cardiology, the First Affiliated Hospital of Sun Yat-Sen University, 58 Zhongshan 2nd Road, Guangzhou, 510080, China
| | - Xin Shen
- Department of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingxi Lin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Cheng Zeng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Teng Zhou
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
10
|
Han S, Nie C, Wang C, Song M, Li J, Cui X, Yang Q, Li Y, Chen Y, Li Q, Cai W, Weng X, Wang Y, Zhu X. Shenlian extract improves atherosclerosis by relieving adventitial inflammation. JOURNAL OF ETHNOPHARMACOLOGY 2024; 320:117339. [PMID: 37866468 DOI: 10.1016/j.jep.2023.117339] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shenlian (SL) extract, a Chinese medicinal compound mainly pointing at inflammation response of atherosclerosis, is composed of Salvia miltiorrhizae Bunge and Andrographis paniculata (Burm.f.) Nees. Salvia miltiorrhizae Bunge has been reported to activate blood to remove stasis, while another herb, Andrographis paniculata (Burm.f.) Nees, has been revealed to clear endogenous heat toxins. The anti-atherosclerotic effects of these two herbs have been reported closely relating to inflammation. However, from the point of view of adventitial inflammation, the in-depth study of SL extract in anti-atherosclerotic effects by relieving adventitial inflammation is still unknown. AIM OF THE STUDY To explore the effects of adventitial inflammation in atherosclerosis progression and if SL extract could reverse the process. MATERIALS AND METHODS A novel atherosclerosis model based on adventitial inflammation was established. High-fat diet-fed ApoE-/- mice were implanted a cotton thread soaked with LPS on the right common carotid artery (RCCA). Meanwhile, three time points were set (week 2, 4, and 12) to accurately evaluate the effect of SL extract on the whole process of atherosclerosis with adventitial inflammation. The pathological changes of phenotype transformation of VAFs, vascular cell proliferation and collagen synthesis were observed dynamically by immunohistochemistry (IHC), BrdU method and sirius red staining. Then primary VAFs were stimulated by LPS to mirror the process of adventitial inflammation in vitro. The VAFS phenotype conversion and its function alterations including proliferation, migration, inflammatory secretion was assessed. Finally, we established a co-culture model of activated VAFs and vascular smooth muscle cells (VSMCs) to observe the impacts of activated VAFs on phenotype transformation and migration of VSMCs. RESULTS SL extract improved atherosclerosis progression by reducing lipid content, adventitial inflammation and plaque formation. HE results showed sham-operated group (Sham) appeared light infiltrated inflammation only in adventitia at week 2, and the degree of inflammation infiltrated in model was more severe than that in Sham at week 2, 4, and 12. At week 12, the sham and model group showed evidently thickened media and intima. The phenotypic transformation, proliferation and migration of vascular adventitial fibroblasts (VAFs) as well as inflammatory secretion enhanced remarkably in vivo and vitro, but SL extract reversed these changes. Moreover, SL extract downregulated JAK2-STAT3-MMP2 signal pathway. The VSMCs transformed from contractile phenotype into synthetic phenotype and the migration of VSMCs increased after co-culture with activated VAFs. In contrast, SL extract could suppress theses effects. CONCLUSIONS Taken together, atherosclerotic inflammation could be a "outside-in" signaling. Adventitial inflammation not only accelerated intimal plaque formation in atherosclerosis, but also worsened the degree of vascular lesion. And SL extract improved atherosclerosis by relieving adventitial inflammation, and the underlying mechanisms could be associated with curbing phenotypic transformation, proliferation and migration of VAFs and VSMCs.
Collapse
Affiliation(s)
- Shuxian Han
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Chunxia Nie
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Chunmiao Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Min Song
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jingjing Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Xihe Cui
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qing Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yujie Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Ying Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qi Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Weiyan Cai
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Xiaogang Weng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yajie Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Xiaoxin Zhu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
11
|
Gillard BK, Rosales C, Gotto AM, Pownall HJ. The pathophysiology of excess plasma-free cholesterol. Curr Opin Lipidol 2023; 34:278-286. [PMID: 37732779 PMCID: PMC10624414 DOI: 10.1097/mol.0000000000000899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
PURPOSE OF REVIEW Several large studies have shown increased mortality due to all-causes and to atherosclerotic cardiovascular disease. In most clinical settings, plasma HDL-cholesterol is determined as a sum of free cholesterol and cholesteryl ester, two molecules with vastly different metabolic itineraries. We examine the evidence supporting the concept that the pathological effects of elevations of plasma HDL-cholesterol are due to high levels of the free cholesterol component of HDL-C. RECENT FINDINGS In a small population of humans, a high plasma HDL-cholesterol is associated with increased mortality. Similar observations in the HDL-receptor deficient mouse (Scarb1 -/- ), a preclinical model of elevated HDL-C, suggests that the pathological component of HDL in these patients is an elevated plasma HDL-FC. SUMMARY Collective consideration of the human and mouse data suggests that clinical trials, especially in the setting of high plasma HDL, should measure free cholesterol and cholesteryl esters and not just total cholesterol.
Collapse
Affiliation(s)
- Baiba K. Gillard
- Center for Bioenergetics, Houston Methodist, Houston, Texas
- Weill Cornell Medicine, New York, New York, USA
| | - Corina Rosales
- Center for Bioenergetics, Houston Methodist, Houston, Texas
- Weill Cornell Medicine, New York, New York, USA
| | - Antonio M. Gotto
- Center for Bioenergetics, Houston Methodist, Houston, Texas
- Weill Cornell Medicine, New York, New York, USA
| | - Henry J. Pownall
- Center for Bioenergetics, Houston Methodist, Houston, Texas
- Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
12
|
Francis GA. The Greatly Under-Represented Role of Smooth Muscle Cells in Atherosclerosis. Curr Atheroscler Rep 2023; 25:741-749. [PMID: 37665492 PMCID: PMC10564813 DOI: 10.1007/s11883-023-01145-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2023] [Indexed: 09/05/2023]
Abstract
PURPOSE OF REVIEW This article summarizes previous and recent research on the fundamental role of arterial smooth muscle cells (SMCs) as drivers of initial and, along with macrophages, later stages of human atherosclerosis. RECENT FINDINGS Studies using human tissues and SMC lineage-tracing mice have reinforced earlier observations that SMCs drive initial atherogenesis in humans and contribute a multitude of phenotypes including foam cell formation hitherto attributed primarily to macrophages in atherosclerosis. Arterial smooth muscle cells (SMCs) are the primary cell type in human pre-atherosclerotic intima and are responsible for the retention of lipoproteins that drive the development of atherosclerosis. Despite this, images of atherogenesis still depict the process as initially devoid of SMCs, primarily macrophage driven, and indicate only relatively minor roles such as fibrous cap formation to intimal SMCs. This review summarizes historical and recent observations regarding the importance of SMCs in the formation of a pre-atherosclerotic intima, initial and later foam cell formation, and the phenotypic changes that give rise to multiple different roles for SMCs in human and mouse lesions. Potential SMC-specific therapies in atherosclerosis are presented.
Collapse
Affiliation(s)
- Gordon A Francis
- Centre for Heart Lung Innovation, Providence Research, St. Paul's Hospital, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
13
|
Oglesby IK, Slattery D, Glynn N, Gupta S, Duggan K, Cuesta M, Dunne E, Garrahy A, Toner S, Kenny D, Agha A. The modulation of platelet function by growth hormone in growth hormone deficient Hypopituitary patients. BMC Endocr Disord 2023; 23:197. [PMID: 37705005 PMCID: PMC10500895 DOI: 10.1186/s12902-023-01448-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 08/31/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND Growth hormone deficiency (GHD) has been implicated in increased cardiovascular and cerebrovascular disease risk seen in hypopituitarism, however the mechanism remains speculative. We hypothesise that platelet abnormalities may play a contributory role. Herein we examined platelet behaviour in GHD hypopituitary patients, pre- and post-growth hormone (GH) replacement. METHODS This study utilizes a physiological flow-based assay to quantify platelet function in whole blood from patient cohorts under arterial shear. Thirteen GH Naïve hypopituitary adults with GHD and thirteen healthy matched controls were studied. Patients were assessed before and after GH treatment. All other pituitary replacements were optimised before the study. In addition to a full endocrine profile, whole blood was labelled and perfused over immobilised von Willibrand factor (vWF). Seven parameters of dynamic platelet-vWF interactions were recorded using digital image microscopy and analysed by customised platelet tracking software. RESULTS We found a significantly altered profile of platelet-vWF interactions in GHD individuals compared to healthy controls. Specifically, we observed a marked increase in platelets shown to form associations such as tethering, rolling and adherence to immobilized vWF, which were reduced post GH treatment. Speed and distance platelets travelled across vWF was similar between controls and pre-therapy GHD patients, however, this was considerably increased post treatment. This may indicate reduced platelet signaling resulting in less stable adhesion of platelets post GH treatment. CONCLUSIONS Taken together observed differences in platelet behaviour may contribute to an increased risk of thrombosis in GHD which can in part be reversed by GH therapy.
Collapse
Affiliation(s)
- Irene K Oglesby
- Irish Centre for Vascular Biology and Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Department of Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - David Slattery
- Department of Endocrinology, Beaumont Hospital, Dublin 9, Dublin, Ireland
| | - Nigel Glynn
- Department of Endocrinology, Beaumont Hospital, Dublin 9, Dublin, Ireland
| | - Saket Gupta
- Department of Endocrinology, Beaumont Hospital, Dublin 9, Dublin, Ireland
| | - Karen Duggan
- Department of Endocrinology, Beaumont Hospital, Dublin 9, Dublin, Ireland
| | - Martin Cuesta
- Department of Endocrinology, Beaumont Hospital, Dublin 9, Dublin, Ireland
| | - Eimear Dunne
- Irish Centre for Vascular Biology and Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Aoife Garrahy
- Department of Endocrinology, Beaumont Hospital, Dublin 9, Dublin, Ireland
| | - Siobhan Toner
- Department of Endocrinology, Beaumont Hospital, Dublin 9, Dublin, Ireland
| | - Dermot Kenny
- Irish Centre for Vascular Biology and Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Amar Agha
- Department of Endocrinology, Beaumont Hospital, Dublin 9, Dublin, Ireland.
| |
Collapse
|
14
|
Zhang Z, Li L, Shi H, Chen B, Li X, Zhang Y, Liu F, Wei W, Zhou Y, Liu K, Xia W, Gu X, Huang J, Tu S, Yin C, Shao A, Jiang L. Role of Circular RNAs in Atherosclerosis through Regulation of Inflammation, Cell Proliferation, Migration, and Apoptosis: Focus on Atherosclerotic Cerebrovascular Disease. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1461. [PMID: 37629751 PMCID: PMC10456328 DOI: 10.3390/medicina59081461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/29/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023]
Abstract
Atherosclerosis (AS) is a disease dangerous to human health and the main pathological cause of ischemic cardiovascular diseases. Although its pathogenesis is not fully understood, numerous basic and clinical studies have shown that AS is a chronic inflammatory disease existing in all stages of atherogenesis. It may be a common link or pathway in the pathogenesis of multiple atherogenic factors. Inflammation is associated with AS complications, such as plaque rupture and ischemic cerebral infarction. In addition to inflammation, apoptosis plays an important role in AS. Apoptosis is a type of programmed cell death, and different apoptotic cells have different or even opposite roles in the process of AS. Unlike linear RNA, circular RNA (circRNA) a covalently closed circular non-coding RNA, is stable and can sponge miRNA, which can affect the stages of AS by regulating downstream pathways. Ultimately, circRNAs play very important roles in AS by regulating inflammation, apoptosis, and some other mechanisms. The study of circular RNAs can provide new ideas for the prediction, prevention, and treatment of AS.
Collapse
Affiliation(s)
- Zheng Zhang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
| | - Lingfei Li
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| | - Huanqing Shi
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
| | - Biao Chen
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
| | - Xiaoqin Li
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
| | - Yuyao Zhang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
| | - Fei Liu
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| | - Wan Wei
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| | - Yongji Zhou
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| | - Keqin Liu
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| | - Wenqing Xia
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| | - Xin Gu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
| | - Jinyu Huang
- Department of Cardiology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China;
| | - Sheng Tu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310006, China;
| | - Congguo Yin
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Disease, Hangzhou 310009, China
| | - Lin Jiang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Z.Z.); (H.S.); (B.C.); (X.L.); (Y.Z.); (X.G.)
- Department of Neurology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; (L.L.); (F.L.); (W.W.); (Y.Z.); (K.L.); (W.X.)
| |
Collapse
|
15
|
Han S, Cai L, Chen P, Kuang W. A study of the correlation between stroke and gut microbiota over the last 20years: a bibliometric analysis. Front Microbiol 2023; 14:1191758. [PMID: 37350780 PMCID: PMC10282156 DOI: 10.3389/fmicb.2023.1191758] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023] Open
Abstract
Purpose This study intends to uncover a more thorough knowledge structure, research hotspots, and future trends in the field by presenting an overview of the relationship between stroke and gut microbiota in the past two decades. Method Studies on stroke and gut microbiota correlations published between 1st January 2002 and 31st December 2021 were retrieved from the Web of Science Core Collection and then visualized and scientometrically analyzed using CiteSpace V. Results A total of 660 papers were included in the study, among which the United States, the United Kingdom, and Germany were the leading research centers. Cleveland Clinic, Southern Medical University, and Chinese Academy of Science were the top three institutions. The NATURE was the most frequently co-cited journal. STANLEY L HAZEN was the most published author, and Tang WHW was the most cited one. The co-occurrence analysis revealed eight clusters (i.e., brain-gut microbiota axis, fecal microbiome transplantation, gut microbiota, hypertension, TMAO, ischemic stroke, neuroinflammation, atopobiosis). "gut microbiota," "Escherichia coli," "cardiovascular disease," "risk," "disease," "ischemic stroke," "stroke," "metabolism," "inflammation," and "phosphatidylcholine" were the most recent keyword explosions. Conclusion Findings suggest that in the next 10 years, the number of publications produced annually may increase significantly. Future research trends tend to concentrate on the mechanisms of stroke and gut microbiota, with the inflammation and immunological mechanisms, TMAO, and fecal transplantation as hotspots. And the relationship between these mechanisms and a particular cardiovascular illness may also be a future research trend.
Collapse
Affiliation(s)
- Shengnan Han
- Clinical Medical College of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Longhui Cai
- First School of Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peipei Chen
- School of Medical Technology, Qiqihar Medical College, Qiqihar, Heilongjiang, China
| | - Weihong Kuang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Dongguan, China
| |
Collapse
|
16
|
Cheng X, Chen X, Zhang M, Wan Y, Ge S, Cheng X. Sparcl1 and Atherosclerosis. J Inflamm Res 2023; 16:2121-2127. [PMID: 37220502 PMCID: PMC10200116 DOI: 10.2147/jir.s406907] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/03/2023] [Indexed: 05/25/2023] Open
Abstract
Atherosclerosis and its complications constitute some of the major diseases affecting humans worldwide. A core component of atherogenesis is endothelial cell damage and dysfunction, which also includes factors such as adhesion and proliferation of various cells. Multiple studies have shown that atherosclerosis and cancer share a common pathophysiological process and exhibit a degree of similarity. Sparcl-1 is a cysteine-rich secretory stromal cell protein present in the extracellular matrix and belongs to the Sparc family of proteins. Its role in tumor development has been widely investigated; however, its role in cardiovascular diseases has rarely been studied. Sparcl-1 is considered an oncogene correlated with the regulation of cell adhesion, migration, and proliferation and is also related to blood vessel integrity. In this review, the potential link between Sparcl-1 and atherosclerosis development is investigated, and recommendations on future research on the role of Sparcl-1 in atherogenesis are provided.
Collapse
Affiliation(s)
- Xu Cheng
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People’s Republic of China
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, Anhui, 230022, People’s Republic of China
| | - Xinyan Chen
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, Anhui, 230022, People’s Republic of China
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People’s Republic of China
| | - Min Zhang
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People’s Republic of China
| | - Yufeng Wan
- Department of Otolaryngology-Head Neck Surgery, the Affiliated Chaohu Hospital of Anhui Medical University, Hefei, Anhui, 238001, People’s Republic of China
| | - Shenglin Ge
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People’s Republic of China
| | - Xiaowen Cheng
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, Anhui, 230022, People’s Republic of China
- Department of Clinical Laboratory, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, People’s Republic of China
- Anhui Province Key Laboratory of Translational Cancer Research, Bengbu Medical College, Bengbu, Anhui, 233030, People’s Republic of China
| |
Collapse
|
17
|
Chen CY, Leu HB, Wang SC, Tsai SH, Chou RH, Lu YW, Tsai YL, Kuo CS, Huang PH, Chen JW, Lin SJ. Inhibition of Trimethylamine N-Oxide Attenuates Neointimal Formation Through Reduction of Inflammasome and Oxidative Stress in a Mouse Model of Carotid Artery Ligation. Antioxid Redox Signal 2023; 38:215-233. [PMID: 35713239 DOI: 10.1089/ars.2021.0115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Aims: Trimethylamine-N-oxide (TMAO) is a metabolite generated from dietary choline, betaine, and l-carnitine, after their oxidization in the liver. TMAO has been identified as a novel independent risk factor for atherosclerosis through the induction of vascular inflammation. However, the effect of TMAO on neointimal formation in response to vascular injury remains unclear. Results: This study was conducted using a murine model of acutely disturbed flow-induced atherosclerosis induced by partial carotid artery ligation. 3,3-Dimethyl-1-butanol (DMB) was used to reduce TMAO concentrations. Wild-type mice were divided into four groups [regular diet, high-TMAO diet, high-choline diet, and high-choline diet+DMB] to investigate the effects of TMAO elevation and its inhibition by DMB. Mice fed high-TMAO and high-choline diets had significantly enhanced neointimal hyperplasia and advanced plaques, elevated arterial elastin fragmentation, increased macrophage infiltration and inflammatory cytokine secretion, and enhanced activation of nuclear factor (NF)-κB, the NLRP3 inflammasome, and endoplasmic reticulum (ER) stress relative to the control group. Mice fed high-choline diets with DMB treatment exhibited attenuated flow-induced atherosclerosis, inflammasome expression, ER stress, and reactive oxygen species expression. Human aortic smooth muscle cells (HASMCs) were used to investigate the mechanism of TMAO-induced injury. The HASMCs were treated with TMAO with or without an ER stress inhibitor to determine whether inhibition of ER stress modulates the TMAO-induced inflammatory response. Innovation: This study demonstrates that TMAO regulates vascular remodeling via ER stress. Conclusion: Our findings demonstrate that TMAO elevation promotes disturbed flow-induced atherosclerosis and that DMB administration mitigates vascular remodeling, suggesting a rationale for a TMAO-targeted strategy for the treatment of atherosclerosis. Antioxid. Redox Signal. 38, 215-233.
Collapse
Affiliation(s)
- Chi-Yu Chen
- Division of Cardiology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hsin-Bang Leu
- Division of Cardiology, Taipei Veterans General Hospital, Taipei, Taiwan.,Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shen-Chih Wang
- Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.,Healthcare and Services Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shih-Hung Tsai
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ruey-Hsing Chou
- Division of Cardiology, Taipei Veterans General Hospital, Taipei, Taiwan.,Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ya-Wen Lu
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Lin Tsai
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chin-Sung Kuo
- Division of Cardiology, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Po-Hsun Huang
- Division of Cardiology, Taipei Veterans General Hospital, Taipei, Taiwan.,Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Pharmacology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jaw-Wen Chen
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Anesthesiology, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shing-Jong Lin
- Division of Cardiology, Taipei Veterans General Hospital, Taipei, Taiwan.,Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan.,Division of Cardiology, Heart Center, Cheng-Hsin General Hospital, Taipei, Taiwan
| |
Collapse
|
18
|
Su C, Lu Y, Wang Z, Guo J, Hou Y, Wang X, Qin Z, Gao J, Sun Z, Dai Y, Liu Y, Liu G, Xian X, Cui X, Zhang J, Tang J. Atherosclerosis: The Involvement of Immunity, Cytokines and Cells in Pathogenesis, and Potential Novel Therapeutics. Aging Dis 2022:AD.2022.1208. [PMID: 37163428 PMCID: PMC10389830 DOI: 10.14336/ad.2022.1208] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 12/08/2022] [Indexed: 05/12/2023] Open
Abstract
As a leading contributor to coronary artery disease (CAD) and stroke, atherosclerosis has become one of the major cardiovascular diseases (CVD) negatively impacting patients worldwide. The endothelial injury is considered to be the initial step of the development of atherosclerosis, resulting in immune cell migration and activation as well as inflammatory factor secretion, which further leads to acute and chronic inflammation. In addition, the inflammation and lipid accumulation at the lesions stimulate specific responses from different types of cells, contributing to the pathological progression of atherosclerosis. As a result, recent studies have focused on using molecular biological approaches such as gene editing and nanotechnology to mediate cellular response during atherosclerotic development for therapeutic purposes. In this review, we systematically discuss inflammatory pathogenesis during the development of atherosclerosis from a cellular level with a focus on the blood cells, including all types of immune cells, together with crucial cells within the blood vessel, such as smooth muscle cells and endothelial cells. In addition, the latest progression of molecular-cellular based therapy for atherosclerosis is also discussed. We hope this review article could be beneficial for the clinical management of atherosclerosis.
Collapse
Affiliation(s)
- Chang Su
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Yongzheng Lu
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Zeyu Wang
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Jiacheng Guo
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Yachen Hou
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Xiaofang Wang
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Zhen Qin
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Jiamin Gao
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Zhaowei Sun
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Yichen Dai
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Yu Liu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Guozhen Liu
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Xunde Xian
- Institute of Cardiovascular Sciences, Peking University, Beijing, China
| | - Xiaolin Cui
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Jinying Zhang
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| | - Junnan Tang
- Department of Cardiology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, Henan, China
- Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, Henan, China
| |
Collapse
|
19
|
Liu L, Cui Q, Song J, Yang Y, Zhang Y, Qi J, Zhao J. Hydroxysafflower Yellow A Inhibits Vascular Adventitial Fibroblast Migration via NLRP3 Inflammasome Inhibition through Autophagy Activation. Int J Mol Sci 2022; 24:ijms24010172. [PMID: 36613617 PMCID: PMC9820330 DOI: 10.3390/ijms24010172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/08/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Inflammation is closely associated with progression of vascular remodeling. The NLRP3 inflammasome is the key molecule that promotes vascular remodeling via activation of vascular adventitia fibroblast (VAF) proliferation and differentiation. VAFs have a vital effect on vascular remodeling that could be improved using hydroxysafflower yellow A (HSYA). However, whether HSYA ameliorates vascular remodeling through inhibition of NLRP3 inflammasome activation has not been explored in detail. Here, we cultured primary VAFs and analyzed the migration of VAFs induced by angiotensin II (ANG II) to determine the potential effects and mechanism of HSYA on VAF migration. The results thereof showed that HSYA remarkably inhibited ANG II-induced VAF migration, NLRP3 inflammasome activation, and the TLR4/NF-κB signaling pathway in a dose-dependent manner. In addition, it is worth noting that LPS promoted ANG II-induced VAF migration and NLRP3 inflammasome assembly, which could be significantly reversed using HSYA. Moreover, HSYA could be used to inhibit NLRP3 inflammasome activation by promoting autophagy. In conclusion, HSYA could inhibit ANG II-induced VAF migration through autophagy activation and inhibition of NLRP3 inflammasome activation through the TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Lin Liu
- College of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Qingzhuo Cui
- College of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Traditional Chinese Medicine Processing Technology Innovation Center of Hebei Province, College of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Junna Song
- College of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yang Yang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Yixin Zhang
- College of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Jiapeng Qi
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Jingshan Zhao
- College of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Traditional Chinese Medicine Processing Technology Innovation Center of Hebei Province, College of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
- Correspondence:
| |
Collapse
|
20
|
Zhang G, Liu Z, Deng J, Liu L, Li Y, Weng S, Guo C, Zhou Z, Zhang L, Wang X, Liu G, Guo J, Bai J, Wang Y, Du Y, Li TS, Tang J, Zhang J. Smooth muscle cell fate decisions decipher a high-resolution heterogeneity within atherosclerosis molecular subtypes. J Transl Med 2022; 20:568. [PMID: 36474294 PMCID: PMC9724432 DOI: 10.1186/s12967-022-03795-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Mounting evidence has revealed the dynamic variations in the cellular status and phenotype of the smooth muscle cell (SMC) are vital for shaping the atherosclerotic plaque microenvironment and ultimately mapping onto heterogeneous clinical outcomes in coronary artery disease. Currently, the underlying clinical significance of SMC evolutions remains unexplored in atherosclerosis. METHODS The dissociated cells from diseased segments within the right coronary artery of four cardiac transplant recipients and 1070 bulk samples with atherosclerosis from six bulk cohorts were retrieved. Following the SMC fate trajectory reconstruction, the MOVICS algorithm integrating the nearest template prediction was used to develop a stable and robust molecular classification. Subsequently, multi-dimensional potential biological implications, molecular features, and cell landscape heterogeneity among distinct clusters were decoded. RESULTS We proposed an SMC cell fate decision signature (SCFDS)-based atherosclerosis stratification system and identified three SCFDS subtypes (C1-C3) with distinguishing features: (i) C1 (DNA-damage repair type), elevated base excision repair (BER), DNA replication, as well as oxidative phosphorylation status. (ii) C2 (immune-activated type), stronger immune activation, hyper-inflammatory state, the complex as well as varied lesion microenvironment, advanced stage, the most severe degree of coronary stenosis severity. (iii) C3 (stromal-rich type), abundant fibrous content, stronger ECM metabolism, immune-suppressed microenvironment. CONCLUSIONS This study uncovered atherosclerosis complex cellular heterogeneity and a differentiated hierarchy of cell populations underlying SMC. The novel high-resolution stratification system could improve clinical outcomes and facilitate individualized management.
Collapse
Affiliation(s)
- Ge Zhang
- grid.412633.10000 0004 1799 0733Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China ,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, 450052 Henan China ,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052 Henan China
| | - Zaoqu Liu
- grid.412633.10000 0004 1799 0733Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Jinhai Deng
- grid.13097.3c0000 0001 2322 6764Richard Dimbleby Laboratory of Cancer Research, School of Cancer & Pharmaceutical Sciences, King’s College London, London, UK
| | - Long Liu
- grid.412633.10000 0004 1799 0733Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Yu Li
- grid.260463.50000 0001 2182 8825Medical College, Nanchang University, Nanchang, 330006 Jiangxi China
| | - Siyuan Weng
- grid.412633.10000 0004 1799 0733Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Chunguang Guo
- grid.412633.10000 0004 1799 0733Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan China
| | - Zhaokai Zhou
- grid.412633.10000 0004 1799 0733Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Li Zhang
- grid.412633.10000 0004 1799 0733Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China ,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, 450052 Henan China ,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052 Henan China
| | - Xiaofang Wang
- grid.412633.10000 0004 1799 0733Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China ,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, 450052 Henan China ,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052 Henan China
| | - Gangqiong Liu
- grid.412633.10000 0004 1799 0733Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China ,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, 450052 Henan China ,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052 Henan China
| | - Jiacheng Guo
- grid.412633.10000 0004 1799 0733Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China ,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, 450052 Henan China ,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052 Henan China
| | - Jing Bai
- grid.412633.10000 0004 1799 0733Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China ,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, 450052 Henan China ,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052 Henan China
| | - Yunzhe Wang
- grid.412633.10000 0004 1799 0733Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China ,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, 450052 Henan China ,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052 Henan China
| | - Youyou Du
- grid.412633.10000 0004 1799 0733Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China ,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, 450052 Henan China ,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052 Henan China
| | - Tao-Sheng Li
- grid.174567.60000 0000 8902 2273Department of Stem Cell Biology, Atomic Bomb Diseases Institute, Nagasaki University, Nagasaki, 852-8523 Japan
| | - Junnan Tang
- grid.412633.10000 0004 1799 0733Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China ,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, 450052 Henan China ,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052 Henan China
| | - Jinying Zhang
- grid.412633.10000 0004 1799 0733Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, No. 1 Eastern Jianshe Road, Zhengzhou, 450052 Henan China ,Henan Province Key Laboratory of Cardiac Injury and Repair, Zhengzhou, 450052 Henan China ,Henan Province Clinical Research Center for Cardiovascular Diseases, Zhengzhou, 450052 Henan China
| |
Collapse
|
21
|
Sesorova IS, Sesorov VV, Soloviev PB, Lakunin KY, Dimov ID, Mironov AA. Role of Endothelial Regeneration and Overloading of Enterocytes with Lipids in Capturing of Lipoproteins by Basement Membrane of Rat Aortic Endothelium. Biomedicines 2022; 10:2858. [PMID: 36359378 PMCID: PMC9687266 DOI: 10.3390/biomedicines10112858] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/11/2022] [Accepted: 10/27/2022] [Indexed: 10/29/2023] Open
Abstract
Atherosclerosis is a complex non-monogenic disease related to endothelial damage in elastic-type arteries and incorrect feeding. Here, using cryodamage of endothelial cells (ECs) of rat abdominal aorta, we examined the role of the EC basement membrane (BM) for re-endothelization endothelial regeneration and its ability to capture low density lipoproteins (LDLs). Regeneration of endothelium induced thickening of the ECBM. Secretion of the BM components occurred in the G2-phase. Multiple regenerations, as well as arterial hypertension and aging, also led to the thickening of the BM. Under these conditions, the speed of re-endothelialization increased. The thick BM captured more LDLs. LDLs formed after overloading of rats with lipids acquired higher affinity to the BM, presumably due to the prolonged transport of chylomicrons through neuraminidase-positive endo-lysosomes. These data provide new molecular and cellular mechanisms of atherogenesis.
Collapse
Affiliation(s)
- Irina S. Sesorova
- Department of Anatomy, Ivanovo State Medical Academy, 153012 Ivanovo, Russia
| | - Vitaly V. Sesorov
- Department of Anatomy, Ivanovo State Medical Academy, 153012 Ivanovo, Russia
| | - Pavel B. Soloviev
- Department of Pathological Anatomy, Ivanovo State Medical Academy, 153012 Ivanovo, Russia
| | | | - Ivan D. Dimov
- Department of Anatomy, Saint Petersburg State Pediatric Medical University, 194100 Saint Petersburg, Russia
| | - Alexander A. Mironov
- Italian Foundation for Cancer Research Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| |
Collapse
|
22
|
Yin Z, Zhang J, Xu S, Liu J, Xu Y, Yu J, Zhao M, Pan W, Wang M, Wan J. The role of semaphorins in cardiovascular diseases: Potential therapeutic targets and novel biomarkers. FASEB J 2022; 36:e22509. [PMID: 36063107 DOI: 10.1096/fj.202200844r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/24/2022] [Accepted: 08/09/2022] [Indexed: 12/17/2022]
Abstract
Semaphorins (Semas), which belongs to the axonal guidance molecules, include 8 classes and could affect axon growth in the nervous system. Recently, semaphorins were found to regulate other pathophysiological processes, such as immune response, oncogenesis, tumor angiogenesis, and bone homeostasis, through binding with their plexin and neuropilin receptors. In this review, we summarized the detailed role of semaphorins and their receptors in the pathological progression of various cardiovascular diseases (CVDs), highlighting that semaphorins may be potential therapeutic targets and novel biomarkers for CVDs.
Collapse
Affiliation(s)
- Zheng Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Shuwan Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Junping Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wei Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
23
|
Zheng Y, Xu L, Dong N, Li F. NLRP3 inflammasome: The rising star in cardiovascular diseases. Front Cardiovasc Med 2022; 9:927061. [PMID: 36204568 PMCID: PMC9530053 DOI: 10.3389/fcvm.2022.927061] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 08/18/2022] [Indexed: 11/21/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the prevalent cause of mortality around the world. Activation of inflammasome contributes to the pathological progression of cardiovascular diseases, including atherosclerosis, abdominal aortic aneurysm, myocardial infarction, dilated cardiomyopathy, diabetic cardiomyopathy, heart failure, and calcific aortic valve disease. The nucleotide oligomerization domain-, leucine-rich repeat-, and pyrin domain-containing protein 3 (NLRP3) inflammasome plays a critical role in the innate immune response, requiring priming and activation signals to provoke the inflammation. Evidence shows that NLRP3 inflammasome not only boosts the cleavage and release of IL-1 family cytokines, but also leads to a distinct cell programmed death: pyroptosis. The significance of NLRP3 inflammasome in the CVDs-related inflammation has been extensively explored. In this review, we summarized current understandings of the function of NLRP3 inflammasome in CVDs and discussed possible therapeutic options targeting the NLRP3 inflammasome.
Collapse
|
24
|
Chen C, Lu T, Wu Z, Xie X, Liu Y, Huang C, Liu Y. A proteomics analysis of neointima formation on decellularized vascular grafts reveals regenerative alterations in protein signature running head: Proteomics analysis of neointima formation. Front Bioeng Biotechnol 2022; 10:894956. [PMID: 36406232 PMCID: PMC9673820 DOI: 10.3389/fbioe.2022.894956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 08/08/2022] [Indexed: 11/25/2022] Open
Abstract
Background: Neointima formation contributes to vascular grafts stenosis and thrombosis. It is a complex reaction that plays a significant role in the performance of vascular grafts. Despite its critical implications, little is known about the mechanisms underlying neointima formation. This study compares neointima proteome in different stages and plasma samples. Methods: Heterogenous acellular native arteries were implanted as abdominal aortic interposition grafts in a rabbit model. Grafts were harvested at 0.5, 1, 4, 6, 7, 14, 21, and 28 days post-surgery for histological and proteomic analysis of the neointima. Results: Histological examination showed a transformed morphological pattern and components, including serum proteins, inflammatory cells, and regenerative cells. Proteomics analysis of the neointima showed distinct characteristics after 14 days of implantation compared to early implantation. Early changes in the neointima samples were proteins involved in acute inflammation and thrombosis, followed by the accumulation of extracellular matrix (ECM) proteins. A total of 110 proteins were found to be differentially expressed in later samples of neointima compared to early controls. The enriched pathways were mainly protein digestion and adsorption, focal adhesion, PI3K-Akt signaling pathway, and ECM-receptor interaction in the late stage. All distributions of proteins in the neointima are different compared to plasma. Conclusion: The biological processes of neointima formation at different stages identified with proteome found developmental characteristics of vascular structure on a decellularized small vascular graft, and significant differences were identified by proteomics in the neointima of early-stage and late-stage after implantation. In the acute unstable phase, the loose and uniform neointima was mainly composed of plasma proteins and inflammatory cells. However, in the relatively stable later stage, the most notable results were an up-regulation of ECM components. The present study demonstrates an interaction between biological matter and vascular graft, provides insights into biological process changes of neointima and facilitates the construction of a functional bioengineered small vascular graft for future clinical applications.
Collapse
Affiliation(s)
- Chunyang Chen
- Department of Cardiovascular surgery, Second Xiangya Hospital of Central South University, Changsha, China
- Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, China
| | - Ting Lu
- Department of Cardiovascular surgery, Second Xiangya Hospital of Central South University, Changsha, China
- Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, China
| | - Zhongshi Wu
- Department of Cardiovascular surgery, Second Xiangya Hospital of Central South University, Changsha, China
- Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, China
| | - Xinlong Xie
- Department of Cardiovascular surgery, Second Xiangya Hospital of Central South University, Changsha, China
- Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, China
| | - Yalin Liu
- Department of Cardiovascular surgery, Second Xiangya Hospital of Central South University, Changsha, China
- Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, China
| | - Can Huang
- Department of Cardiovascular surgery, Second Xiangya Hospital of Central South University, Changsha, China
- Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, China
| | - Yuhong Liu
- Department of Cardiovascular surgery, Second Xiangya Hospital of Central South University, Changsha, China
- Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, China
| |
Collapse
|
25
|
Zhang X, Ren Z, Xu W, Jiang Z. Necroptosis in atherosclerosis. Clin Chim Acta 2022; 534:22-28. [PMID: 35809652 DOI: 10.1016/j.cca.2022.07.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 11/26/2022]
Abstract
Atherosclerosis, a chronic inflammatory disease, is a leading cause of death worldwide. Vascular endothelial cells (VECs), vascular smooth muscle cells (VSMCs) and macrophages play extremely vital roles in the formation of atherosclerotic plaques and subsequent atherosclerosis. Necroptosis, a caspase-independent programmed cell necrosis, occurs in advanced atherosclerotic plaques and has been implicated in VEC, VSMC and macrophage function. Although necroptosis may have considered as a defensive line against intracellular infection, it can induce a pro-inflammatory state, which will accelerate the disease process. Accordingly, necroptosis plays an important pathophysiologic role. In this review, we explore the role of necroptosis in VECs, VSMCs and macrophages in atherosclerotic plaques and their connection to atherosclerosis.
Collapse
Affiliation(s)
- Xiaofan Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Hengyang Medical School, University of South China, Hengyang 421001, China.
| | - Zhong Ren
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Hengyang Medical School, University of South China, Hengyang 421001, China.
| | - Wenxin Xu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Hengyang Medical School, University of South China, Hengyang 421001, China.
| | - Zhisheng Jiang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, Hengyang Medical School, University of South China, Hengyang 421001, China.
| |
Collapse
|
26
|
Lee HW, Gu MJ, Yoo G, Choi IW, Lee SH, Kim Y, Ha SK. Glycolaldehyde induces synergistic effects on vascular inflammation in TNF-α-stimulated vascular smooth muscle cells. PLoS One 2022; 17:e0270249. [PMID: 35788200 PMCID: PMC9255721 DOI: 10.1371/journal.pone.0270249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/07/2022] [Indexed: 11/18/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease that contributes to disease progression is associated with the expression of adhesion molecules in vascular smooth muscle cells (VSMCs). Glycolaldehyde (GA) has been shown to impair cellular function in various disorders, including diabetes, and renal diseases. This study investigated the effect of GA on the expression of adhesion molecules in the mouse VSMC line, MOVAS-1. Co-incubation of VSMCs with GA (25–50 μM) dose-dependently increased the protein and mRNA level of Vcam-1 and ICAM-1. Additionally, GA upregulated intracellular ROS production and phosphorylation of MAPK and NK-κB. GA also elevated TNF-α-induced PI3K-AKT activation. Furthermore, GA enhanced TNF-α-activated IκBα kinase activation, subsequent IκBα degradation, and nuclear translocation of NF-κB. These findings suggest that GA stumulated VSMC adhesive capacity and the induction of VCAM-1 and ICAM-1 in VSMCs through inhibition of MAPK and NF-κB signaling pathways, providing insights into the effect of GA to induce inflammation within atherosclerotic lesions.
Collapse
Affiliation(s)
- Hee-Weon Lee
- Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Min Ji Gu
- Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Guijae Yoo
- Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - In-Wook Choi
- Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Sang-Hoon Lee
- Korea Food Research Institute, Wanju-gun, Republic of Korea
- Division of Food Biotechnology, University of Science and Technology, Daejeon, Korea
| | - Yoonsook Kim
- Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Sang Keun Ha
- Korea Food Research Institute, Wanju-gun, Republic of Korea
- Division of Food Biotechnology, University of Science and Technology, Daejeon, Korea
- * E-mail:
| |
Collapse
|
27
|
The Functional Interaction of EGFR with AT1R or TP in Primary Vascular Smooth Muscle Cells Triggers a Synergistic Regulation of Gene Expression. Cells 2022; 11:cells11121936. [PMID: 35741065 PMCID: PMC9222111 DOI: 10.3390/cells11121936] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/11/2022] [Accepted: 06/15/2022] [Indexed: 02/01/2023] Open
Abstract
In vivo, cells are simultaneously exposed to multiple stimuli whose effects are difficult to distinguish. Therefore, they are often investigated in experimental cell culture conditions where stimuli are applied separately. However, it cannot be presumed that their individual effects simply add up. As a proof-of-principle to address the relevance of transcriptional signaling synergy, we investigated the interplay of the Epidermal Growth Factor Receptor (EGFR) with the Angiotensin-II (AT1R) or the Thromboxane-A2 (TP) receptors in murine primary aortic vascular smooth muscle cells. Transcriptome analysis revealed that EGFR-AT1R or EGFR-TP simultaneous activations led to different patterns of regulated genes compared to individual receptor activations (qualitative synergy). Combined EGFR-TP activation also caused a variation of amplitude regulation for a defined set of genes (quantitative synergy), including vascular injury-relevant ones (Klf15 and Spp1). Moreover, Gene Ontology enrichment suggested that EGFR and TP-induced gene expression changes altered processes critical for vascular integrity, such as cell cycle and senescence. These bioinformatics predictions regarding the functional relevance of signaling synergy were experimentally confirmed. Therefore, by showing that the activation of more than one receptor can trigger a synergistic regulation of gene expression, our results epitomize the necessity to perform comprehensive network investigations, as the study of individual receptors may not be sufficient to understand their physiological or pathological impact.
Collapse
|
28
|
Green tea polyphenols in cardiometabolic health: A critical appraisal on phytogenomics towards personalized green tea. PHARMANUTRITION 2022. [DOI: 10.1016/j.phanu.2022.100296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
29
|
Weng Y, Peng Y, Xu Y, Wang L, Wu B, Xiang H, Ji K, Guan X. The Ratio of Red Blood Cell Distribution Width to Albumin Is Correlated With All-Cause Mortality of Patients After Percutaneous Coronary Intervention – A Retrospective Cohort Study. Front Cardiovasc Med 2022; 9:869816. [PMID: 35686040 PMCID: PMC9170887 DOI: 10.3389/fcvm.2022.869816] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 04/14/2022] [Indexed: 12/14/2022] Open
Abstract
Objectives The purpose of this study was to investigate the independent effect of the ratio of red blood cell distribution width (RDW) to albumin (RA) on all-cause mortality in patients after percutaneous coronary intervention (PCI). Methods Clinical data were obtained from the Multiparameter Intelligent Monitoring in Intensive Care-III (MIMIC-III) database version 1.4 and the database of Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University. We used the MIMIC-III database for model training, and data collected from the Second Affiliated Hospital of Wenzhou Medical University for validation. The primary outcome of our study was 90-day mortality. Cox proportional hazards regression model was used to estimate hazard ratio (HR) for the association between RA and all-cause mortality in patients after PCI. Pearson correlation analysis was conducted to assess the relationship between RA and Gensini score or cardiac troponin I (cTnI). Results A total of 707 patients were eligible in MIMIC-III database, including 432 males, with a mean age of 70.29 years. For 90-day all-cause mortality, in the adjusted multivariable model, the adjusted HRs [95% confidence intervals (CIs)] for the second (RA: 3.7–4.5 ml/g) and third (RA >4.5 ml/g) tertiles were 2.27 (1.11, 4.64) and 3.67 (1.82, 7.40), respectively, compared to the reference group (RA <3.7 ml/g) (p < 0.05). A similar relationship was also observed for 30-day all-cause mortality and 1-year all-cause mortality. No significant interaction was observed in subgroup analysis. Receiver operating characteristic (ROC) curve analysis proved that the ability of RA to predict the 90-day mortality was better than that of RDW or albumin alone. The correlation coefficient between Gensini score and RA was 0.254, and that between cTnI and RA was 0.323. Conclusion RA is an independent risk factor for all-cause mortality in patients after PCI. The higher the RA, the higher the mortality. RA has a good predictive ability for all-cause mortality in patients after PCI, which is better than RDW or albumin alone. RA may be positively correlated with the severity of coronary artery disease (CAD) in patients with CAD.
Collapse
|
30
|
Guo Y, Qin J, Zhao Q, Yang J, Wei X, Huang Y, Xie M, Zhang C, Li Y. Plaque-Targeted Rapamycin Spherical Nucleic Acids for Synergistic Atherosclerosis Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105875. [PMID: 35344289 PMCID: PMC9165522 DOI: 10.1002/advs.202105875] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/21/2022] [Indexed: 05/04/2023]
Abstract
Atherosclerosis with unstable plaques is the dominant pathological basis of lethal cardio-cerebrovascular diseases, which can cause acute death due to the rupture of plaques. Plaque-targeted drug delivery to achieve promoted treatment remains the main challenge because of the systemic occurrence of atheroma. Herein, a rapamycin (RAP) spherical nucleic acid (SNA) structure, capable of specifically accumulating in plaques for synergistic atherosclerosis treatment is constructed. By designing consecutive phosphorothioate (PS) at 3' terminus of the deoxyribonucleic acid (DNA) strand, multiple hydrophobic RAPs are covalently grafted onto the PS segment to form an amphiphilic drug-grafted DNA (RAP-DNA), which successively self-assembles into micellar SNA (RAP-SNA). Moreover, the phosphodiester-DNA segment constitutes the outer shell of RAP-SNA, enabling further hybridization with functional siRNA (targeting lectin-like oxidized low-density lipoprotein receptor-1, LOX-1) to obtain the drug codelivered SNA (LOX-1/RAP-SNA). With two active ingredients inside, LOX-1/RAP-SNA can not only induce robust autophagy and decrease the evil apoptosis of the pathological macrophages, but also simultaneously prohibit the LOX-1-mediated formation of damageable foam cells, realizing the effect of synergistic therapy. As a result, the LOX-1/RAP-SNA significantly reduces the progression of atheroma and stabilizes the plaques, providing a new strategy for synergistically targeted atherosclerosis treatment.
Collapse
Affiliation(s)
- Yuanyuan Guo
- Department of RadiologyShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai Jiao Tong University School of Medicine600 Yi Shan RoadShanghai200233China
| | - Jingcan Qin
- Department of RadiologyShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai Jiao Tong University School of Medicine600 Yi Shan RoadShanghai200233China
| | - Qianqian Zhao
- Department of RadiologyShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai Jiao Tong University School of Medicine600 Yi Shan RoadShanghai200233China
| | - Jiapei Yang
- School of Chemistry and Chemical EngineeringFrontiers Science Center for Transformative MoleculesShanghai Jiao Tong University800 Dongchuan RoadShanghai200240China
| | - Xiaoer Wei
- Department of RadiologyShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai Jiao Tong University School of Medicine600 Yi Shan RoadShanghai200233China
| | - Yu Huang
- Department of RadiologyShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai Jiao Tong University School of Medicine600 Yi Shan RoadShanghai200233China
| | - Miao Xie
- School of Chemistry and Chemical EngineeringFrontiers Science Center for Transformative MoleculesShanghai Jiao Tong University800 Dongchuan RoadShanghai200240China
| | - Chuan Zhang
- School of Chemistry and Chemical EngineeringFrontiers Science Center for Transformative MoleculesShanghai Jiao Tong University800 Dongchuan RoadShanghai200240China
| | - Yuehua Li
- Department of RadiologyShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai Jiao Tong University School of Medicine600 Yi Shan RoadShanghai200233China
| |
Collapse
|
31
|
Hou X, Dai H, Zheng Y. Circular RNA hsa_circ_0008896 accelerates atherosclerosis by promoting the proliferation, migration and invasion of vascular smooth muscle cells via hsa-miR-633/CDC20B (cell division cycle 20B) axis. Bioengineered 2022; 13:5987-5998. [PMID: 35212610 PMCID: PMC8973975 DOI: 10.1080/21655979.2022.2039467] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Circular RNAs, a class of circularly closed non-coding RNAs, play essential roles in the formation of atherosclerosis, which is a frequent cause of cardiovascular and cerebrovascular diseases. Although many circular RNAs are found to be involved in the progression of atherosclerosis, more circular RNA regulators still need to be identified, to improve the understanding of the regulatory networks of atherosclerosis. Here, we found that hsa_circ_0008896 was significantly up-regulated in both in vitro and in vivo atherosclerosis models, indicating hsa_circ_0008896 was involved in the progression of atherosclerosis. Further functional analyses confirmed that knockdown of hsa_circ_0008896 decreased proliferation, migration, and invasion of VSMCs. In addition, we conducted bioinformatics analysis and found that hsa-miR-633 could directly bind to hsa_circ_0008896, which was confirmed by RNA immune-precipitation (RIP) assays. Results of proliferation, migration, and invasion assays showed that hsa-miR-633 inhibitor reversed the si-circ_0008896 phenotypes, indicating that hsa_circ_0008896 functionally bound to hsa-miR-633. At last, combining bioinformatics and experimental analyses, we found the protein target of hsa_circ_0008896/hsa-miR-633, CDC20B (cell division cycle 20B). The expression level of CDC20B was regulated by hsa-miR-633, and knockdown of CDC20B decreased proliferation, migration, and invasion of VSMCs. Taken together, hsa_circ_0008896 regulated the expression of CDC20B by sponging hsa-miR-633, and then enhanced proliferation, migration, and invasion of VSMCs to promote the progression of atherosclerosis.
Collapse
Affiliation(s)
- Xumin Hou
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Huangdong Dai
- Department of Cardiovascular Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yue Zheng
- Department of Cardiovascular Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
32
|
Mammoto A, Matus K, Mammoto T. Extracellular Matrix in Aging Aorta. Front Cell Dev Biol 2022; 10:822561. [PMID: 35265616 PMCID: PMC8898904 DOI: 10.3389/fcell.2022.822561] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/07/2022] [Indexed: 12/11/2022] Open
Abstract
The aging population is booming all over the world and arterial aging causes various age-associated pathologies such as cardiovascular diseases (CVDs). The aorta is the largest elastic artery, and transforms pulsatile flow generated by the left ventricle into steady flow to maintain circulation in distal tissues and organs. Age-associated structural and functional changes in the aortic wall such as dilation, tortuousness, stiffening and losing elasticity hamper stable peripheral circulation, lead to tissue and organ dysfunctions in aged people. The extracellular matrix (ECM) is a three-dimensional network of macromolecules produced by resident cells. The composition and organization of key ECM components determine the structure-function relationships of the aorta and therefore maintaining their homeostasis is critical for a healthy performance. Age-associated remodeling of the ECM structural components, including fragmentation of elastic fibers and excessive deposition and crosslinking of collagens, is a hallmark of aging and leads to functional stiffening of the aorta. In this mini review, we discuss age-associated alterations of the ECM in the aortic wall and shed light on how understanding the mechanisms of aortic aging can lead to the development of efficient strategy for aortic pathologies and CVDs.
Collapse
Affiliation(s)
- Akiko Mammoto
- Department of Pediatrics, Milwaukee, WI, United States
- Department of Cell Biology, Neurobiology and Anatomy, Milwaukee, WI, United States
- *Correspondence: Akiko Mammoto, ; Tadanori Mammoto,
| | - Kienna Matus
- Department of Pediatrics, Milwaukee, WI, United States
| | - Tadanori Mammoto
- Department of Pediatrics, Milwaukee, WI, United States
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, United States
- *Correspondence: Akiko Mammoto, ; Tadanori Mammoto,
| |
Collapse
|
33
|
Tao J, Cao X, Yu B, Qu A. Vascular Stem/Progenitor Cells in Vessel Injury and Repair. Front Cardiovasc Med 2022; 9:845070. [PMID: 35224067 PMCID: PMC8866648 DOI: 10.3389/fcvm.2022.845070] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Vascular repair upon vessel injury is essential for the maintenance of arterial homeostasis and function. Stem/progenitor cells were demonstrated to play a crucial role in regeneration and replenishment of damaged vascular cells during vascular repair. Previous studies revealed that myeloid stem/progenitor cells were the main sources of tissue regeneration after vascular injury. However, accumulating evidences from developing lineage tracing studies indicate that various populations of vessel-resident stem/progenitor cells play specific roles in different process of vessel injury and repair. In response to shear stress, inflammation, or other risk factors-induced vascular injury, these vascular stem/progenitor cells can be activated and consequently differentiate into different types of vascular wall cells to participate in vascular repair. In this review, mechanisms that contribute to stem/progenitor cell differentiation and vascular repair are described. Targeting these mechanisms has potential to improve outcome of diseases that are characterized by vascular injury, such as atherosclerosis, hypertension, restenosis, and aortic aneurysm/dissection. Future studies on potential stem cell-based therapy are also highlighted.
Collapse
Affiliation(s)
- Jiaping Tao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- The Key Laboratory of Cardiovascular Remodeling-Related Diseases, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
| | - Xuejie Cao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- The Key Laboratory of Cardiovascular Remodeling-Related Diseases, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
| | - Baoqi Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- The Key Laboratory of Cardiovascular Remodeling-Related Diseases, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
- *Correspondence: Baoqi Yu
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- The Key Laboratory of Cardiovascular Remodeling-Related Diseases, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Metabolic Disorder-Related Cardiovascular Diseases, Beijing, China
- Aijuan Qu
| |
Collapse
|
34
|
An W, Luong LA, Bowden NP, Yang M, Wu W, Zhou X, Liu C, Niu K, Luo J, Zhang C, Sun X, Poston R, Zhang L, Evans PC, Xiao Q. Cezanne is a critical regulator of pathological arterial remodelling by targeting β-catenin signalling. Cardiovasc Res 2022; 118:638-653. [PMID: 33599243 PMCID: PMC8803089 DOI: 10.1093/cvr/cvab056] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 12/16/2020] [Accepted: 02/15/2021] [Indexed: 12/12/2022] Open
Abstract
AIMS Pathological arterial remodelling including neointimal hyperplasia and atherosclerosis is the main underlying cause for occluding arterial diseases. Cezanne is a novel deubiquitinating enzyme, functioning as a NF-кB negative regulator, and plays a key role in renal inflammatory response and kidney injury induced by ischaemia. Here we attempted to examine its pathological role in vascular smooth muscle cell (VSMC) pathology and arterial remodelling. METHODS AND RESULTS Cezanne expression levels were consistently induced by various atherogenic stimuli in VSMCs, and in remodelled arteries upon injury. Functionally, VSMCs over-expressing wild-type Cezanne, but not the mutated catalytically-inactive Cezanne (C209S), had an increased proliferative ability and mobility, while the opposite was observed in VSMCs with Cezanne knockdown. Surprisingly, we observed no significant effects of Cezanne on VSMC apoptosis, NF-κB signalling, or inflammation. RNA-sequencing and biochemical studies showed that Cezanne drives VSMC proliferation by regulating CCN family member 1 (CCN1) by targeting β-catenin for deubiquitination. Importantly, local correction of Cezanne expression in the injured arteries greatly decreased VSMC proliferation, and prevented arterial inward remodelling. Interestingly, global Cezanne gene deletion in mice led to smaller atherosclerotic plaques, but with a lower level of plaque stability. Translating, we observed a similar role for Cezanne in human VSMCs, and higher expression levels of Cezanne in human atherosclerotic lesions. CONCLUSION Cezanne is a key regulator of VSMC proliferation and migration in pathological arterial remodelling. Our findings have important implications for therapeutic targeting Cezanne signalling and VSMC pathology in vascular diseases.
Collapse
MESH Headings
- Animals
- Aorta/metabolism
- Aorta/pathology
- Apoptosis
- Atherosclerosis/enzymology
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Cell Movement
- Cell Proliferation
- Cells, Cultured
- Cysteine-Rich Protein 61/genetics
- Cysteine-Rich Protein 61/metabolism
- Disease Models, Animal
- Endopeptidases/genetics
- Endopeptidases/metabolism
- Humans
- Inflammation Mediators/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- NF-kappa B/metabolism
- Neointima
- Ubiquitination
- Vascular Remodeling
- Wnt Signaling Pathway
- beta Catenin/genetics
- beta Catenin/metabolism
- Mice
Collapse
Affiliation(s)
- Weiwei An
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Le A Luong
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Neil P Bowden
- Department of Infection, Immunity and Cardiovascular Disease, Bateson Centre, and Insigneo Institute for In Silico Medicine, University of Sheffield, Beech Hill Rd, Sheffield S10 2RX, UK
| | - Mei Yang
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
- Department of Cardiology, and Institute for Cardiovascular Development and Regenerative Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei Wu
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Xinmiao Zhou
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Chenxin Liu
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Kaiyuan Niu
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Jun Luo
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Cheng Zhang
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Xiaolei Sun
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Robin Poston
- Centre for Microvascular Research, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Li Zhang
- Department of Cardiology, and Institute for Cardiovascular Development and Regenerative Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Paul C Evans
- Department of Infection, Immunity and Cardiovascular Disease, Bateson Centre, and Insigneo Institute for In Silico Medicine, University of Sheffield, Beech Hill Rd, Sheffield S10 2RX, UK
| | - Qingzhong Xiao
- Centre for Clinical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
- Key Laboratory of Cardiovascular Diseases at The Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
35
|
Vaidyanathan K, Wang C, Krajnik A, Yu Y, Choi M, Lin B, Jang J, Heo SJ, Kolega J, Lee K, Bae Y. A machine learning pipeline revealing heterogeneous responses to drug perturbations on vascular smooth muscle cell spheroid morphology and formation. Sci Rep 2021; 11:23285. [PMID: 34857846 PMCID: PMC8640073 DOI: 10.1038/s41598-021-02683-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 11/22/2021] [Indexed: 02/06/2023] Open
Abstract
Machine learning approaches have shown great promise in biology and medicine discovering hidden information to further understand complex biological and pathological processes. In this study, we developed a deep learning-based machine learning algorithm to meaningfully process image data and facilitate studies in vascular biology and pathology. Vascular injury and atherosclerosis are characterized by neointima formation caused by the aberrant accumulation and proliferation of vascular smooth muscle cells (VSMCs) within the vessel wall. Understanding how to control VSMC behaviors would promote the development of therapeutic targets to treat vascular diseases. However, the response to drug treatments among VSMCs with the same diseased vascular condition is often heterogeneous. Here, to identify the heterogeneous responses of drug treatments, we created an in vitro experimental model system using VSMC spheroids and developed a machine learning-based computational method called HETEROID (heterogeneous spheroid). First, we established a VSMC spheroid model that mimics neointima-like formation and the structure of arteries. Then, to identify the morphological subpopulations of drug-treated VSMC spheroids, we used a machine learning framework that combines deep learning-based spheroid segmentation and morphological clustering analysis. Our machine learning approach successfully showed that FAK, Rac, Rho, and Cdc42 inhibitors differentially affect spheroid morphology, suggesting that multiple drug responses of VSMC spheroid formation exist. Overall, our HETEROID pipeline enables detailed quantitative drug characterization of morphological changes in neointima formation, that occurs in vivo, by single-spheroid analysis.
Collapse
Affiliation(s)
- Kalyanaraman Vaidyanathan
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14203, USA
| | - Chuangqi Wang
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01609, USA
| | - Amanda Krajnik
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14203, USA
| | - Yudong Yu
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01609, USA
| | - Moses Choi
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01609, USA
| | - Bolun Lin
- Department of Computer Science, Worcester Polytechnic Institute, Worcester, MA, 01609, USA
| | - Junbong Jang
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Su-Jin Heo
- Department of Orthopedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - John Kolega
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14203, USA
| | - Kwonmoo Lee
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, 01609, USA.
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, 02115, USA.
- Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA.
| | - Yongho Bae
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, 14203, USA.
| |
Collapse
|
36
|
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: 130] [Impact Index Per Article: 32.5] [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
|
37
|
Kjeldsen EW, Thomassen JQ, Frikke-Schmidt R. HDL cholesterol concentrations and risk of atherosclerotic cardiovascular disease - Insights from randomized clinical trials and human genetics. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1867:159063. [PMID: 34637926 DOI: 10.1016/j.bbalip.2021.159063] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/12/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022]
Abstract
Through seven decades the inverse association between HDL cholesterol concentrations and risk of atherosclerotic cardiovascular disease (ASCVD) has been observed in case-control and prospective cohort studies. This robust inverse association fuelled the enthusiasm towards development of HDL cholesterol increasing drugs, exemplified by the cholesteryl ester transfer protein (CETP) inhibitor trials and the extended-release niacin HPS2-THRIVE trial. These HDL cholesterol increasing trials were launched without conclusive evidence from human genetics, and despite discrepant species dependent evidence from animal studies. Evidence from human genetics and from randomized clinical trials over the last 13 years now point in the direction that concentrations of HDL cholesterol, do not appear to be a viable future path to target therapeutically for prevention of ASCVD. A likely explanation for the strong observational association between low HDL cholesterol and high ASCVD risk is the concomitant inverse association between HDL cholesterol and atherogenic triglyceride-rich lipoproteins. The purpose of the present review is to bring HDL cholesterol increasing trials into a human genetics context exemplified by candidate gene studies of key players in HDL biogenesis as well as by HDL cholesterol related genome-wide association studies.
Collapse
Affiliation(s)
- Emilie Westerlin Kjeldsen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Qvist Thomassen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Ruth Frikke-Schmidt
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
38
|
Dynamic Crosstalk between Vascular Smooth Muscle Cells and the Aged Extracellular Matrix. Int J Mol Sci 2021; 22:ijms221810175. [PMID: 34576337 PMCID: PMC8468233 DOI: 10.3390/ijms221810175] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/31/2021] [Accepted: 09/03/2021] [Indexed: 01/15/2023] Open
Abstract
Vascular aging is accompanied by the fragmentation of elastic fibers and collagen deposition, leading to reduced distensibility and increased vascular stiffness. A rigid artery facilitates elastin to degradation by MMPs, exposing vascular cells to greater mechanical stress and triggering signaling mechanisms that only exacerbate aging, creating a self-sustaining inflammatory environment that also promotes vascular calcification. In this review, we highlight the role of crosstalk between smooth muscle cells and the vascular extracellular matrix (ECM) and how aging promotes smooth muscle cell phenotypes that ultimately lead to mechanical impairment of aging arteries. Understanding the underlying mechanisms and the role of associated changes in ECM during aging may contribute to new approaches to prevent or delay arterial aging and the onset of cardiovascular diseases.
Collapse
|
39
|
Guo J, Yang Z, Wang X, Xu Y, Lu Y, Qin Z, Zhang L, Xu J, Wang W, Zhang J, Tang J. Advances in Nanomaterials for Injured Heart Repair. Front Bioeng Biotechnol 2021; 9:686684. [PMID: 34513807 PMCID: PMC8424111 DOI: 10.3389/fbioe.2021.686684] [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: 03/27/2021] [Accepted: 08/09/2021] [Indexed: 11/30/2022] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is one of the leading causes of mortality worldwide. Because of the limited regenerative capacity of adult myocardium to compensate for the loss of heart tissue after ischemic infarction, scientists have been exploring the possible mechanisms involved in the pathological process of ASCVD and searching for alternative means to regenerate infarcted cardiac tissue. Although numerous studies have pursued innovative solutions for reversing the pathological process of ASCVD and improving the effectiveness of delivering therapeutics, the translation of those advances into downstream clinical applications remains unsatisfactory because of poor safety and low efficacy. Recently, nanomaterials (NMs) have emerged as a promising new strategy to strengthen both the efficacy and safety of ASCVD therapy. Thus, a comprehensive review of NMs used in ASCVD treatment will be useful. This paper presents an overview of the pathophysiological mechanisms of ASCVD and the multifunctional mechanisms of NM-based therapy, including antioxidative, anti-inflammation and antiapoptosis mechanisms. The technological improvements of NM delivery are summarized and the clinical transformations concerning the use of NMs to treat ASCVD are examined. Finally, this paper discusses the challenges and future perspectives of NMs in cardiac regeneration to provide insightful information for health professionals on the latest advancements in nanotechnologies for ASCVD treatment.
Collapse
Affiliation(s)
- Jiacheng Guo
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Zhenzhen Yang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xu Wang
- Department of Medical Record Management, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanyan Xu
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Yongzheng Lu
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Zhen Qin
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Li Zhang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Jing Xu
- Department of Cardiac Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Wang
- Henan Medical Association, Zhengzhou, China
| | - Jinying Zhang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Junnan Tang
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| |
Collapse
|
40
|
Schereider IRG, Vassallo DV, Simões MR. Chronic mercury exposure induces oxidative stress in female rats by endothelial nitric oxide synthase uncoupling and cyclooxygenase-2 activation, without affecting oestrogen receptor function. Basic Clin Pharmacol Toxicol 2021; 129:470-485. [PMID: 34491608 DOI: 10.1111/bcpt.13655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 08/04/2021] [Accepted: 09/02/2021] [Indexed: 11/29/2022]
Abstract
Mercury has been shown to be a significant health risk factor and is positively associated with cardiovascular diseases. Evidence reveals that men are more likely to develop cardiovascular diseases than women during reproductive age. However, the effects of mercury in females remain poorly investigated, despite the finding that female hormones demonstrate a cardioprotective role. In the present study, we evaluated whether chronic mercury chloride exposure could alter blood pressure and vascular function of the female rat aorta. Ten-week-old female Wistar rats were divided into two groups: control (vehicle) and mercury treated (first dose of 4.6 μg/kg, subsequent daily doses of 0.07 μg/kg), im. Mercury treatment did not modify systolic blood pressure (SBP) but increased vascular reactivity due to the reduction of nitric oxide bioavailability associated with the increase in reactive oxygen species from endothelial nitric oxide synthase (eNOS) uncoupling. Furthermore, increased participation of the cyclooxygenase-2 pathway occurred through an imbalance in thromboxane 2 and prostacyclin 2. However, the oestrogen signalling pathway was not altered in either group. These results demonstrated that chronic exposure to mercury in females induced endothelial dysfunction and, consequently, increased aortic vascular reactivity, causing vascular damage to the female rat aorta and representing a risk of cardiovascular diseases.
Collapse
Affiliation(s)
| | - Dalton Valentim Vassallo
- Department of Physiological Sciences, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil.,Health Science Center of Vitória, School of Sciences of Santa Casa de Misericórdia de Vitória - EMESCAM, Vitória, Espírito Santo, Brazil
| | - Maylla Ronacher Simões
- Department of Physiological Sciences, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| |
Collapse
|
41
|
He S, Jiang H, Zhuo C, Jiang W. Trimethylamine/Trimethylamine-N-Oxide as a Key Between Diet and Cardiovascular Diseases. Cardiovasc Toxicol 2021; 21:593-604. [PMID: 34003426 DOI: 10.1007/s12012-021-09656-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 04/27/2021] [Indexed: 02/08/2023]
Abstract
Trimethylamine (TMA) is a gut microbiota-derived metabolite which comes from diets rich of choline, betaine or L-carnitine and could be further converted to Trimethylamine-N-oxide (TMAO) in the liver. As the function of gut microbiota and its metabolites being explored so far, studies suggest that TMAO may be a potential risk factor of cardiovascular diseases independent of other traditional risk factors. However, the precise role of TMAO is controversial as some converse results were discovered. In recent studies, it is hypothesized that TMA may also participate in the progression of cardiovascular diseases and some cytotoxic effect of TMA has been discovered. Thus, exploring the relationship between TMA, TMAO and CVD may bring a novel insight into the diagnosis and therapy of cardiovascular diseases. In this review, we discussed the factors which influence the TMA/TMAO's process of metabolism in the human body. We have also summarized the pathogenic effect of TMA/TMAO in cardiovascular diseases, as well as the limitation of some controversial discoveries.
Collapse
Affiliation(s)
- Siyu He
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Hong Jiang
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Caili Zhuo
- The Laboratory of Cardiovascular Diseases, Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Wei Jiang
- The Laboratory of Cardiovascular Diseases, Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
42
|
Kraler S, Libby P, Evans PC, Akhmedov A, Schmiady MO, Reinehr M, Camici GG, Lüscher TF. Resilience of the Internal Mammary Artery to Atherogenesis: Shifting From Risk to Resistance to Address Unmet Needs. Arterioscler Thromb Vasc Biol 2021; 41:2237-2251. [PMID: 34107731 PMCID: PMC8299999 DOI: 10.1161/atvbaha.121.316256] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Fueled by the global surge in aging, atherosclerotic cardiovascular disease reached pandemic dimensions putting affected individuals at enhanced risk of myocardial infarction, stroke, and premature death. Atherosclerosis is a systemic disease driven by a wide spectrum of factors, including cholesterol, pressure, and disturbed flow. Although all arterial beds encounter a similar atherogenic milieu, the development of atheromatous lesions occurs discontinuously across the vascular system. Indeed, the internal mammary artery possesses unique biological properties that confer protection to intimal growth and atherosclerotic plaque formation, thus making it a conduit of choice for coronary artery bypass grafting. Its endothelium abundantly expresses nitric oxide synthase and shows accentuated nitric oxide release, while its vascular smooth muscle cells exhibit reduced tissue factor expression, high tPA (tissue-type plasminogen activator) production and blunted migration and proliferation, which may collectively mitigate intimal thickening and ultimately the evolution of atheromatous plaques. We aim here to provide insights into the anatomy, physiology, cellular, and molecular aspects of the internal mammary artery thereby elucidating its remarkable resistance to atherogenesis. We propose a change in perspective from risk to resilience to decipher mechanisms of atheroresistance and eventually identification of novel therapeutic targets presently not addressed by currently available remedies.
Collapse
Affiliation(s)
- Simon Kraler
- Center for Molecular Cardiology, University of Zürich, 8952 Schlieren, Switzerland
| | - Peter Libby
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, USA
| | - Paul C. Evans
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zürich, 8952 Schlieren, Switzerland
| | - Martin O. Schmiady
- Clinic for Cardiac Surgery, University Heart Centre, University Hospital Zurich, Zurich, Switzerland
| | - Michael Reinehr
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Giovanni G. Camici
- Center for Molecular Cardiology, University of Zürich, 8952 Schlieren, Switzerland
- University Heart Center, Department of Cardiology, University Hospital, Zurich, Switzerland
- Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Thomas F. Lüscher
- Center for Molecular Cardiology, University of Zürich, 8952 Schlieren, Switzerland
- Royal Brompton and Harefield Hospitals and Imperial College, London, United Kingdom
| |
Collapse
|
43
|
Libby P. Inflammation in Atherosclerosis-No Longer a Theory. Clin Chem 2021; 67:131-142. [PMID: 33393629 DOI: 10.1093/clinchem/hvaa275] [Citation(s) in RCA: 182] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 10/23/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND Inflammation links to atherosclerosis and its complications in various experimental investigations. Animal studies have implicated numerous inflammatory mediators in the initiation and complication of atherosclerosis. Numerous studies in humans have shown associations of biomarkers of inflammation with cardiovascular events provoked by atheromata. Inflammatory status, determined by the biomarker C-reactive protein, can guide the allocation of statin therapy to individuals without elevated low-density lipoprotein (LDL) concentrations to prevent first ever adverse cardiovascular events. CONTENT Until recently, no direct evidence has shown that an intervention that selectively limits inflammation can improve outcomes in patients with atherosclerosis. A recent study, based on decades of preclinical investigation, treated patients who had sustained a myocardial infarction and whose LDL was well-controlled on statin treatment with an antibody that neutralizes interleukin-1 beta. This trial, conducted in over 10 000 individuals, showed a reduction in major adverse cardiac events, establishing for the first time the clinical efficacy of an anti-inflammatory intervention in atherosclerosis. Two large subsequent studies have shown that colchicine treatment can also prevent recurrent events in patients recovering from an acute coronary syndrome or in the stable phase of coronary artery disease. These clinical trials have transformed inflammation in atherosclerosis from theory to practice. SUMMARY Much work remains to optimize further anti-inflammatory interventions, minimize unwanted actions, and refine patient selection. This long road from discovery in the laboratory to successful clinical trials represents a victory for medical science, and opens a new avenue to reducing the risk that remains despite current treatments for atherosclerosis.
Collapse
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
44
|
Liu QK. Triglyceride-lowering and anti-inflammatory mechanisms of omega-3 polyunsaturated fatty acids for atherosclerotic cardiovascular risk reduction. J Clin Lipidol 2021; 15:556-568. [PMID: 34172393 DOI: 10.1016/j.jacl.2021.05.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 05/28/2021] [Accepted: 05/30/2021] [Indexed: 01/08/2023]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is the leading cause of death globally. Omega-3 polyunsaturated fatty acids (PUFAs) including eicosapentaenoic acid and docosahexaenoic acid have been extensively studied as both dietary supplement and pharmaceutical agent for the prevention of ASCVD. Epidemiological and retrospective studies have long shown the inverse relationship of omega-3 PUFA consumption and ASCVD event but results of previous large randomized controlled trials have not consistently shown the same effect. Meta-analysis and a recent clinical trial using a high dose of eicosapentaenoic acid showed convincing protective effects of omega-3 PUFAs on ASCVD. Emerging evidence shows that both chronic inflammation and hypertriglyceridemia increase the risk of atherosclerosis. Amelioration of the inflammatory process and reduction of hypertriglyceridemia provide two mechanisms on the prevention and management of ASCVD, and agents with both of these effects are more potent and desirable. Omega-3 PUFAs exert anti-hypertriglyceridemia effect, ameliorate inflammation, and maintain the resolution of inflammation homeostasis pleiotropically through multiple molecular and cellular mechanisms. This review presents the pathophysiology of atherosclerosis, the mechanisms of omega-3 PUFAs on the reduction of the atherosclerotic risk, and the current clinical utilities of omega-3 PUFAs on the prevention of ASCVD.
Collapse
Affiliation(s)
- Qiyuan Keith Liu
- MedStar Medical Group, MedStar Montgomery Medical Center, Olney, MD 20832, USA.
| |
Collapse
|
45
|
Guo Z, Yu B, Li X, Yang X, Wang C, Fan L. Circadian misalignment promotes vascular smooth muscle cell apoptosis via defective autophagy. J Mol Histol 2021; 52:799-808. [PMID: 34131827 DOI: 10.1007/s10735-021-10000-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 06/12/2021] [Indexed: 01/28/2023]
Abstract
Defective autophagy in vascular smooth muscle cells (VSMCs) in response to oxidative stress can lead to cellular apoptosis and plaque instability. Previous studies have revealed that the circadian clock system is involved in autophagic regulation and plaque progression. However, the mechanism by which circadian rhythmicity influences VSMC autophagy and plaque stability remains unclear. Our study described the circadian profiles in atheromatous plaques and verified the role of circadian misalignment in VSMC autophagy and apoptosis. We found that the mRNA expression levels of circadian locomotor output cycles protein kaput (CLOCK) and Beclin 1 were significantly decreased in unstable plaques compared with stable plaques. No significant differences were observed in other circadian rhythm genes. VSMCs treated with oxidized low-density lipoprotein (ox-LDL, 80 μg/ml) exhibited abnormal circadian rhythmicity and impaired autophagy, as evidenced by consistent decreases in CLOCK and Beclin 1 expression, suggesting a correlation between CLOCK and autophagy. CLOCK protein expression was inhibited by ox-LDL, accompanied by defective autophagy and an increased apoptosis rates (P < 0.05). Administration of rapamycin (10 nM) reversed the effect of ox-LDL on VSMC autophagy and apoptosis. Finally, CLOCK silencing led to a considerable decrease in autophagy. VSMCs with stable CLOCK silencing also showed an increased apoptosis rate. In addition, gene silencing of CLOCK in VSMCs counteracted the effects of moderate rapamycin concentrations on autophagy and apoptosis. In conclusion, these findings suggested that the CLOCK-dependent rapamycin signaling pathway is a critical mediator in ox-LDL-induced VSMCs with defective autophagy that exacerbates plaque destabilization.
Collapse
Affiliation(s)
- Zhenyu Guo
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, 1158 Park Road, Qingpu, Shanghai, 201700, China
| | - Baixue Yu
- Biomedical Research Centre, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xu Li
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, 1158 Park Road, Qingpu, Shanghai, 201700, China
| | - Xiaohu Yang
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, 1158 Park Road, Qingpu, Shanghai, 201700, China
| | - Chen Wang
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Longhua Fan
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, 1158 Park Road, Qingpu, Shanghai, 201700, China. .,Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
46
|
Rodrigues CDSA, Bazan R, Reis FM, Silveira CFSMPD, Hueb LMS, Carvalho FCD, Nunes HRDC, Okoshi K, Hueb JC, Bazan SGZ. Carotid Artery Atherosclerotic Profile as Risk Predictor for Restenosis After Coronary Stenting. Arq Bras Cardiol 2021; 116:727-733. [PMID: 33886718 PMCID: PMC8121410 DOI: 10.36660/abc.20190650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 12/27/2019] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The incidence of restenosis of the coronary artery after a bare-metal stent implant has been lower than in simple balloon angioplasty; however, it still shows relatively high rates. OBJECTIVE The aim of this study was to find new risk indicators for in-stent restenosis using carotid ultrasonography, that, in addition to the already existing indicators, would help in decision-making for stent selection. METHODS We carried out a cross-sectional prospective study including 121 consecutive patients with chronic coronary artery disease who had undergone percutaneous coronary intervention with repeat angiography in the previous 12 months. After all cases of in-stent restenosis were identified, patients underwent carotid ultrasonography to evaluate carotid intima-media thickness and atherosclerosis plaques. The data were analyzed by Cox multiple regression. The significance level was set a p<0.05. RESULTS Median age of patients was 60 years (1st quartile = 55, 3rd quartile = 68), and 64.5% of patients were male. Coronary angiography showed that 57 patients (47.1%) presented in-stent restenosis. Fifty-five patients (45.5%) had echolucent atherosclerotic plaques in carotid arteries and 54.5% had echogenic plaques or no plaques. Of patients with who had echolucent plaques, 90.9% presented coronary in-stent restenosis. Of those who had echogenic plaques or no plaques, 10.6% presented in-stent restenosis. The presence of echolucent plaques in carotid arteries increased the risk of coronary in-stent restenosis by 8.21 times (RR=8.21; 95%CI: 3.58-18.82; p<0.001). CONCLUSIONS The presence of echolucent atherosclerotic plaques in carotid artery constitutes a risk predictor of coronary instent restenosis and should be considered in the selection of the type of stent to be used in coronary angioplasty.
Collapse
Affiliation(s)
| | - Rodrigo Bazan
- Universidade Estadual Paulista Julio de Mesquita Filho - Faculdade de Medicina Campus de Botucatu, Botucatu, SP - Brasil
| | - Fabrício Moreira Reis
- Universidade Estadual Paulista Julio de Mesquita Filho - Faculdade de Medicina Campus de Botucatu, Botucatu, SP - Brasil
| | | | - Lívia Maria Severino Hueb
- Universidade Estadual Paulista Julio de Mesquita Filho - Faculdade de Medicina Campus de Botucatu, Botucatu, SP - Brasil
| | - Fábio Cardoso de Carvalho
- Universidade Estadual Paulista Julio de Mesquita Filho - Faculdade de Medicina Campus de Botucatu, Botucatu, SP - Brasil
| | - Hélio Rubens de Carvalho Nunes
- Universidade Estadual Paulista Julio de Mesquita Filho - Faculdade de Medicina Campus de Botucatu, Botucatu, SP - Brasil
| | - Katashi Okoshi
- Universidade Estadual Paulista Julio de Mesquita Filho - Faculdade de Medicina Campus de Botucatu, Botucatu, SP - Brasil
| | - João Carlos Hueb
- Universidade Estadual Paulista Julio de Mesquita Filho - Faculdade de Medicina Campus de Botucatu, Botucatu, SP - Brasil
| | - Silméia Garcia Zanati Bazan
- Universidade Estadual Paulista Julio de Mesquita Filho - Faculdade de Medicina Campus de Botucatu, Botucatu, SP - Brasil
| |
Collapse
|
47
|
Herrmann J, Gummi MR, Xia M, van der Giet M, Tölle M, Schuchardt M. Vascular Calcification in Rodent Models-Keeping Track with an Extented Method Assortment. BIOLOGY 2021; 10:biology10060459. [PMID: 34067504 PMCID: PMC8224561 DOI: 10.3390/biology10060459] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/12/2021] [Accepted: 05/20/2021] [Indexed: 02/07/2023]
Abstract
Simple Summary Arterial vessel diseases are the leading cause of death in the elderly and their accelerated pathogenesis is responsible for premature death in patients with chronic renal failure. Since no functioning therapy concepts exist so far, the identification of the main signaling pathways is of current research interest. To develop therapeutic concepts, different experimental rodent models are needed, which should be subject to the 3R principle of Russel and Burch: “Replace, Reduce and Refine”. This review aims to summarize the current available experimental rodent models for studying vascular calcification and their quantification methods. Abstract Vascular calcification is a multifaceted disease and a significant contributor to cardiovascular morbidity and mortality. The calcification deposits in the vessel wall can vary in size and localization. Various pathophysiological pathways may be involved in disease progression. With respect to the calcification diversity, a great number of research models and detection methods have been established in basic research, relying mostly on rodent models. The aim of this review is to provide an overview of the currently available rodent models and quantification methods for vascular calcification, emphasizing animal burden and assessing prospects to use available methods in a way to address the 3R principles of Russel and Burch: “Replace, Reduce and Refine”.
Collapse
Affiliation(s)
- Jaqueline Herrmann
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (J.H.); (M.R.G.); (M.X.); (M.v.d.G.); (M.T.)
- Department of Chemistry, Biochemistry and Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2+4, 14195 Berlin, Germany
| | - Manasa Reddy Gummi
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (J.H.); (M.R.G.); (M.X.); (M.v.d.G.); (M.T.)
| | - Mengdi Xia
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (J.H.); (M.R.G.); (M.X.); (M.v.d.G.); (M.T.)
| | - Markus van der Giet
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (J.H.); (M.R.G.); (M.X.); (M.v.d.G.); (M.T.)
| | - Markus Tölle
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (J.H.); (M.R.G.); (M.X.); (M.v.d.G.); (M.T.)
| | - Mirjam Schuchardt
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (J.H.); (M.R.G.); (M.X.); (M.v.d.G.); (M.T.)
- Correspondence: ; Tel.: +49-30-450-514-690
| |
Collapse
|
48
|
Sun JX, Zhang C, Cheng ZB, Tang MY, Liu YZ, Jiang JF, Xiao X, Huang L. Chemerin in atherosclerosis. Clin Chim Acta 2021; 520:8-15. [PMID: 34022243 DOI: 10.1016/j.cca.2021.05.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/15/2021] [Accepted: 05/18/2021] [Indexed: 02/06/2023]
Abstract
Atherosclerosis (AS), a chronic arterial disease, is characterized by endothelial dysfunction, inflammatory reactions and lipid accumulation in parallel with aberrant angiogenesis and vascular smooth muscle cell (VSMC) proliferation. Adipose tissue has been suggested to have an integral influence on metabolism and endocrine secretion, while there have been increasing concerns about the possible involvement of adipokines in cardiovascular diseases, including AS. Here, we focused on chemerin, an adipokine highly expressed in adipose tissue, with strong evidence of an association with inflammation, endothelial dysfunction, metabolic disorder, aberrant angiogenesis, VSMC proliferation and calcification. In this review, we discuss chemerin and its receptors in the pathogenesis of AS. However, the existing data assign various, even contradictory, roles to chemerin in atherosclerosis, such as inhibiting vascular calcification and impairing endothelial function. Current studies focusing on its anti- and pro-atherogenic effects have pinpointed its distinct role in specific cell types and contexts in the pathogenesis of atherosclerosis. Therefore, the gaps in current knowledge regarding the specific role played by chemerin in the etiology of AS require additional future studies. It seems reasonable to suggest that targeted chemerin therapy can be developed as an innovative approach for treating AS.
Collapse
Affiliation(s)
- Jia-Xiang Sun
- Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Chi Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Zhe-Bin Cheng
- Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Stomatology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Mu-Yao Tang
- Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Yi-Zhang Liu
- Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Jie-Feng Jiang
- Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Xuan Xiao
- Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China; Departments of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Liang Huang
- Research Laboratory of Translational Medicine, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, People's Republic of China.
| |
Collapse
|
49
|
Gielen S. The new EAPC core curriculum for preventive cardiology: the keystone of the EAPC transformation. Eur J Prev Cardiol 2021; 29:246-250. [PMID: 34003241 DOI: 10.1093/eurjpc/zwab079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Stephan Gielen
- Klinikum Lippe, Klinik für Innere Medizin/Kardiologie, Angiologie und Internistische Intensivmedizin, Röntgenstr. 18, 32756 Detmold, Germany.,Martin-Luther-Universität Halle-Wittenberg, Medizinische Fakultät, Magdeburger Str.8, 06108 Halle/Saale, Germany
| |
Collapse
|
50
|
Walther BK, Rajeeva Pandian NK, Gold KA, Kiliç ES, Sama V, Gu J, Gaharwar AK, Guiseppi-Elie A, Cooke JP, Jain A. Mechanotransduction-on-chip: vessel-chip model of endothelial YAP mechanobiology reveals matrix stiffness impedes shear response. LAB ON A CHIP 2021; 21:1738-1751. [PMID: 33949409 PMCID: PMC9761985 DOI: 10.1039/d0lc01283a] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Endothelial mechanobiology is a key consideration in the progression of vascular dysfunction, including atherosclerosis. However mechanistic connections between the clinically associated physical stimuli, vessel stiffness and shear stress, and how they interact to modulate plaque progression remain incompletely characterized. Vessel-chip systems are excellent candidates for modeling vascular mechanobiology as they may be engineered from the ground up, guided by the mechanical parameters present in human arteries and veins, to recapitulate key features of the vasculature. Here, we report extensive validation of a vessel-chip model of endothelial yes-associated protein (YAP) mechanobiology, a protein sensitive to both matrix stiffness and shearing forces and, importantly, implicated in atherosclerotic progression. Our model captures the established endothelial mechanoresponse, with endothelial alignment, elongation, reduction of adhesion molecules, and YAP cytoplasmic retention under high laminar shear. Conversely, we observed disturbed morphology, inflammation, and nuclear partitioning under low, high, and high oscillatory shear. Examining targets of YAP transcriptional co-activation, connective tissue growth factor (CTGF) is strongly downregulated by high laminar shear, whereas it is strongly upregulated by low shear or oscillatory flow. Ankyrin repeat domain 1 (ANKRD1) is only upregulated by high oscillatory shear. Verteporfin inhibition of YAP reduced the expression of CTGF but did not affect ANKRD1. Lastly, substrate stiffness modulated the endothelial shear mechanoresponse. Under high shear, softer substrates showed the lowest nuclear localization of YAP whereas stiffer substrates increased nuclear localization. Low shear strongly increased nuclear localization of YAP across stiffnesses. Together, we have validated a model of endothelial mechanobiology and describe a clinically relevant biological connection between matrix stiffness, shear stress, and endothelial activation via YAP mechanobiology.
Collapse
Affiliation(s)
- Brandon K Walther
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, USA. and Department of Cardiovascular Sciences, Houston Methodist Institute for Academic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, Texas 77030, USA.
| | | | - Karli A Gold
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, USA.
| | - Ecem S Kiliç
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, USA.
| | - Vineeth Sama
- Department of Biomedical Engineering, Clemson University, Clemson, South Carolina 29634, USA.
| | - Jianhua Gu
- Department of Cardiovascular Sciences, Houston Methodist Institute for Academic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, Texas 77030, USA.
| | - Akhilesh K Gaharwar
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, USA. and Department of Materials Science, Texas A&M University, College Station, Texas 77843, USA
| | - Anthony Guiseppi-Elie
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, USA. and Department of Cardiovascular Sciences, Houston Methodist Institute for Academic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, Texas 77030, USA. and ABTECH Scientific, Inc., Biotechnology Research Park, 800 East Leigh Street, Richmond, Virginia 23219, USA and Department of Biomedical Engineering, Anderson University, Anderson, South Carolina 29621, USA.
| | - John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Institute for Academic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, Texas 77030, USA.
| | - Abhishek Jain
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843, USA. and Department of Cardiovascular Sciences, Houston Methodist Institute for Academic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, Texas 77030, USA. and Department of Medical Physiology, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA
| |
Collapse
|