1
|
Wei SY, Fu WS, Liu CH, Wang WL, Shih YT, Chien S, Chiu JJ. Identification of KU-55933 as an anti-atherosclerosis compound by using a hemodynamic-based high-throughput drug screening platform. Proc Natl Acad Sci U S A 2024; 121:e2318718121. [PMID: 38252820 PMCID: PMC10835076 DOI: 10.1073/pnas.2318718121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 12/19/2023] [Indexed: 01/24/2024] Open
Abstract
Several compounds have been used for atherosclerosis treatment, including clinical trials; however, no anti-atherosclerotic drugs based on hemodynamic force-mediated atherogenesis have been discovered. Our previous studies demonstrated that "small mothers against decapentaplegic homolog 1/5" (Smad1/5) is a convergent signaling molecule for chemical [e.g., bone morphogenetic proteins (BMPs)] and mechanical (e.g., disturbed flow) stimulations and hence may serve as a promising hemodynamic-based target for anti-atherosclerosis drug development. The goal of this study was to develop a high-throughput screening (HTS) platform to identify potential compounds that can inhibit disturbed flow- and BMP-induced Smad1/5 activation and atherosclerosis. Through HTS using a Smad1/5 downstream target inhibitor of DNA binding 1 (Id-1) as a luciferase reporter, we demonstrated that KU-55933 and Apicidin suppressed Id-1 expression in AD-293 cells. KU-55933 (10 μM), Apicidin (10 μM), and the combination of half doses of each [1/2(K + A)] inhibited disturbed flow- and BMP4-induced Smad1/5 activation in human vascular endothelial cells (ECs). KU-55933, Apicidin, and 1/2(K + A) treatments caused 50.6%, 47.4%, and 73.3% inhibitions of EC proliferation induced by disturbed flow, respectively, whereas EC inflammation was only suppressed by KU-55933 and 1/2(K + A), but not Apicidin alone. Administrations of KU-55933 and 1/2(K + A) to apolipoprotein E-deficient mice inhibited Smad1/5 activation in ECs in athero-susceptible regions, thereby suppressing endothelial proliferation and inflammation, with the attenuation of atherosclerotic lesions in these mice. A unique drug screening platform has been developed to demonstrate that KU-55933 and its combination with Apicidin are promising therapeutic compounds for atherosclerosis based on hemodynamic considerations.
Collapse
Affiliation(s)
- Shu-Yi Wei
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli350, Taiwan
| | - Wei-Shan Fu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli350, Taiwan
| | - Chang-Hsuan Liu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli350, Taiwan
| | - Wei-Li Wang
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli350, Taiwan
| | - Yu-Tsung Shih
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli350, Taiwan
- Department of Anatomy and Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei110, Taiwan
| | - Shu Chien
- Department of Bioengineering and Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA93093
- Department of Medicine, University of California, San Diego, La Jolla, CA93093
| | - Jeng-Jiann Chiu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli350, Taiwan
- College of Medical Science and Technology, Taipei Medical University, Taipei110, Taiwan
- Taipei Heart Institute, Taipei Medical University, Taipei110, Taiwan
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu300, Taiwan
| |
Collapse
|
2
|
Belhoul-Fakir H, Brown ML, Thompson PL, Hamzah J, Jansen S. Connecting the Dots: How Injury in the Arterial Wall Contributes to Atherosclerotic Disease. Clin Ther 2023; 45:1092-1098. [PMID: 37891144 DOI: 10.1016/j.clinthera.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/22/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023]
Abstract
PURPOSE The occurrence and development of atherosclerotic cardiovascular disease, which can result in severe outcomes, such as myocardial infarction, stroke, loss of limb, renal failure, and infarction of the gut, are strongly associated with injury to the intimal component of the arterial wall whether via the inside-out or outside-in pathways. The role of injury to the tunica media as a pathway of atherosclerosis initiation is an underresearched area. This review focuses on potential pathways to vessel wall injury as well as current experimental and clinical research in the middle-aged and elderly populations, including the role of exercise, as it relates to injury to the tunica media. METHODS A database search using PubMed and Google Scholar was conducted for research articles published between 1909 and 2023 that focused on pathways of atherogenesis and the impact of mechanical forces on wall injury. The following key words were searched: wall injury, tunica media, atherogenesis, vascular aging, and wall strain. Studies were analyzed, and the relevant information was extracted from each study. FINDINGS A link between high mechanical stress in the arterial wall and reduced vascular compliance was found. The stiffening and calcification of the arterial wall with aging induce high blood pressure and pulse pressure, thereby causing incident hypertension and cardiovascular disease. In turn, prolonged high mechanical stress, particularly wall strain, applied to the arterial wall during vigorous exercise, results in stiffening and calcification of tunica media, accelerated arterial aging, and cardiovascular disease events. In both scenarios, the tunica media is the primary target of mechanical stress and the first to respond to hemodynamic changes. The cyclical nature of these impacts confounds the results of each because they are not mutually exclusive. IMPLICATIONS The role of stress in the tunica media appears to be overlooked despite its relevance, and further research into new primary preventive therapies is needed aside from cautioning the role of vigorous exercise in the elderly population.
Collapse
Affiliation(s)
- Hanane Belhoul-Fakir
- Curtin Medical School, Curtin University, Bentley, Perth, Western Australia, Australia; Targeted Drug Delivery, Imaging & Therapy, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia; Heart & Vascular Research Institute, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia.
| | - Michael Lawrence Brown
- School of Population Health, Curtin University, Bently, Perth, Western Australia, Australia
| | - Peter L Thompson
- Heart & Vascular Research Institute, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia
| | - Juliana Hamzah
- Curtin Medical School, Curtin University, Bentley, Perth, Western Australia, Australia; Targeted Drug Delivery, Imaging & Therapy, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia; Heart & Vascular Research Institute, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia
| | - Shirley Jansen
- Curtin Medical School, Curtin University, Bentley, Perth, Western Australia, Australia; Targeted Drug Delivery, Imaging & Therapy, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia; Heart & Vascular Research Institute, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, Western Australia, Australia; Department of Vascular and Endovascular Surgery, Sir Charles Gairdner Hospital, Nedlands, Perth, Western Australia, Australia.
| |
Collapse
|
3
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [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
|
4
|
Gao X, Xu Z, Chen C, Hao P, He F, Zhang X. Full-scale numerical simulation of hemodynamics based on left ventricular assist device. Front Physiol 2023; 14:1192610. [PMID: 37304828 PMCID: PMC10248007 DOI: 10.3389/fphys.2023.1192610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/16/2023] [Indexed: 06/13/2023] Open
Abstract
Ventricular assist devices have been widely used and accepted to treat patients with end-stage heart failure. The role of VAD is to improve circulatory dysfunction or temporarily maintain the circulatory status of patients. In order to be closer to the medical practice, a multi-Domain model of the left ventricular coupled axial flow artificial heart was considered to study the effect of its hemodynamics on the aorta. Because whether LVAD itself was connected between the left ventricular apex and the ascending aorta by catheter in the loop was not very important for the analysis of simulation results, on the premise of ensuring the multi-Domain simulation, the simulation data of the import and export ends of LVAD were imported to simplify the model. In this paper, the hemodynamic parameters in the ascending aorta, such as blood flow velocity vector, wall shear stress distribution, vorticity current intensity, vorticity flow generation, etc., have been calculated. The numerical conclusion of this study showed the vorticity intensity under LVAD was significantly higher than that under patients' conditions and the overall condition is similar to that of a healthy ventricular spin, which can improve heart failure patients' condition while minimizing other pitfalls. In addition, high velocity blood flow during left ventricular assist surgery is mainly concentrated near the lining of the ascending aorta lumen. What's more, the paper proposes to use Q criterion to determine the generation of vorticity flow. The Q criterion of LVAD is much higher than that of patients with heart failure, and the closer the LVAD is to the wall of the ascending aorta, the greater the Q criterion is. All these are beneficial to the effectiveness of LVAD in the treatment of heart failure patients and provide clinical suggestions for the LVAD implantation in clinical practice.
Collapse
Affiliation(s)
- Xinyi Gao
- Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing, China
| | - Zhike Xu
- Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing, China
| | - Chenghan Chen
- Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing, China
| | - Pengfei Hao
- Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing, China
- Tsinghua University (School of Materials Science and Engineering)—AVIC Aerodynamics Research Institute Joint Research Center for Advanced Materials and Anti-Icing, Beijing, China
| | - Feng He
- Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing, China
| | - Xiwen Zhang
- Applied Mechanics Laboratory, Department of Engineering Mechanics, Tsinghua University, Beijing, China
| |
Collapse
|
5
|
Jiang Y, Ji JY. Progerin-Induced Impairment in Wound Healing and Proliferation in Vascular Endothelial Cells. FRONTIERS IN AGING 2022; 3:844885. [PMID: 35821855 PMCID: PMC9261432 DOI: 10.3389/fragi.2022.844885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 01/27/2022] [Indexed: 11/29/2022]
Abstract
Progerin as a mutated isoform of lamin A protein was first known to induce premature atherosclerosis progression in patients with Hutchinson-Gilford progeria syndrome (HGPS), and its role in provoking an inflammatory response in vascular cells and accelerating cell senescence has been investigated recently. However, how progerin triggers endothelial dysfunction that often occurs at the early stage of atherosclerosis in a mechanical environment has not been studied intensively. Here, we generated a stable endothelial cell line that expressed progerin and examined its effects on endothelial wound repair under laminar flow. We found decreased wound healing rate in progerin-expressing ECs under higher shear stress compared with those under low shear. Furthermore, the decreased wound recovery could be due to reduced number of cells at late mitosis, suggesting potential interference by progerin with endothelial proliferation. These findings provided insights into how progerin affects endothelial mechanotransduction and may contribute to the disruption of endothelial integrity in HGPS vasculature, as we continue to examine the mechanistic effect of progerin in shear-induced endothelial functions.
Collapse
|
6
|
Wang T, Zhou J, Zhang X, Wu Y, Jin K, Wang Y, Xu R, Yang G, Li W, Jiao L. X-box Binding Protein 1: An Adaptor in the Pathogenesis of Atherosclerosis. Aging Dis 2022; 14:350-369. [PMID: 37008067 PMCID: PMC10017146 DOI: 10.14336/ad.2022.0824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/24/2022] [Indexed: 11/18/2022] Open
Abstract
Atherosclerosis (AS), the formation of fibrofatty lesions in the vessel wall, is the primary cause of heart disease and stroke and is closely associated with aging. Disrupted metabolic homeostasis is a primary feature of AS and leads to endoplasmic reticulum (ER) stress, which is an abnormal accumulation of unfolded proteins. By orchestrating signaling cascades of the unfolded protein response (UPR), ER stress functions as a double-edged sword in AS, where adaptive UPR triggers synthetic metabolic processes to restore homeostasis, whereas the maladaptive response programs the cell to the apoptotic pathway. However, little is known regarding their precise coordination. Herein, an advanced understanding of the role of UPR in the pathological process of AS is reviewed. In particular, we focused on a critical mediator of the UPR, X-box binding protein 1 (XBP1), and its important role in balancing adaptive and maladaptive responses. The XBP1 mRNA is processed from the unspliced isoform (XBP1u) to the spliced isoform of XBP1 (XBP1s). Compared with XBP1u, XBP1s predominantly functions downstream of inositol-requiring enzyme-1α (IRE1α) and transcript genes involved in protein quality control, inflammation, lipid metabolism, carbohydrate metabolism, and calcification, which are critical for the pathogenesis of AS. Thus, the IRE1α/XBP1 axis is a promising pharmaceutical candidate against AS.
Collapse
Affiliation(s)
- Tao Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- China International Neuroscience Institute (China-INI), Beijing, China.
| | - Jia Zhou
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| | - Xiao Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- China International Neuroscience Institute (China-INI), Beijing, China.
| | - Yujie Wu
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China.
| | - Kehan Jin
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| | - Yilin Wang
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.
| | - Ran Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- China International Neuroscience Institute (China-INI), Beijing, China.
| | - Ge Yang
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China.
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China.
- Correspondence should be addressed to: Dr. Ge Yang, Chinese Academy of Sciences, Beijing, China. , Dr. Wenjing Li, Chinese Academy of Sciences, Beijing, China. ; Dr. Liqun Jiao, Xuanwu Hospital, Capital Medical University, Beijing, China. .
| | - Wenjing Li
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China.
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China.
- Correspondence should be addressed to: Dr. Ge Yang, Chinese Academy of Sciences, Beijing, China. , Dr. Wenjing Li, Chinese Academy of Sciences, Beijing, China. ; Dr. Liqun Jiao, Xuanwu Hospital, Capital Medical University, Beijing, China. .
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- China International Neuroscience Institute (China-INI), Beijing, China.
- Department of Interventional Radiology, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Correspondence should be addressed to: Dr. Ge Yang, Chinese Academy of Sciences, Beijing, China. , Dr. Wenjing Li, Chinese Academy of Sciences, Beijing, China. ; Dr. Liqun Jiao, Xuanwu Hospital, Capital Medical University, Beijing, China. .
| |
Collapse
|
7
|
Jiang LP, Yu XH, Chen JZ, Hu M, Zhang YK, Lin HL, Tang WY, He PP, Ouyang XP. Histone Deacetylase 3: A Potential Therapeutic Target for Atherosclerosis. Aging Dis 2022; 13:773-786. [PMID: 35656103 PMCID: PMC9116907 DOI: 10.14336/ad.2021.1116] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 11/16/2021] [Indexed: 11/17/2022] Open
Abstract
Atherosclerosis, the pathological basis of most cardiovascular disease, is characterized by plaque formation in the intima. Secondary lesions include intraplaque hemorrhage, plaque rupture, and local thrombosis. Vascular endothelial function impairment and smooth muscle cell migration lead to vascular dysfunction, which is conducive to the formation of macrophage-derived foam cells and aggravates inflammatory response and lipid accumulation that cause atherosclerosis. Histone deacetylase (HDAC) is an epigenetic modifying enzyme closely related to chromatin structure and gene transcriptional regulation. Emerging studies have demonstrated that the Class I member HDAC3 of the HDAC super family has cell-specific functions in atherosclerosis, including 1) maintenance of endothelial integrity and functions, 2) regulation of vascular smooth muscle cell proliferation and migration, 3) modulation of macrophage phenotype, and 4) influence on foam cell formation. Although several studies have shown that HDAC3 may be a promising therapeutic target, only a few HDAC3-selective inhibitors have been thoroughly researched and reported. Here, we specifically summarize the impact of HDAC3 and its inhibitors on vascular function, inflammation, lipid accumulation, and plaque stability in the development of atherosclerosis with the hopes of opening up new opportunities for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Li-Ping Jiang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hunan, China.
| | - Xiao-Hua Yu
- Institute of Clinical Medicine, the Second Affiliated Hospital of Hainan Medical University, Haikou, China.
| | - Jin-Zhi Chen
- Department of Physiology, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hunan, China.
| | - Mi Hu
- Department of Physiology, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hunan, China.
| | - Yang-Kai Zhang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hunan, China.
| | - Hui-Ling Lin
- Department of Physiology, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hunan, China.
| | - Wan-Ying Tang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hunan, China.
| | - Ping-Ping He
- School of Nursing, University of South China, Hunan, China
- Correspondence should be addressed to: Dr. Ping-Ping He, School of Nursing, University of South China, Hunan, China. and Dr. Xin-Ping Ouyang, Department of Physiology, University of South China, Hunan, China. .
| | - Xin-Ping Ouyang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hunan, China.
- Correspondence should be addressed to: Dr. Ping-Ping He, School of Nursing, University of South China, Hunan, China. and Dr. Xin-Ping Ouyang, Department of Physiology, University of South China, Hunan, China. .
| |
Collapse
|
8
|
Botts SR, Fish JE, Howe KL. Dysfunctional Vascular Endothelium as a Driver of Atherosclerosis: Emerging Insights Into Pathogenesis and Treatment. Front Pharmacol 2021; 12:787541. [PMID: 35002720 PMCID: PMC8727904 DOI: 10.3389/fphar.2021.787541] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/06/2021] [Indexed: 12/28/2022] Open
Abstract
Atherosclerosis, the chronic accumulation of cholesterol-rich plaque within arteries, is associated with a broad spectrum of cardiovascular diseases including myocardial infarction, aortic aneurysm, peripheral vascular disease, and stroke. Atherosclerotic cardiovascular disease remains a leading cause of mortality in high-income countries and recent years have witnessed a notable increase in prevalence within low- and middle-income regions of the world. Considering this prominent and evolving global burden, there is a need to identify the cellular mechanisms that underlie the pathogenesis of atherosclerosis to discover novel therapeutic targets for preventing or mitigating its clinical sequelae. Despite decades of research, we still do not fully understand the complex cell-cell interactions that drive atherosclerosis, but new investigative approaches are rapidly shedding light on these essential mechanisms. The vascular endothelium resides at the interface of systemic circulation and the underlying vessel wall and plays an essential role in governing pathophysiological processes during atherogenesis. In this review, we present emerging evidence that implicates the activated endothelium as a driver of atherosclerosis by directing site-specificity of plaque formation and by promoting plaque development through intracellular processes, which regulate endothelial cell proliferation and turnover, metabolism, permeability, and plasticity. Moreover, we highlight novel mechanisms of intercellular communication by which endothelial cells modulate the activity of key vascular cell populations involved in atherogenesis, and discuss how endothelial cells contribute to resolution biology - a process that is dysregulated in advanced plaques. Finally, we describe important future directions for preclinical atherosclerosis research, including epigenetic and targeted therapies, to limit the progression of atherosclerosis in at-risk or affected patients.
Collapse
Affiliation(s)
- Steven R. Botts
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jason E. Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Kathryn L. Howe
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Division of Vascular Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
9
|
Imbalance between the circulating endothelium-derived apoptotic microparticles and the endothelial colony-forming units of progenitor cells in patients undergoing diagnostic coronary angiography. Adv Med Sci 2021; 66:396-402. [PMID: 34375819 DOI: 10.1016/j.advms.2021.07.011] [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: 02/16/2021] [Revised: 04/04/2021] [Accepted: 07/31/2021] [Indexed: 11/21/2022]
Abstract
PURPOSE The involvement of the circulating endothelium-derived microparticles (EMPs) and the endothelial progenitor cells (EPCs) has been shown in the pathogenesis of coronary artery disease (CAD). The current study aimed to explore whether the Friesinger index is associated with the levels of the apoptotic CD144+/CD31+/annexin V+ EMPs and the number of endothelial colony-forming units of progenitor cells in patients undergoing coronary angiography. PATIENTS AND METHODS Fifty-seven patients with a median age of 62 years (range: 48-84 years) were enrolled. Quantification of the apoptotic CD144+/CD31+/annexin V+ EMPs was performed by flow cytometry. The number of endothelial colony-forming units defined by CFU-Hill was assessed by cell culture. RESULTS There was a positive correlation between the Friesinger index and the circulating levels of the apoptotic CD144+/CD31+/annexin V+ EMPs (rho=0.817, p<0.001), whereas a negative correlation was found with the number of CFU-Hill (rho = - 0.649, p<0.001). Multivariable logistic analysis showed that the risk of having moderate/severe CAD was five times greater among male patients (OR:5.32; 95% CI: 1.19 - 16.33; p=0.038) and almost one and a half times higher among those with a higher level of apoptotic CD144+/CD31+/annexin V+ EMPs (OR:1.74; 95% CI: 1.23 - 2.28; p=0.001). Finally, the circulating levels of apoptotic EMPs labelled for CD144+/CD31+/annexin V+ presented a good discrimination of moderate/severe CAD, with an AUC of 0.85 (95% CI = 0.74 - 0.96; p< 0.001). CONCLUSIONS Moderate or severe CAD is associated with increased levels of apoptotic EMPs and reduced EPC colony-forming capacity, increasing the occurrence of endothelial injuries.
Collapse
|
10
|
Chen L, Shaw DWW, Dager SR, Corrigan NM, Chu B, Kleinhans NM, Kuhl PK, Hwang JN, Yuan C. Quantitative Assessment of the Intracranial Vasculature of Infants and Adults Using iCafe (Intracranial Artery Feature Extraction). Front Neurol 2021; 12:668298. [PMID: 34122310 PMCID: PMC8193571 DOI: 10.3389/fneur.2021.668298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/16/2021] [Indexed: 11/13/2022] Open
Abstract
Comprehensive quantification of intracranial artery features may help to assess and understand regional variations of blood supply during early brain development and aging. We analyzed vasculature features of 27 healthy infants during natural sleep, 13 infants at 7-months (7.3 ± 1.0 month), and 14 infants at 12-months (11.7 ± 0.4 month), and 13 older healthy, awake adults (62.8 ± 8.7 years) to investigate age-related vascular differences as a preliminary study of vascular changes associated with brain development. 3D time-of-flight (TOF) magnetic resonance angiography (MRA) acquisitions were processed in iCafe, a technique to quantify arterial features (http://icafe.clatfd.cn), to characterize intracranial vasculature. Overall, adult subjects were found to have increased ACA length, tortuosity, and vasculature density compared to both 7-month-old and 12-month-old infants, as well as MCA length compared to 7-month-old infants. No brain laterality differences were observed for any vascular measures in either infant or adult age groups. Reduced skull and brain sharpness, indicative of increased head motion and brain/vascular pulsation, respectively, were observed in infants but not correlated with length, tortuosity, or vasculature density measures. Quantitative analysis of TOF MRA using iCafe may provide an objective approach for systematic study of infant brain vascular development and for clinical assessment of adult and pediatric brain vascular diseases.
Collapse
Affiliation(s)
- Li Chen
- University of Washington, Seattle, WA, United States
| | | | | | | | | | | | | | | | - Chun Yuan
- University of Washington, Seattle, WA, United States
| |
Collapse
|
11
|
Coronary Shear Stress after Implantation of Bioresorbable Scaffolds – a Modern Interdisciplinary Concept at the Border between Interventional Cardiology and Cardiac Imaging. JOURNAL OF INTERDISCIPLINARY MEDICINE 2019. [DOI: 10.2478/jim-2019-0007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Abstract
Bioresorbable scaffolds/stents offer new and exciting perspectives in the treatment of patients with acute coronary syndromes, especially after the recent development of invasive imaging techniques, such as optical coherence tomography, which allow complete assessment of vascu-lar segments. A particular advantage of bioresorbable scaffolds is that once the biosorption of the scaffold is complete, the vascular segment regains its normal physiological functions, thus eliminating the risk of late complications. New studies show the importance of shear stress in the progression of vascular atherosclerosis or in accelerating endothelial turnover. Based on the current knowledge in this field, a future standardized determination of shear stress may help in the long-term follow-up of patients that have suffered or are at risk of developing an acute coronary syndrome.
Collapse
|
12
|
Savoji H, Mohammadi MH, Rafatian N, Toroghi MK, Wang EY, Zhao Y, Korolj A, Ahadian S, Radisic M. Cardiovascular disease models: A game changing paradigm in drug discovery and screening. Biomaterials 2019; 198:3-26. [PMID: 30343824 PMCID: PMC6397087 DOI: 10.1016/j.biomaterials.2018.09.036] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 09/11/2018] [Accepted: 09/22/2018] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease is the leading cause of death worldwide. Although investment in drug discovery and development has been sky-rocketing, the number of approved drugs has been declining. Cardiovascular toxicity due to therapeutic drug use claims the highest incidence and severity of adverse drug reactions in late-stage clinical development. Therefore, to address this issue, new, additional, replacement and combinatorial approaches are needed to fill the gap in effective drug discovery and screening. The motivation for developing accurate, predictive models is twofold: first, to study and discover new treatments for cardiac pathologies which are leading in worldwide morbidity and mortality rates; and second, to screen for adverse drug reactions on the heart, a primary risk in drug development. In addition to in vivo animal models, in vitro and in silico models have been recently proposed to mimic the physiological conditions of heart and vasculature. Here, we describe current in vitro, in vivo, and in silico platforms for modelling healthy and pathological cardiac tissues and their advantages and disadvantages for drug screening and discovery applications. We review the pathophysiology and the underlying pathways of different cardiac diseases, as well as the new tools being developed to facilitate their study. We finally suggest a roadmap for employing these non-animal platforms in assessing drug cardiotoxicity and safety.
Collapse
Affiliation(s)
- Houman Savoji
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada
| | - Mohammad Hossein Mohammadi
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada; Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada
| | - Naimeh Rafatian
- Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada
| | - Masood Khaksar Toroghi
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada
| | - Erika Yan Wang
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada
| | - Yimu Zhao
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada
| | - Anastasia Korolj
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada
| | - Samad Ahadian
- Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada
| | - Milica Radisic
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 170 College St, Toronto, Ontario, M5S 3G9, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College St, Toronto, Ontario, M5S 3E5, Canada; Toronto General Research Institute, University Health Network, University of Toronto, 200 Elizabeth St, Toronto, Ontario, M5G 2C4, Canada.
| |
Collapse
|
13
|
Head T, Daunert S, Goldschmidt-Clermont PJ. The Aging Risk and Atherosclerosis: A Fresh Look at Arterial Homeostasis. Front Genet 2017; 8:216. [PMID: 29312440 PMCID: PMC5735066 DOI: 10.3389/fgene.2017.00216] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/04/2017] [Indexed: 12/14/2022] Open
Abstract
A considerable volume of research over the last decade has focused on understanding the fundamental mechanisms for the progression of atherosclerosis-the underlying cause for the vast majority of all cardiovascular (CVD)-related complications. Aging is the dominant risk factor for clinically significant atherosclerotic lesion formation, yet the heightened impact of aging on the disease is not accounted for by changes in traditional risk factors, such as lack of physical activity, smoking, hypertension, hyperlipidemia, or diabetes mellitus. This review will examine the pathological and biochemical processes of atherosclerotic plaque formation and growth, with particular focus on the aging risk vis-a-vis arterial homeostasis. Particular focus will be placed on the impact of a number of important contributors to arterial homeostasis including bone marrow (BM)-derived vascular progenitor cells, differential monocyte subpopulations, and the role of cellular senescence. Finally, this review will explore many critical observations in the way the disease process has been reassessed both by clinicians and researchers, and will highlight recent advances in this field that have provided a greater understanding of this aging-driven disease.
Collapse
Affiliation(s)
- Trajen Head
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Sylvia Daunert
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, United States
| | | |
Collapse
|
14
|
Sargowo D, Ovianti N, Susilowati E, Ubaidillah N, Widya Nugraha A, Vitriyaturrida, Siwi Proboretno K, Failasufi M, Ramadhan F, Wulandari H, Waranugraha Y, Hayuning Putri D. The role of polysaccharide peptide of Ganoderma lucidum as a potent antioxidant against atherosclerosis in high risk and stable angina patients. Indian Heart J 2017; 70:608-614. [PMID: 30392496 PMCID: PMC6204443 DOI: 10.1016/j.ihj.2017.12.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 10/23/2017] [Accepted: 12/13/2017] [Indexed: 12/22/2022] Open
Abstract
Objectives Antioxidants can reduce oxidative radicals that affect the early phase of atherogenesis, that is endothelial dysfunction. Polysaccharide Peptide (PsP) derived from Ganoderma lucidum has an active substance in the form of β-glucan. Previous studies have proven the PsP of Ganoderma lucidum as an effective antioxidant in atherosclerotic rats and shows no toxicity in animal model. This study aims to prove the effect of PsP as potent antioxidant in high risk and stable angina patients. Method This is a clinical trial conducted to 37 high risk and 34 stable angina patients, which were determined based on ESC Stable CAD Guidelines and Framingham risk score, with pre and post test design without control group. The parameters are superoxide dimustase (SOD) and malondialdehyde (MDA) concentration, circulating endothelial cell (CEC) and endothelial progenitor cell (EPC) counts. The patients were given PsP 750 mg/day in 3 divided dose for 90 days. Paired t-test was performed for normally distributed data, and Wilcoxon test for not normally distributed data, and significant level of p ≤ 0,05. Results SOD level in high risk patients slightly increased but not statistically significant with p = 0,22. Level of SOD in stable angina group significantly increased with p = 0,001. MDA concentration significantly reduced in high risk and stable angina patients with p = 0.000. CEC significantly reduced both in high risk and stable angina patients, with p = 0.000 in both groups. EPC count significantly reduced in high risk and stable angina with p = 0.000. Conclusion PsP of Ganoderma lucidum is a potent antioxidant against pathogenesis of atherosclerosis in stable angina and high risk patients
Collapse
Affiliation(s)
- Djanggan Sargowo
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Brawijaya University/Saiful Anwar General Hospital, Malang, Indonesia
| | - Nadia Ovianti
- Master Program of Biomedical Science, Faculty of Medicine, Brawijaya University, Malang, Indonesia.
| | - Eliana Susilowati
- Master Program of Biomedical Science, Faculty of Medicine, Brawijaya University, Malang, Indonesia
| | - Nizamuddin Ubaidillah
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Brawijaya University/Saiful Anwar General Hospital, Malang, Indonesia
| | - Adriyawan Widya Nugraha
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Brawijaya University/Saiful Anwar General Hospital, Malang, Indonesia
| | - Vitriyaturrida
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Brawijaya University/Saiful Anwar General Hospital, Malang, Indonesia
| | - Kartika Siwi Proboretno
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Brawijaya University/Saiful Anwar General Hospital, Malang, Indonesia
| | - Mirza Failasufi
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Brawijaya University/Saiful Anwar General Hospital, Malang, Indonesia
| | - Fadhli Ramadhan
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Brawijaya University/Saiful Anwar General Hospital, Malang, Indonesia
| | - Hesti Wulandari
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Brawijaya University/Saiful Anwar General Hospital, Malang, Indonesia
| | - Yoga Waranugraha
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Brawijaya University/Saiful Anwar General Hospital, Malang, Indonesia
| | - Dinarsari Hayuning Putri
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Brawijaya University/Saiful Anwar General Hospital, Malang, Indonesia
| |
Collapse
|
15
|
Pan L, Hong Z, Yu L, Gao Y, Zhang R, Feng H, Su L, Wang G. Shear stress induces human aortic endothelial cell apoptosis via interleukin‑1 receptor‑associated kinase 2‑induced endoplasmic reticulum stress. Mol Med Rep 2017; 16:7205-7212. [PMID: 28944871 PMCID: PMC5865847 DOI: 10.3892/mmr.2017.7524] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 05/12/2017] [Indexed: 02/05/2023] Open
Abstract
Atherosclerosis is characterized by localized lesions distributed in the arterial tree due to the shear stress produced by blood flow. Endothelial cells are directly affected by alterations in blood flow. Dysfunction and injury to endothelial cells has been hypothesized to initiate the pathological processes of atherosclerosis. The present study aimed to investigate the mechanism of shear stress‑induced endothelial cellular apoptosis. Shear stress was generated using an artificial device to mimic the impact of disturbed blood flow on cultured human aortic endothelial cells (HAECs). Cellular apoptosis was assessed using a terminal deoxynucleotidyl transferase dUTP nick end labeling assay; an ELISA assay was used to detect the produced interleukin (IL)‑1β; specific small interfering (si)RNA was used to knockdown the expression of interleukin‑1 receptor‑associated kinase 2 (IRAK2) in HAECs and the expression levels of 78 kDa glucose‑regulated protein, DNA damage‑inducible transcript 3 protein (CHOP), IRAK2 and IL‑1β were evaluated using western blotting. The results of the present study demonstrated that artificial shear stress induced endoplasmic reticulum (ER) stress, IL‑1β production and apoptosis in HAECs in a time‑dependent manner. The inhibition of ER stress, and treatment with interleukin‑1 receptor antagonist protein and siRNA against IRAK2 attenuated shear stress‑induced CHOP signaling‑mediated cellular apoptosis. Therefore, overproduction of IL‑1β exacerbated shear stress‑induced ER stress‑mediated apoptosis via the IRAK2/CHOP signaling pathway in endothelial cells.
Collapse
Affiliation(s)
- Longfei Pan
- Department of Emergency Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Zhou Hong
- Department of Emergency Medicine, The Fifth Hospital of Xi'an, Xi'an, Shaanxi 710082, P.R. China
| | - Lei Yu
- Department of Basic Medicine, Xi'an Medical College, Xi'an, Shaanxi 710068, P.R. China
| | - Yanxia Gao
- Department of Emergency Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Rui Zhang
- Department of Emergency Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Hui Feng
- Department of Emergency Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Lijuan Su
- Department of Emergency Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Gang Wang
- Department of Emergency Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
16
|
Yu B, Kiechl S, Qi D, Wang X, Song Y, Weger S, Mayr A, Le Bras A, Karamariti E, Zhang Z, Barco Barrantes ID, Niehrs C, Schett G, Hu Y, Wang W, Willeit J, Qu A, Xu Q. A Cytokine-Like Protein Dickkopf-Related Protein 3 Is Atheroprotective. Circulation 2017; 136:1022-1036. [PMID: 28674110 PMCID: PMC5598907 DOI: 10.1161/circulationaha.117.027690] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 06/06/2017] [Indexed: 12/28/2022]
Abstract
Supplemental Digital Content is available in the text. Background: Dickkopf-related protein 3 (DKK3) is a secreted protein that is involved in the regulation of cardiac remodeling and vascular smooth muscle cell differentiation, but little is known about its role in atherosclerosis. Methods: We tested the hypothesis that DKK3 is atheroprotective using both epidemiological and experimental approaches. Blood DKK3 levels were measured in the Bruneck Study in 2000 (n=684) and then in 2005 (n=574). DKK3-deficient mice were crossed with apolipoprotein E-/- mice to evaluate atherosclerosis development and vessel injury-induced neointimal formation. Endothelial cell migration and the underlying mechanisms were studied using in vitro cell culture models. Results: In the prospective population-based Bruneck Study, the level of plasma DKK3 was inversely related to carotid artery intima-media thickness and 5-year progression of carotid atherosclerosis independently from standard risk factors for atherosclerosis. Experimentally, we analyzed the area of atherosclerotic lesions, femoral artery injury-induced reendothelialization, and neointima formation in both DKK3-/-/apolipoprotein E-/- and DKK3+/+/apolipoprotein E-/- mice. It was demonstrated that DKK3 deficiency accelerated atherosclerosis and delayed reendothelialization with consequently exacerbated neointima formation. To explore the underlying mechanisms, we performed transwell and scratch migration assays using cultured human endothelial cells, which exhibited a significant induction in cell migration in response to DKK3 stimulation. This DKK3-induced migration activated ROR2 and DVL1, activated Rac1 GTPases, and upregulated JNK and c-jun phosphorylation in endothelial cells. Knockdown of the ROR2 receptor using specific siRNA or transfection of a dominant-negative form of Rac1 in endothelial cells markedly inhibited cell migration and downstream JNK and c-jun phosphorylation. Conclusions: This study provides the evidence for a role of DKK3 in the protection against atherosclerosis involving endothelial migration and repair, with great therapeutic potential implications against atherosclerosis.
Collapse
Affiliation(s)
- Baoqi Yu
- From Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (B.Y., X.W., A.L.B., E.K., Z.Z., Y.H., Q.X.); Department of Neurology, Medical University of Innsbruck, Austria (S.K., J.W.); Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China (D.Q., Y.S., A.Q.); Department of Internal and Laboratory Medicine, Bruneck Hospital, Italy (S.W., A.M.); Division of Molecular Embryology, German Cancer Research Center (DKFZ) Heidelberg Germany and Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH) Alliance, Heidelberg, Germany (I.d.B.B., C.N.); Institute of Molecular Biology, Mainz, Germany (C.N.); Department of Internal Medicine, Institute for Clinical Immunology, Friedrich-Alexander-University Erlangen-Nuremberg, Germany (G.S.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (Y.H., Q.X.); and Institute of Bioengineering, Queen Mary University of London, United Kingdom (W.W.)
| | - Stefan Kiechl
- From Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (B.Y., X.W., A.L.B., E.K., Z.Z., Y.H., Q.X.); Department of Neurology, Medical University of Innsbruck, Austria (S.K., J.W.); Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China (D.Q., Y.S., A.Q.); Department of Internal and Laboratory Medicine, Bruneck Hospital, Italy (S.W., A.M.); Division of Molecular Embryology, German Cancer Research Center (DKFZ) Heidelberg Germany and Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH) Alliance, Heidelberg, Germany (I.d.B.B., C.N.); Institute of Molecular Biology, Mainz, Germany (C.N.); Department of Internal Medicine, Institute for Clinical Immunology, Friedrich-Alexander-University Erlangen-Nuremberg, Germany (G.S.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (Y.H., Q.X.); and Institute of Bioengineering, Queen Mary University of London, United Kingdom (W.W.)
| | - Dan Qi
- From Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (B.Y., X.W., A.L.B., E.K., Z.Z., Y.H., Q.X.); Department of Neurology, Medical University of Innsbruck, Austria (S.K., J.W.); Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China (D.Q., Y.S., A.Q.); Department of Internal and Laboratory Medicine, Bruneck Hospital, Italy (S.W., A.M.); Division of Molecular Embryology, German Cancer Research Center (DKFZ) Heidelberg Germany and Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH) Alliance, Heidelberg, Germany (I.d.B.B., C.N.); Institute of Molecular Biology, Mainz, Germany (C.N.); Department of Internal Medicine, Institute for Clinical Immunology, Friedrich-Alexander-University Erlangen-Nuremberg, Germany (G.S.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (Y.H., Q.X.); and Institute of Bioengineering, Queen Mary University of London, United Kingdom (W.W.)
| | - Xiaocong Wang
- From Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (B.Y., X.W., A.L.B., E.K., Z.Z., Y.H., Q.X.); Department of Neurology, Medical University of Innsbruck, Austria (S.K., J.W.); Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China (D.Q., Y.S., A.Q.); Department of Internal and Laboratory Medicine, Bruneck Hospital, Italy (S.W., A.M.); Division of Molecular Embryology, German Cancer Research Center (DKFZ) Heidelberg Germany and Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH) Alliance, Heidelberg, Germany (I.d.B.B., C.N.); Institute of Molecular Biology, Mainz, Germany (C.N.); Department of Internal Medicine, Institute for Clinical Immunology, Friedrich-Alexander-University Erlangen-Nuremberg, Germany (G.S.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (Y.H., Q.X.); and Institute of Bioengineering, Queen Mary University of London, United Kingdom (W.W.)
| | - Yanting Song
- From Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (B.Y., X.W., A.L.B., E.K., Z.Z., Y.H., Q.X.); Department of Neurology, Medical University of Innsbruck, Austria (S.K., J.W.); Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China (D.Q., Y.S., A.Q.); Department of Internal and Laboratory Medicine, Bruneck Hospital, Italy (S.W., A.M.); Division of Molecular Embryology, German Cancer Research Center (DKFZ) Heidelberg Germany and Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH) Alliance, Heidelberg, Germany (I.d.B.B., C.N.); Institute of Molecular Biology, Mainz, Germany (C.N.); Department of Internal Medicine, Institute for Clinical Immunology, Friedrich-Alexander-University Erlangen-Nuremberg, Germany (G.S.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (Y.H., Q.X.); and Institute of Bioengineering, Queen Mary University of London, United Kingdom (W.W.)
| | - Siegfried Weger
- From Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (B.Y., X.W., A.L.B., E.K., Z.Z., Y.H., Q.X.); Department of Neurology, Medical University of Innsbruck, Austria (S.K., J.W.); Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China (D.Q., Y.S., A.Q.); Department of Internal and Laboratory Medicine, Bruneck Hospital, Italy (S.W., A.M.); Division of Molecular Embryology, German Cancer Research Center (DKFZ) Heidelberg Germany and Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH) Alliance, Heidelberg, Germany (I.d.B.B., C.N.); Institute of Molecular Biology, Mainz, Germany (C.N.); Department of Internal Medicine, Institute for Clinical Immunology, Friedrich-Alexander-University Erlangen-Nuremberg, Germany (G.S.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (Y.H., Q.X.); and Institute of Bioengineering, Queen Mary University of London, United Kingdom (W.W.)
| | - Agnes Mayr
- From Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (B.Y., X.W., A.L.B., E.K., Z.Z., Y.H., Q.X.); Department of Neurology, Medical University of Innsbruck, Austria (S.K., J.W.); Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China (D.Q., Y.S., A.Q.); Department of Internal and Laboratory Medicine, Bruneck Hospital, Italy (S.W., A.M.); Division of Molecular Embryology, German Cancer Research Center (DKFZ) Heidelberg Germany and Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH) Alliance, Heidelberg, Germany (I.d.B.B., C.N.); Institute of Molecular Biology, Mainz, Germany (C.N.); Department of Internal Medicine, Institute for Clinical Immunology, Friedrich-Alexander-University Erlangen-Nuremberg, Germany (G.S.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (Y.H., Q.X.); and Institute of Bioengineering, Queen Mary University of London, United Kingdom (W.W.)
| | - Alexandra Le Bras
- From Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (B.Y., X.W., A.L.B., E.K., Z.Z., Y.H., Q.X.); Department of Neurology, Medical University of Innsbruck, Austria (S.K., J.W.); Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China (D.Q., Y.S., A.Q.); Department of Internal and Laboratory Medicine, Bruneck Hospital, Italy (S.W., A.M.); Division of Molecular Embryology, German Cancer Research Center (DKFZ) Heidelberg Germany and Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH) Alliance, Heidelberg, Germany (I.d.B.B., C.N.); Institute of Molecular Biology, Mainz, Germany (C.N.); Department of Internal Medicine, Institute for Clinical Immunology, Friedrich-Alexander-University Erlangen-Nuremberg, Germany (G.S.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (Y.H., Q.X.); and Institute of Bioengineering, Queen Mary University of London, United Kingdom (W.W.)
| | - Eirini Karamariti
- From Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (B.Y., X.W., A.L.B., E.K., Z.Z., Y.H., Q.X.); Department of Neurology, Medical University of Innsbruck, Austria (S.K., J.W.); Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China (D.Q., Y.S., A.Q.); Department of Internal and Laboratory Medicine, Bruneck Hospital, Italy (S.W., A.M.); Division of Molecular Embryology, German Cancer Research Center (DKFZ) Heidelberg Germany and Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH) Alliance, Heidelberg, Germany (I.d.B.B., C.N.); Institute of Molecular Biology, Mainz, Germany (C.N.); Department of Internal Medicine, Institute for Clinical Immunology, Friedrich-Alexander-University Erlangen-Nuremberg, Germany (G.S.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (Y.H., Q.X.); and Institute of Bioengineering, Queen Mary University of London, United Kingdom (W.W.)
| | - Zhongyi Zhang
- From Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (B.Y., X.W., A.L.B., E.K., Z.Z., Y.H., Q.X.); Department of Neurology, Medical University of Innsbruck, Austria (S.K., J.W.); Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China (D.Q., Y.S., A.Q.); Department of Internal and Laboratory Medicine, Bruneck Hospital, Italy (S.W., A.M.); Division of Molecular Embryology, German Cancer Research Center (DKFZ) Heidelberg Germany and Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH) Alliance, Heidelberg, Germany (I.d.B.B., C.N.); Institute of Molecular Biology, Mainz, Germany (C.N.); Department of Internal Medicine, Institute for Clinical Immunology, Friedrich-Alexander-University Erlangen-Nuremberg, Germany (G.S.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (Y.H., Q.X.); and Institute of Bioengineering, Queen Mary University of London, United Kingdom (W.W.)
| | - Ivan Del Barco Barrantes
- From Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (B.Y., X.W., A.L.B., E.K., Z.Z., Y.H., Q.X.); Department of Neurology, Medical University of Innsbruck, Austria (S.K., J.W.); Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China (D.Q., Y.S., A.Q.); Department of Internal and Laboratory Medicine, Bruneck Hospital, Italy (S.W., A.M.); Division of Molecular Embryology, German Cancer Research Center (DKFZ) Heidelberg Germany and Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH) Alliance, Heidelberg, Germany (I.d.B.B., C.N.); Institute of Molecular Biology, Mainz, Germany (C.N.); Department of Internal Medicine, Institute for Clinical Immunology, Friedrich-Alexander-University Erlangen-Nuremberg, Germany (G.S.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (Y.H., Q.X.); and Institute of Bioengineering, Queen Mary University of London, United Kingdom (W.W.)
| | - Christof Niehrs
- From Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (B.Y., X.W., A.L.B., E.K., Z.Z., Y.H., Q.X.); Department of Neurology, Medical University of Innsbruck, Austria (S.K., J.W.); Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China (D.Q., Y.S., A.Q.); Department of Internal and Laboratory Medicine, Bruneck Hospital, Italy (S.W., A.M.); Division of Molecular Embryology, German Cancer Research Center (DKFZ) Heidelberg Germany and Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH) Alliance, Heidelberg, Germany (I.d.B.B., C.N.); Institute of Molecular Biology, Mainz, Germany (C.N.); Department of Internal Medicine, Institute for Clinical Immunology, Friedrich-Alexander-University Erlangen-Nuremberg, Germany (G.S.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (Y.H., Q.X.); and Institute of Bioengineering, Queen Mary University of London, United Kingdom (W.W.)
| | - Georg Schett
- From Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (B.Y., X.W., A.L.B., E.K., Z.Z., Y.H., Q.X.); Department of Neurology, Medical University of Innsbruck, Austria (S.K., J.W.); Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China (D.Q., Y.S., A.Q.); Department of Internal and Laboratory Medicine, Bruneck Hospital, Italy (S.W., A.M.); Division of Molecular Embryology, German Cancer Research Center (DKFZ) Heidelberg Germany and Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH) Alliance, Heidelberg, Germany (I.d.B.B., C.N.); Institute of Molecular Biology, Mainz, Germany (C.N.); Department of Internal Medicine, Institute for Clinical Immunology, Friedrich-Alexander-University Erlangen-Nuremberg, Germany (G.S.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (Y.H., Q.X.); and Institute of Bioengineering, Queen Mary University of London, United Kingdom (W.W.)
| | - Yanhua Hu
- From Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (B.Y., X.W., A.L.B., E.K., Z.Z., Y.H., Q.X.); Department of Neurology, Medical University of Innsbruck, Austria (S.K., J.W.); Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China (D.Q., Y.S., A.Q.); Department of Internal and Laboratory Medicine, Bruneck Hospital, Italy (S.W., A.M.); Division of Molecular Embryology, German Cancer Research Center (DKFZ) Heidelberg Germany and Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH) Alliance, Heidelberg, Germany (I.d.B.B., C.N.); Institute of Molecular Biology, Mainz, Germany (C.N.); Department of Internal Medicine, Institute for Clinical Immunology, Friedrich-Alexander-University Erlangen-Nuremberg, Germany (G.S.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (Y.H., Q.X.); and Institute of Bioengineering, Queen Mary University of London, United Kingdom (W.W.)
| | - Wen Wang
- From Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (B.Y., X.W., A.L.B., E.K., Z.Z., Y.H., Q.X.); Department of Neurology, Medical University of Innsbruck, Austria (S.K., J.W.); Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China (D.Q., Y.S., A.Q.); Department of Internal and Laboratory Medicine, Bruneck Hospital, Italy (S.W., A.M.); Division of Molecular Embryology, German Cancer Research Center (DKFZ) Heidelberg Germany and Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH) Alliance, Heidelberg, Germany (I.d.B.B., C.N.); Institute of Molecular Biology, Mainz, Germany (C.N.); Department of Internal Medicine, Institute for Clinical Immunology, Friedrich-Alexander-University Erlangen-Nuremberg, Germany (G.S.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (Y.H., Q.X.); and Institute of Bioengineering, Queen Mary University of London, United Kingdom (W.W.)
| | - Johann Willeit
- From Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (B.Y., X.W., A.L.B., E.K., Z.Z., Y.H., Q.X.); Department of Neurology, Medical University of Innsbruck, Austria (S.K., J.W.); Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China (D.Q., Y.S., A.Q.); Department of Internal and Laboratory Medicine, Bruneck Hospital, Italy (S.W., A.M.); Division of Molecular Embryology, German Cancer Research Center (DKFZ) Heidelberg Germany and Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH) Alliance, Heidelberg, Germany (I.d.B.B., C.N.); Institute of Molecular Biology, Mainz, Germany (C.N.); Department of Internal Medicine, Institute for Clinical Immunology, Friedrich-Alexander-University Erlangen-Nuremberg, Germany (G.S.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (Y.H., Q.X.); and Institute of Bioengineering, Queen Mary University of London, United Kingdom (W.W.)
| | - Aijuan Qu
- From Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (B.Y., X.W., A.L.B., E.K., Z.Z., Y.H., Q.X.); Department of Neurology, Medical University of Innsbruck, Austria (S.K., J.W.); Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China (D.Q., Y.S., A.Q.); Department of Internal and Laboratory Medicine, Bruneck Hospital, Italy (S.W., A.M.); Division of Molecular Embryology, German Cancer Research Center (DKFZ) Heidelberg Germany and Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH) Alliance, Heidelberg, Germany (I.d.B.B., C.N.); Institute of Molecular Biology, Mainz, Germany (C.N.); Department of Internal Medicine, Institute for Clinical Immunology, Friedrich-Alexander-University Erlangen-Nuremberg, Germany (G.S.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (Y.H., Q.X.); and Institute of Bioengineering, Queen Mary University of London, United Kingdom (W.W.)
| | - Qingbo Xu
- From Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom (B.Y., X.W., A.L.B., E.K., Z.Z., Y.H., Q.X.); Department of Neurology, Medical University of Innsbruck, Austria (S.K., J.W.); Department of Physiology and Pathophysiology, Capital Medical University, Beijing, China (D.Q., Y.S., A.Q.); Department of Internal and Laboratory Medicine, Bruneck Hospital, Italy (S.W., A.M.); Division of Molecular Embryology, German Cancer Research Center (DKFZ) Heidelberg Germany and Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH) Alliance, Heidelberg, Germany (I.d.B.B., C.N.); Institute of Molecular Biology, Mainz, Germany (C.N.); Department of Internal Medicine, Institute for Clinical Immunology, Friedrich-Alexander-University Erlangen-Nuremberg, Germany (G.S.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (Y.H., Q.X.); and Institute of Bioengineering, Queen Mary University of London, United Kingdom (W.W.)
| |
Collapse
|
17
|
Trimethylamine N-oxide in atherogenesis: impairing endothelial self-repair capacity and enhancing monocyte adhesion. Biosci Rep 2017; 37:BSR20160244. [PMID: 28153917 PMCID: PMC5333780 DOI: 10.1042/bsr20160244] [Citation(s) in RCA: 175] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 01/04/2023] Open
Abstract
Several studies have reported a strong association between high plasma level of trimethylamine N-oxide (TMAO) and atherosclerosis development. However, the exact mechanism underlying this correlation is unknown. In the present study, we try to explore the impact of TMAO on endothelial dysfunction. After TMAO treatment, human umbilical vein endothelial cells (HUVECs) showed significant impairment in cellular proliferation and HUVECs-extracellular matrix (ECM) adhesion compared with control. Likewise, TMAO markedly suppressed HUVECs migration in transwell migration assay and wound healing assay. In addition, we found TMAO up-regulated vascular cell adhesion molecule-1 (VCAM-1) expression, promoted monocyte adherence, activated protein kinase C (PKC) and p-NF-κB. Interestingly, TMAO-stimulated VCAM-1 expression and monocyte adherence were diminished by PKC inhibitor. These results demonstrate that TMAO promotes early pathological process of atherosclerosis by accelerating endothelial dysfunction, including decreasing endothelial self-repair and increasing monocyte adhesion. Furthermore, TMAO-induced monocyte adhesion is partly attributable to activation of PKC/NF-κB/VCAM-1.
Collapse
|
18
|
Zaromytidou M, Siasos G, Coskun AU, Lucier M, Antoniadis AP, Papafaklis MI, Koskinas KC, Andreou I, Feldman CL, Stone PH. Intravascular hemodynamics and coronary artery disease: New insights and clinical implications. Hellenic J Cardiol 2016; 57:389-400. [PMID: 27894949 DOI: 10.1016/j.hjc.2016.11.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 07/26/2016] [Indexed: 11/23/2022] Open
Abstract
Intracoronary hemodynamics play a pivotal role in the initiation and progression of the atherosclerotic process. Low pro-inflammatory endothelial shear stress impacts vascular physiology and leads to the occurrence of coronary artery disease and its implications.
Collapse
Affiliation(s)
- Marina Zaromytidou
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Gerasimos Siasos
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Ahmet U Coskun
- Mechanical and Industrial Engineering, Northeastern University, Boston, MA, United States
| | - Michelle Lucier
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Antonios P Antoniadis
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Michail I Papafaklis
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Konstantinos C Koskinas
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Ioannis Andreou
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Charles L Feldman
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Peter H Stone
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
19
|
Andreou I, Antoniadis AP, Shishido K, Papafaklis MI, Koskinas KC, Chatzizisis YS, Coskun AU, Edelman ER, Feldman CL, Stone PH. How do we prevent the vulnerable atherosclerotic plaque from rupturing? Insights from in vivo assessments of plaque, vascular remodeling, and local endothelial shear stress. J Cardiovasc Pharmacol Ther 2015; 20:261-275. [PMID: 25336461 DOI: 10.1177/1074248414555005] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 08/14/2014] [Indexed: 01/13/2023]
Abstract
Coronary atherosclerosis progresses both as slow, gradual enlargement of focal plaque and also as a more dynamic process with periodic abrupt changes in plaque geometry, size, and morphology. Systemic vasculoprotective therapies such as statins, angiotensin-converting enzyme inhibitors, and antiplatelet agents are the cornerstone of prevention of plaque rupture and new adverse clinical outcomes, but such systemic therapies are insufficient to prevent the majority of new cardiac events. Invasive imaging methods have been able to identify both the anatomic features of high-risk plaque and the ongoing pathobiological stimuli responsible for progressive plaque inflammation and instability and may provide sufficient information to formulate preventive local mechanical strategies (eg, preemptive percutaneous coronary interventions) to avert cardiac events. Local endothelial shear stress (ESS) triggers vascular phenomena that synergistically exacerbate atherosclerosis toward an unstable phenotype. Specifically, low ESS augments lipid uptake and catabolism, induces plaque inflammation and oxidation, downregulates the production, upregulates the degradation of extracellular matrix, and increases cellular apoptosis ultimately leading to thin-cap fibroatheromas and/or endothelial erosions. Increases in blood thrombogenicity that result from either high or low ESS also contribute to plaque destabilization. An understanding of the actively evolving vascular phenomena, as well as the development of in vivo imaging methodologies to identify the presence and severity of the different processes, may enable early identification of a coronary plaque destined to acquire a high-risk state and allow for highly selective, focal preventive interventions to avert the adverse natural history of that particular plaque. In this review, we focus on the role of ESS in the pathobiologic processes responsible for plaque destabilization, leading either to accelerated plaque growth or to acute coronary events, and emphasize the potential to utilize in vivo risk stratification of individual coronary plaques to optimize prevention strategies to preclude new cardiac events.
Collapse
Affiliation(s)
- Ioannis Andreou
- The Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Antonios P Antoniadis
- The Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Koki Shishido
- The Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Michail I Papafaklis
- The Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Konstantinos C Koskinas
- The Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Yiannis S Chatzizisis
- The Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Ahmet U Coskun
- The Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Elazer R Edelman
- The Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Charles L Feldman
- The Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Peter H Stone
- The Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
20
|
Briançon-Marjollet A, Henri M, Pépin JL, Lemarié E, Lévy P, Tamisier R. Altered in vitro endothelial repair and monocyte migration in obstructive sleep apnea: implication of VEGF and CRP. Sleep 2014; 37:1825-32. [PMID: 25364078 DOI: 10.5665/sleep.4180] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
STUDY OBJECTIVES Although obstructive sleep apnea (OSA) causes cardiovascular morbidities through atherosclerosis induced by inflammation and endothelial dysfunction, OSA patients exhibit elevated plasma vascular endothelial growth factor (VEGF), which may represent an adaptive response to intermittent hypoxia. The aims of this study were to investigate whether in vitro endothelial wound healing and monocyte migration are affected by patient serum, and to determine the implication of circulating factors (VEGF and C-reactive protein). PATIENTS Serum was collected from healthy controls (HC), "healthy" OSA, and metabolic syndrome (MS) patients with or without OSA. MEASUREMENTS AND RESULTS Along with the presence of OSA and/or MS, both VEGF and hsCRP were significantly elevated in patient serum. Their specific role was tested with blocking antibodies on primary endothelial cells for wound healing assay and on human monocytes for migration assay. Endothelial wound healing was reduced with OSA compared to HC serum, and even more significantly using MS+OSA patient serum. Altered wound healing with OSA serum was unmasked when blocking VEGF and restored when blocking CRP. Monocyte migration was activated with OSA serum, and further enhanced by MS+OSA patient serum. Blocking CRP in serum inhibited this migration. CONCLUSIONS Serum from OSA patient alters in vitro endothelial cell repair function and activates monocyte migration; this is further aggravated with the presence of metabolic syndrome. These effects are partly driven by VEGF and CRP, suggesting an unfavorable balance between the pro healing (VEGF) and pro injury (CRP) factors that may promote vascular injury in OSA with and without metabolic syndrome.
Collapse
Affiliation(s)
- Anne Briançon-Marjollet
- Univ Grenoble Alpes, HP2, F-38000 Grenoble, France ; INSERM U1042, HP2, F-38000 Grenoble, France
| | - Marion Henri
- Univ Grenoble Alpes, HP2, F-38000 Grenoble, France ; INSERM U1042, HP2, F-38000 Grenoble, France
| | - Jean-Louis Pépin
- Univ Grenoble Alpes, HP2, F-38000 Grenoble, France ; INSERM U1042, HP2, F-38000 Grenoble, France ; CHU de Grenoble, HP2, F-38000 Grenoble, France
| | - Emeline Lemarié
- Univ Grenoble Alpes, HP2, F-38000 Grenoble, France ; INSERM U1042, HP2, F-38000 Grenoble, France
| | - Patrick Lévy
- Univ Grenoble Alpes, HP2, F-38000 Grenoble, France ; INSERM U1042, HP2, F-38000 Grenoble, France ; CHU de Grenoble, HP2, F-38000 Grenoble, France
| | - Renaud Tamisier
- Univ Grenoble Alpes, HP2, F-38000 Grenoble, France ; INSERM U1042, HP2, F-38000 Grenoble, France ; CHU de Grenoble, HP2, F-38000 Grenoble, France
| |
Collapse
|
21
|
Experimental tools to monitor the dynamics of endothelial barrier function: a survey of in vitro approaches. Cell Tissue Res 2014; 355:485-514. [DOI: 10.1007/s00441-014-1810-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 01/13/2014] [Indexed: 02/05/2023]
|
22
|
Abstract
It is well known that the altered blood flow is related to vascular diseases, including atherosclerosis, restenosis, and arteriosclerosis, which preferentially located at areas with the disturbed blood flow, suggesting that altered biomechanical stress may exert their effect on the vascular disease. Recent evidence indicated the presence of abundant stem/progenitor cells in the vessel wall, in which laminar shear stress can stimulate these cells to differentiate towards endothelial lineage, while cyclic strain results in smooth muscle differentiation. In line with this, it was evidenced that altered biomechanical stress in stented vessels may lead to 'wrong' direction of vascular stem cell differentiation resulting in restenosis. However, the underlying mechanisms are not well understood. In this article, we will give an overview of the effect of the local flow pattern on stem/progenitor cell differentiation and the possible mechanism on how the blood flow influences stem cell behaviours in the development of vascular diseases.
Collapse
Affiliation(s)
- Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Shandong University Qilu Hospital, Jinan, China
| | | | | | | |
Collapse
|
23
|
Pan B, Ma Y, Ren H, He Y, Wang Y, Lv X, Liu D, Ji L, Yu B, Wang Y, Chen YE, Pennathur S, Smith JD, Liu G, Zheng L. Diabetic HDL is dysfunctional in stimulating endothelial cell migration and proliferation due to down regulation of SR-BI expression. PLoS One 2012; 7:e48530. [PMID: 23133640 PMCID: PMC3487724 DOI: 10.1371/journal.pone.0048530] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 09/26/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Diabetic HDL had diminished capacity to stimulate endothelial cell (EC) proliferation, migration, and adhesion to extracellular matrix. The mechanism of such dysfunction is poorly understood and we therefore sought to determine the mechanistic features of diabetic HDL dysfunction. METHODOLOGY/PRINCIPAL FINDINGS We found that the dysfunction of diabetic HDL on human umbilical vein endothelial cells (HUVECs) was associated with the down regulation of the HDL receptor protein, SR-BI. Akt-phosphorylation in HUVECs was induced in a biphasic manner by normal HDL. While diabetic HDL induced Akt phosphorylation normally after 20 minutes, the phosphorylation observed 24 hours after diabetic HDL treatment was reduced. To determine the role of SR-BI down regulation on diminished EC responses of diabetic HDL, Mouse aortic endothelial cells (MAECs) were isolated from wild type and SR-BI (-/-) mice, and treated with normal and diabetic HDL. The proliferative and migratory effects of normal HDL on wild type MAECs were greatly diminished in SR-BI (-/-) cells. In contrast, response to diabetic HDL was impaired in both types suggesting diminished effectiveness of diabetic HDL on EC proliferation and migration might be due to the down regulation of SR-BI. Additionally, SR-BI down regulation diminishes diabetic HDL's capacity to activate Akt chronically. CONCLUSIONS/SIGNIFICANCE Diabetic HDL was dysfunctional in promoting EC proliferation, migration, and adhesion to matrix which was associated with the down-regulation of SR-BI. Additionally, SR-BI down regulation diminishes diabetic HDL's capacity to activate Akt chronically.
Collapse
Affiliation(s)
- Bing Pan
- The Institute of Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Health Science Center, Beijing, China
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Health Science Center, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Health Science Center, Beijing, China
| | - Yijing Ma
- The Institute of Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Health Science Center, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Health Science Center, Beijing, China
| | - Hui Ren
- The Institute of Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Health Science Center, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Health Science Center, Beijing, China
| | - Yubin He
- The Military General Hospital of Beijing, Beijing, China
| | - Yongyu Wang
- Department of Pathology, Shantou University Medical College, Shantou, Guangdong, China
| | - Xiaofeng Lv
- The Military General Hospital of Beijing, Beijing, China
| | - Donghui Liu
- The Institute of Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Health Science Center, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Health Science Center, Beijing, China
| | - Liang Ji
- The Institute of Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Health Science Center, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Health Science Center, Beijing, China
| | - Baoqi Yu
- The Institute of Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Health Science Center, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Health Science Center, Beijing, China
| | - Yuhui Wang
- The Institute of Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Health Science Center, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Health Science Center, Beijing, China
| | - Y. Eugene Chen
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Subramaniam Pennathur
- Department of Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Jonathan D. Smith
- Department of Cell Biology, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - George Liu
- The Institute of Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Health Science Center, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Health Science Center, Beijing, China
| | - Lemin Zheng
- The Institute of Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Health Science Center, Beijing, China
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Health Science Center, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Health Science Center, Beijing, China
| |
Collapse
|
24
|
High-fat diet increases and the polyphenol, S17834, decreases acetylation of the sirtuin-1-dependent lysine-382 on p53 and apoptotic signaling in atherosclerotic lesion-prone aortic endothelium of normal mice. J Cardiovasc Pharmacol 2012; 58:263-71. [PMID: 21654327 DOI: 10.1097/fjc.0b013e3182239eb7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Our purpose was to determine if high-fat diet and treatment with a polyphenol regulate the acetylation of lysine-382 of p53, the site regulated by sirtuin-1, and apoptosis in the endothelium of the atherosclerotic lesion-prone mouse aortic arch. In cultured endothelial cells, 2 atherogenic stimuli, hydrogen peroxide and tumor necrosis factor-α, increased the acetylation of p53 lysine-382, and caspase-3 cleavage, an indicator of apoptotic signaling. The polyphenol, S17834, significantly prevented these changes. In low-density lipoprotein receptor-deficient mice, a high-fat diet increased, and treatment with S17834 attenuated early atherosclerotic lesions on the lesser curvature of the aortic arch. In wild-type C57BL6 mice fed the same diet, no atherosclerotic lesions were observed in this lesion-prone area, but p53 acetylation and caspase-3 cleavage increased in the endothelium. In high-fat fed mice, S17834 increased sirtuin-1 protein in the lesion-prone endothelium and prevented both the increase in p53 acetylation and caspase-3 cleavage without affecting blood lipids. These results indicate that high-fat diet increases and S17834 decreases the acetylation of p53 in lesion-prone aortic endothelial cells of normal mice independently of blood lipids, suggesting that the polyphenol may regulate endothelial cell p53 acetylation and apoptosis via local actions.
Collapse
|
25
|
Li R, Mittelstein D, Fang K, Beebe T, Quigley K, Berliner J, Hsiai TK. Angiopoeitin-2 modulates Survivin expression in OxLDL-induced endothelial cell apoptosis. Biochem Biophys Res Commun 2011; 417:619-22. [PMID: 22182412 DOI: 10.1016/j.bbrc.2011.12.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Accepted: 12/05/2011] [Indexed: 01/11/2023]
Abstract
Angiopoeitin-2 (Ang-2) antagonizes Angiopeitin-1 (Ang-1)-mediated Tie-2 signaling. Ang-1 is reported to up-regulate anti-apoptotic Survivin expression. Here, we investigated the interplay between Ang-2 and Survivin in response to oxidized low density lipoprotein (OxLDL)-induced apoptosis. We demonstrate that treatment of human aortic endothelial cells (HAEC) with 100 μg/ml of OxLDL down-regulated Ang-2 expression as early as 4h after treatment and persisted up to 24h (p<0.05, n=3), but did not down-regulate Survivin until the 24h point. Further, treatment of HAEC with recombinant Ang-2 up-regulated Survivin expression (at Ang-2 ≥200 ng/ml, p<0.05, n=3) and attenuated the OxLDL-mediated down-regulation of Survivin (p<0.05, n=3). Knockdown of Ang-2 further down-regulated Survivin expression, whereas over-expression of Survivin attenuated OxLDL-induced HAEC apoptosis (p<0.05, n=3). Hence, Ang-2 mediated Survivin expression in response to OxLDL-induced endothelial apoptosis.
Collapse
Affiliation(s)
- Rongsong Li
- Biomedical Engineering and Cardiovascular Medicine, University of Southern California, Los Angeles, CA 90089, United States
| | | | | | | | | | | | | |
Collapse
|
26
|
Xu Q, Metzler B, Jahangiri M, Mandal K. Molecular chaperones and heat shock proteins in atherosclerosis. Am J Physiol Heart Circ Physiol 2011; 302:H506-14. [PMID: 22058161 DOI: 10.1152/ajpheart.00646.2011] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In response to stress stimuli, mammalian cells activate an ancient signaling pathway leading to the transient expression of heat shock proteins (HSPs). HSPs are a family of proteins serving as molecular chaperones that prevent the formation of nonspecific protein aggregates and assist proteins in the acquisition of their native structures. Physiologically, HSPs play a protective role in the homeostasis of the vessel wall but have an impact on immunoinflammatory processes in pathological conditions involved in the development of atherosclerosis. For instance, some members of HSPs have been shown to have immunoregulatory properties and modification of innate and adaptive response to HSPs, and can protect the vessel wall from the disease. On the other hand, a high degree of sequence homology between microbial and mammalian HSPs, due to evolutionary conservation, carries a risk of misdirected autoimmunity against HSPs expressed on the stressed cells of vascular endothelium. Furthermore, HSPs and anti-HSP antibodies have been shown to elicit production of proinflammatory cytokines. Potential therapeutic use of HSP in prevention of atherosclerosis involves achieving optimal balance between protective and immunogenic effects of HSPs and in the progress of research on vaccination. In this review, we update the progress of studies on HSPs and the integrity of the vessel wall, discuss the mechanism by which HSPs exert their role in the disease development, and highlight the potential clinic translation in the research field.
Collapse
Affiliation(s)
- Qingbo Xu
- Cardiovascular Division, King's British Heart Foundation Center, King's College London, London, United Kingdom
| | | | | | | |
Collapse
|
27
|
Rippe C, Blimline M, Magerko KA, Lawson BR, LaRocca TJ, Donato AJ, Seals DR. MicroRNA changes in human arterial endothelial cells with senescence: relation to apoptosis, eNOS and inflammation. Exp Gerontol 2011; 47:45-51. [PMID: 22037549 DOI: 10.1016/j.exger.2011.10.004] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 10/05/2011] [Accepted: 10/07/2011] [Indexed: 01/21/2023]
Abstract
A senescent phenotype in endothelial cells is associated with increased apoptosis, reduced endothelial nitric oxide synthase (eNOS) and inflammation, which are implicated in arterial dysfunction and disease in humans. We tested the hypothesis that changes in microRNAs are associated with a senescent phenotype in human aortic endothelial cells (HAEC). Compared with early-passage HAEC, late-passage HAEC had a reduced proliferation rate and increased staining for senescence-associated beta-galactosidase and the tumor suppressor p16(INK4a). Late-passage senescent HAEC had reduced expression of proliferation-stimulating/apoptosis-suppressing miR-21, miR-214 and miR-92 and increased expression of tumor suppressors and apoptotic markers. eNOS-suppressing miR-221 and miR-222 were increased and eNOS protein and eNOS activation (phosphorylation at serine1177) were lower in senescent HAEC. Caveolin-1 inhibiting miR-133a was reduced and caveolin-1, a negative regulator of eNOS activity, was elevated in senescent HAEC. Inflammation-repressing miR-126 was reduced and inflammation-stimulating miR-125b was increased, whereas inflammatory proteins were greater in senescent HAEC. Development of a senescent arterial endothelial cell phenotype featuring reduced cell proliferation, enhanced apoptosis and inflammation and reduced eNOS is associated with changes in miRNAs linked to the regulation of these processes. Our results support the hypothesis that miRNAs could play a critical role in arterial endothelial cell senescence.
Collapse
Affiliation(s)
- Catarina Rippe
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, CO 80309, United States.
| | | | | | | | | | | | | |
Collapse
|
28
|
Zhu F, Wang Q, Guo C, Wang X, Cao X, Shi Y, Gao F, Ma C, Zhang L. IL-17 induces apoptosis of vascular endothelial cells: a potential mechanism for human acute coronary syndrome. Clin Immunol 2011; 141:152-60. [PMID: 21872532 DOI: 10.1016/j.clim.2011.07.003] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 07/14/2011] [Accepted: 07/17/2011] [Indexed: 11/17/2022]
Abstract
Th17 cells producing IL-17 are involved in the pathogenesis of atherosclerosis, but the underlying mechanisms remain unclear. In this study, we investigated the effects of IL-17 on human vascular endothelial cells and showed that IL-17 induced cell death of the vascular endothelial cells, which played a pivotal role in plaque destabilization triggering acute coronary syndrome (ACS). We showed that circulating Th17 cells and IL-17 increased in patients with ACS compared to the patients with stable angina or health individuals; the plasma levels of IL-6 increased but TGF-β decreased in ACS patients, exhibiting a positive and negative correlation with that of IL-17, respectively. Importantly, we uncovered that IL-17 promoted the production of von Willebrand factor by endothelial cells and induced endothelial apoptosis by activating caspase-3, caspase-9 and up-regulating the ratio of Bax/Bcl-2, indicating the function of IL-17 in vascular endothelial damage as a potential mechanism for the pathogenesis of human ACS.
Collapse
Affiliation(s)
- Faliang Zhu
- Department of Immunology, Shandong University School of Medicine, Jinan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Masi LN, Portioli-Sanches EP, Lima-Salgado TM, Curi R. Toxicity of fatty acids on ECV-304 endothelial cells. Toxicol In Vitro 2011; 25:2140-6. [PMID: 21723937 DOI: 10.1016/j.tiv.2011.06.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 06/13/2011] [Accepted: 06/16/2011] [Indexed: 01/26/2023]
Abstract
The effects of stearic (saturated) or oleic (monounsaturated) acids and their combination with ω-3 and ω-6 polyunsaturated fatty acids (PUFA) on death of endothelial cells (ECV-304 cell line) were investigated. We examined: loss of plasma membrane integrity, DNA fragmentation, accumulation of neutral lipids (NL) and release of reactive oxygen species (ROS). The fatty acids studied were: stearic (SA), oleic (OA), docosahexaenoic (DHA), eicosapentaenoic (EPA), linoleic (LA) and gamma-linolenic (γA) acids. SA at 150 μM induced cell death, did not lead to accumulation of NL and raised the release of ROS. ω-3 PUFA decreased ROS production, increased NL content but did not protect against ECV-304 cell death induced by SA. ω-6 PUFA inhibited SA-induced cell death, increased NL content and decreased ROS production. OA caused cell death but did not increase NL content and ROS production even at 300 μM. ω-3 and ω-6 FA associated with OA further increased cell death with no change in ROS production and NL content. Concluding, ω-6 PUFA had a greater protective effect than ω-3 PUFA on the deleterious effects caused by SA whereas OA had low cytotoxicity but, when associated with PUFA, presented marked toxic effects on ECV-304 endothelial cells.
Collapse
Affiliation(s)
- Laureane Nunes Masi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, Av. Prof. Lineu Prestes, 1524, 05508-900 São Paulo, Brazil.
| | | | | | | |
Collapse
|
30
|
MicroRNA-21 targets peroxisome proliferators-activated receptor-alpha in an autoregulatory loop to modulate flow-induced endothelial inflammation. Proc Natl Acad Sci U S A 2011; 108:10355-60. [PMID: 21636785 DOI: 10.1073/pnas.1107052108] [Citation(s) in RCA: 275] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Adhesion of circulating monocytes to vascular endothelial cells (ECs) is a critical event leading to vascular inflammation and, hence, development of atherosclerosis. MicroRNAs (miRs) are a class of endogenous, highly conserved, noncoding small RNAs that play important roles in regulating gene expression and cellular function, as well as pathogenesis of atherosclerosis. Here, we showed that oscillatory shear stress (OSS) induces the expression of miR-21 at the transcriptional level in cultured human umbilical vein ECs via an increased binding of c-Jun, which is a component of transcription factor activator protein-1 (AP-1), to the promoter region of miR-21. OSS induction of miR-21 inhibited the translation, but not transcription, of peroxisome proliferators-activated receptor-α (PPARα) by 3'-UTR targeting. Overexpression of miR-21 up-regulated AP-1 activation, which was attenuated by exogenous expression of PPARα. OSS and overexpression of miR-21 enhanced the expression of adhesion molecules vascular cell adhesion molecule-1 and monocyte chemotactic protein-1 and the consequential adhesion of monocytes to ECs. Overexpression of PPARα significantly attenuated the AP-1-mediated miR-21 expression. These results demonstrate a unique mechanism by which OSS induces AP-1-dependent miR-21 expression, which directly targets PPARα to inhibit its expression, thereby allowing activation of AP-1 and the promotion of monocyte adhesion. Our findings suggest the presence of a positive feedback loop that enables the sustained induction of miR-21, thus contributing to the proinflammatory responses of vascular endothelium under OSS.
Collapse
|
31
|
Kliche K, Jeggle P, Pavenstädt H, Oberleithner H. Role of cellular mechanics in the function and life span of vascular endothelium. Pflugers Arch 2011; 462:209-17. [DOI: 10.1007/s00424-011-0929-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 01/13/2011] [Accepted: 01/16/2011] [Indexed: 01/17/2023]
|