1
|
Wang Y, Zhang X, Li X, Cheng M, Cui X. The vascular microenvironment and its stem cells regulate vascular homeostasis. Front Cell Dev Biol 2025; 13:1544129. [PMID: 40114970 PMCID: PMC11922910 DOI: 10.3389/fcell.2025.1544129] [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: 12/12/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
The vascular microenvironment comprises of anatomical structures, extracellular matrix components, and various cell populations, which play a crucial role in regulating vascular homeostasis and influencing vascular structure and function. Under physiological conditions, intrinsic regulation of the vascular microenvironment is required to sustain vascular homeostasis. In contrast, under pathological conditions, alterations to this microenvironment lead to vascular injury and pathological remodeling. According to the anatomy, the vascular microenvironment can be subdivided into three sections from the inside out. The vascular endothelial microenvironment, centered on vascular endothelial cells (VECs), includes the extracellular matrix and various vascular physicochemical factors. The VECs interact with vascular physicochemical factors to regulate the function of various parenchymal cells, including hepatocytes, neurons and tumor cells. The vascular wall microenvironment, comprising the vasa vasorum and their unique stem/progenitor cell niches, plays a pivotal role in vascular inflammation and pathological remodeling. Additionally, the perivascular microenvironment, which includes perivascular adipose tissue, consists of adipocytes and stem cells, which contribute to the pathological processes of atherosclerosis. It is anticipated that targeted regulation of the vascular microenvironment will emerge as a novel approach for the treatment of various diseases. Accordingly, this review will examine the structure of the vascular microenvironment, the regulation of vascular function by vascular cells and stem/progenitor cells, and the role of the vascular microenvironment in regulating cardiovascular diseases.
Collapse
Affiliation(s)
- Yanhui Wang
- Medical Physiology Laboratory, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Xiaoyun Zhang
- Medical Physiology Laboratory, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Xin Li
- Medical Physiology Laboratory, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Min Cheng
- Medical Physiology Laboratory, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Xiaodong Cui
- Medical Physiology Laboratory, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| |
Collapse
|
2
|
Ketata I, Ellouz E. Multiple sclerosis and vascular nexus: A systematic review and meta-analysis of incidence and mortality. Rev Neurol (Paris) 2025; 181:184-199. [PMID: 39627054 DOI: 10.1016/j.neurol.2024.10.007] [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: 07/02/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 03/21/2025]
Abstract
BACKGROUND Vascular diseases (VDs) are increasingly studied in multiple sclerosis (MS) due to their significant impact on disability and mortality. Our aim was to evaluate VD incidence rates and determine the prevalence of VD-related mortality. MATERIAL AND METHODS Our study followed the PRISMA 2020 guidelines. We searched PubMed, Google Scholar, Europe PMC, and Web of Science from the inception up to April 30th, 2024. Data analysis was performed using the R statistics package program. Sensitivity analysis, subgroup analysis, and meta-regression were conducted for significant heterogeneity. RESULTS We collected 23 studies. Patients with MS exhibited significantly higher rates of VD compared with controls. Patients with MS showed a significantly higher rate of heart failure (HF), venous thromboembolism (VTE), peripheral vascular disease (PVD), and stroke, although the incidence rate of atrial fibrillation/flutter was significantly lower compared with controls. High blood pressure tended to be lower compared with controls. The highest rate of VD was within the first 10years of follow-up, significantly for myocardial infraction, VTE, and stroke, compared with controls. Regarding mortality, the prevalence of VD-related mortality among deceased individuals with MS was 16.7% [95% CI 11.8;22.0]. CONCLUSION Our meta-analysis revealed a significant dynamic nexus between MS and VD. MS may act as an independent risk factor for certain VDs, yet it could also be a protective factor against others. Physicians should consider VD and mortality related to VD when managing patients with MS.
Collapse
Affiliation(s)
- I Ketata
- Neurology Department, University Hospital of Gabes, 6014 Gabes, Tunisia; Sfax Faculty of Medicine, Sfax University, Sfax, Tunisia.
| | - E Ellouz
- Neurology Department, University Hospital of Gabes, 6014 Gabes, Tunisia; Sfax Faculty of Medicine, Sfax University, Sfax, Tunisia
| |
Collapse
|
3
|
Huang H, Shen Y. Bezafibrate mitigates oxidized-low density lipoprotein (ox-LDL)-induced the attachment of monocytes to endothelial cells: An implication in atherosclerosis. Fundam Clin Pharmacol 2024; 38:958-966. [PMID: 39009501 DOI: 10.1111/fcp.13025] [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: 02/27/2024] [Revised: 05/18/2024] [Accepted: 06/11/2024] [Indexed: 07/17/2024]
Abstract
BACKGROUND Oxidized forms of low-density lipoproteins (ox-LDL)-associated endothelial dysfunction and subsequent monocyte adhesion play an important role in the development of atherosclerosis (AS). Bezafibrate (BEZ) is a peroxisome proliferator-activated receptor (pan-PPAR) agonist licensed as a hypolipidemic drug. However, the effects of BEZ on endothelial dysfunction are less reported. OBJECTIVES In this study, we aim to investigate the protective effects of BEZ on ox-LDL-challenged vascular endothelial cells to evaluate its potential value in treating AS. METHODS Human aortic endothelial cells (HAECs) and THP-1 cells were used to establish an In Vitro AS model. Cell Counting Kit-8 (CCK-8) assay, Real-time PCR, Western blot analysis, and Enzyme-linked immunosorbent assay (ELISA) were used to test the data. RESULTS As expected, treatment with BEZ suppressed the expression of vascular endothelial growth factor A (VEGF-A), tissue factor (TF), Interleukin 12 (IL-12), tumor necrosis factor (TNF-α), and monocyte chemoattractant protein-1 (MCP-1). BEZ was also found to inhibit ox-LDL-induced expression of the endothelial adhesion molecules vascular cellular adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1) in HAECs. Correspondingly, BEZ prevented attachment of THP-1 monocytes to ox-LDL-incubated HAECs. Mechanically, BEZ was found to prevent NF-κB activation by reducing the levels of nuclear NF-κB p65 and inhibiting luciferase activity of NF-κB. CONCLUSION Our study revealed the pharmacological function of BEZ in protecting endothelial dysfunction against ox-LDL, which may provide valuable insight for the clinical application of BEZ.
Collapse
Affiliation(s)
- Huijun Huang
- Department of Cardiology, Shanghai Eighth People's Hospital, Shanghai, China
| | - Yan Shen
- Department of Cardiology, Shanghai Eighth People's Hospital, Shanghai, China
| |
Collapse
|
4
|
Liu X, Huang R, Wan J, Niu T. LncRNA MIR4697HG Alleviates Endothelial Cell Injury and Atherosclerosis Progression in Mice via the FUS/ANXA5 Axis. Biochem Genet 2024; 62:3155-3173. [PMID: 38082058 DOI: 10.1007/s10528-023-10542-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 10/02/2023] [Indexed: 07/31/2024]
Abstract
Atherosclerosis (AS) manifests with arterial intimal injury, lipid deposition and chronic inflammation, which is a key pathogenic cause of cardio-cerebrovascular disorders. LncRNA MIR4697HG was downregulated in human advanced atherosclerotic plaques. This study probed the precise biological functions and downstream regulatory mechanisms of MIR4697HG during AS progression. MIR4697HG levels in atherosclerotic plaque tissues and normal arterial intima were measured by RT-qPCR. An injury model of human umbilical vein endothelial cells (HUVECs) was induced through treating with oxidative low-density lipoprotein (ox-LDL). MIR4697HG overexpression plasmids (pcDNA-MIR4697HG) was transfected into ox-LDL-treated HUVECs, and then cell viability, apoptosis, reactive oxygen species (ROS) level, oxidative stress marker protein malondialdehyde (MDA) level and superoxide dismutase (SOD) activity, and adhesion molecule intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) levels in HUVECs were determined. Moreover, the binding between MIR4697HG and fused in sarcoma (FUS) was checked with RNA pull-down assay. The interaction between FUS and annexin A5 (ANXA5) was gauged with Co-immunoprecipitation. Then MIR4697HG/FUS/ANXA5 axis mediated HUVEC functions were accessed with rescue experiments. Additionally, an AS model was established via feeding a high-fat diet for ApoE-/- mice, and lentivirus MIR4697HG overexpression vector (Lv-MIR4697HG) was injected into AS mice followed by detection of atherosclerotic plaque area in mice. MIR4697HG was downregulated in atherosclerotic plaque tissues and HUVECs stimulated by ox-LDL. MIR4697HG overexpression attenuated ox-LDL-induced HUVEC viability inhibition, apoptosis, oxidative stress and adhesion molecule release. Moreover, MIR4697HG bound with FUS and facilitated FUS expression in HUVECs. FUS knockdown abrogated the functions of lncRNA MIR4697HG overexpression in ox-LDL induced HUVEC injury. Besides, FUS could bind with ANXA5. FUS overexpression inhibited ox-LDL induced HUVEC injury, while ANXA5 knockdown reversed these effects. Additionally, Lv-MIR4697HG reduced atherosclerotic plaque area in ApoE-/- mice. LncRNA MIR4697HG mitigated ox-LDL-induced apoptosis, oxidative stress and adhesion molecule release in HUVECs and alleviated AS progression in mice through the FUS/ANXA5 axis.
Collapse
Affiliation(s)
- Xue Liu
- Department of Cardiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, He-Ping District, Shenyang, 110004, Liaoning, China
| | - Rui Huang
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Jiye Wan
- Department of Cardiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, He-Ping District, Shenyang, 110004, Liaoning, China
| | - Tiesheng Niu
- Department of Cardiology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, He-Ping District, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
5
|
Gao F, Zhao Y, Zhang B, Xiao C, Sun Z, Gao Y, Dou X. Orientin alleviates ox-LDL-induced oxidative stress, inflammation and apoptosis in human vascular endothelial cells by regulating Sestrin 1 (SESN1)-mediated autophagy. J Mol Histol 2024; 55:109-120. [PMID: 38165567 DOI: 10.1007/s10735-023-10176-z] [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: 07/21/2023] [Accepted: 11/04/2023] [Indexed: 01/04/2024]
Abstract
Endothelial cells are a crucial component of the vessel-tissue wall and exert an important role in atherosclerosis (AS). To explore the role of Orientin in AS, human vascular endothelial cells (HUVECs) were induced by oxidized low-density lipoprotein (ox-LDL) to simulate the vascular endothelial injury during AS. Cell viability was detected by CCK-8 assay. Oxidative stress and inflammation related markers were measured using kits, RT-qPCR or western blot. Besides, cell apoptosis was assessed with TUNEL staining and cell autophagy was evaluated by LC3 immunofluorescent staining. Additionally, western blot was utilized to evaluate the expression of Sestrin 1 (SESN1) and proteins in AMPK/mTOR signaling. Afterwards, SESN1 was silenced to determine the expression of autophagy-related proteins. The further application of autophagy inhibitor 3-methyladenine (3-MA) was used to clarify the regulatory mechanism of Orientin on autophagy. Results showed that the decreased viability of HUVECs caused by ox-LDL induction was elevated by Orientin. Oxidative stress and inflammation were also attenuated after Orientin addition in HUVECs under ox-LDL condition. Moreover, Orientin suppressed apoptosis and induced autophagy of HUVECs stimulated by ox-LDL, accompanied by enhanced level of phospho (p)-AMPK and declined level of p-mTOR. Interestingly, SESN1 level was elevated by Orientin, and SESN1 depletion alleviated autophagy and reduced p-AMPK expression but enhanced p-mTOR expression. The further experiments indicated that SESN1 silencing or 3-MA addition reversed the inhibitory effects of Orientin on the oxidative stress, inflammation and apoptosis of HUVECs. Collectively, Orientin could induce autophagy by activating SESN1 expression, thereby regulating AMPK/mTOR signaling in ox-LDL-induced HUVECs.
Collapse
Affiliation(s)
- Feng Gao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China.
| | - Yongcheng Zhao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| | - Bin Zhang
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| | - Chunwei Xiao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| | - Zhanfa Sun
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| | - Yuan Gao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| | - Xueyong Dou
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| |
Collapse
|
6
|
Gu M, Liu K, Xiong H, You Q. MiR-130a-3p inhibits endothelial inflammation by regulating the expression of MAPK8 in endothelial cells. Heliyon 2024; 10:e24541. [PMID: 38298633 PMCID: PMC10828701 DOI: 10.1016/j.heliyon.2024.e24541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 12/07/2023] [Accepted: 01/10/2024] [Indexed: 02/02/2024] Open
Abstract
MicroRNA-130a-3p (miR-130a-3p) has been reported as closely related to atherosclerosis (AS). This study is to survey the effects of miR-130a-3p in endothelial cells (ECs) treated with oxidized low-density lipoprotein (ox-LDL) and explore underlying mechanisms. The proliferation and apoptosis of ox-LDL-treated HUVEC cells were determined by CCK-8, EdU, and flow cytometry assays. ELISA and Western blot analysis measured the expressions of cytokines and protein levels. Bioinformatics and dual-luciferase reporter assay were performed to predict and confirm that Mitogen-activated protein kinase 8 (MAPK8) was a direct target of miR-130a-3p, and MAPK8 was negatively associated with miR-130a-3p. As expected, miR-130a-3p was down-regulated in ox-LDL-treated HUVEC cells, and up-regulation of miR-130a-3p promoted proliferation and inhibited apoptosis of ox-LDL-treated HUVEC cells. Furthermore, miR-130a-3p mimics suppressed the expressions of TNF-α and IL-6 and decreased the protein levels of VCAM-1, ICAM-1 and E-selectin. MAPK8 was highly expressed in ox-LDL-treated HUVEC cells, and silence of MAPK8 promoted proliferation inhibited apoptosis, suppressed inflammatory responses, and decreased the levels of VCAM-1, ICAM-1, and E-selectin, over-expression of MAPK8 partially restored the functional effects of miR-130a-3p on proliferation, inflammatory responses, and the expressions of VCAM-1, ICAM-1 and E-selectin. This study indicates that miR-130a-3p may emerge as an effective target for treating AS.
Collapse
Affiliation(s)
- Mingming Gu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Kun Liu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Hui Xiong
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Qingsheng You
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China
| |
Collapse
|
7
|
Tang F, Liu D, Zhang L, Xu LY, Zhang JN, Zhao XL, Ao H, Peng C. Targeting endothelial cells with golden spice curcumin: A promising therapy for cardiometabolic multimorbidity. Pharmacol Res 2023; 197:106953. [PMID: 37804925 DOI: 10.1016/j.phrs.2023.106953] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 09/20/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Cardiometabolic multimorbidity (CMM) is an increasingly significant global public health concern. It encompasses the coexistence of multiple cardiometabolic diseases, including hypertension, stroke, heart disease, atherosclerosis, and T2DM. A crucial component to the development of CMM is the disruption of endothelial homeostasis. Therefore, therapies targeting endothelial cells through multi-targeted and multi-pathway approaches hold promise for preventing and treatment of CMM. Curcumin, a widely used dietary supplement derived from the golden spice Carcuma longa, has demonstrated remarkable potential in treatment of CMM through its interaction with endothelial cells. Numerous studies have identified various molecular targets of curcumin (such as NF-κB/PI3K/AKT, MAPK/NF-κB/IL-1β, HO-1, NOs, VEGF, ICAM-1 and ROS). These findings highlight the efficacy of curcumin as a therapeutic agent against CMM through the regulation of endothelial function. It is worth noting that there is a close relationship between the progression of CMM and endothelial damage, characterized by oxidative stress, inflammation, abnormal NO bioavailability and cell adhesion. This paper provides a comprehensive review of curcumin, including its availability, pharmacokinetics, pharmaceutics, and therapeutic application in treatment of CMM, as well as the challenges and future prospects for its clinical translation. In summary, curcumin shows promise as a potential treatment option for CMM, particularly due to its ability to target endothelial cells. It represents a novel and natural lead compound that may offer significant therapeutic benefits in the management of CMM.
Collapse
Affiliation(s)
- Fei Tang
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Dong Liu
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Li Zhang
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Li-Yue Xu
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Jing-Nan Zhang
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Xiao-Lan Zhao
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Hui Ao
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Cheng Peng
- Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
8
|
Yang X, Wang C, Zhu G, Guo Z, Fan L. METTL14/YTHDF1 axis-modified UCHL5 aggravates atherosclerosis by activating the NLRP3 inflammasome. Exp Cell Res 2023; 427:113587. [PMID: 37044315 DOI: 10.1016/j.yexcr.2023.113587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/28/2023] [Accepted: 04/04/2023] [Indexed: 04/14/2023]
Abstract
BACKGROUND Vascular smooth muscle cell (VSMC) phenotypic switching contributes to VSMC proliferation and migration in atherosclerosis (AS). Nevertheless, the regulatory mechanism of VSMC phenotypic switching during AS progression is unclear. Here, the role and regulatory mechanism of UCHL5 in VSMC phenotypic switching during AS progression were investigated. METHODS ApoE-/- mice were fed with high fat diet to establish AS model in vivo. VSMCs stimulated by ox-LDL were used as AS cellular model. VSMC proliferation and migration were examined by CCK8 assay and transwell assay, respectively. The levels of pro-inflammatory cytokines were assessed using ELISA. The interactions between METTL14/YTHDF1, UCHL5 and NLRP3 were analyzed using RIP and/or dual-luciferase reporter gene and/or Co-IP assays. NLRP3 ubiquitination was analyzed by ubiquitination analysis. RESULTS UCHL5 was significantly upregulated in AS patients and ox-LDL-treated VSMCs. UCHL5 silencing ameliorated plaque formation and vascular remodeling in vivo and suppressed ox-LDL-induced VSMC proliferation, migration, inflammation and phenotypic switching in vitro. Moreover, METTL14 could increase UCHL5 mRNA m6A level and promoted UCHL5 expression by recruiting YTHDF1. Moreover, UCHL5 overexpression enhanced protein stability by deubiquitinating NLRP3. Rescue studies revealed that NLRP3 overexpression abrogated UCHL5 silencing-mediated biological effects in ox-LDL-treated VSMCs. CONCLUSION UCHL5 modified by METTL14/YTHDF1 axis could facilitate the inflammation and vascular remodeling in atherosclerosis by activating the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Xiaohu Yang
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 201700, China
| | - Chen Wang
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 201700, China
| | - Guanglang Zhu
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 201700, China
| | - Zhenyu Guo
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 201700, China
| | - Longhua Fan
- Department of Vascular Surgery, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 201700, China.
| |
Collapse
|
9
|
Zhi W, Liu Y, Wang X, Zhang H. Recent advances of traditional Chinese medicine for the prevention and treatment of atherosclerosis. JOURNAL OF ETHNOPHARMACOLOGY 2023; 301:115749. [PMID: 36181983 DOI: 10.1016/j.jep.2022.115749] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 09/13/2022] [Accepted: 09/19/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Atherosclerosis (AS) is a common systemic disease with increasing morbidity and mortality worldwide. Traditional Chinese medicine (TCM) with characteristics of multiple pathways and targets, presents advantages in the diagnosis and treatment of atherosclerosis. AIM OF THE STUDY With the modernization of TCM, the active ingredients and molecular mechanisms of TCM for AS treatment have been gradually revealed. Therefore, it is necessary to examine the existing studies on TCM therapies aimed at regulating AS over the past two decades. MATERIALS AND METHODS Using "atherosclerosis" and "Traditional Chinese medicine" as keywords, all relevant TCM literature published in the last 10 years was collected from electronic databases (such as Elsevier, Springer, PubMed, CNKI, and Web of Science), books and papers until March 2022, and the critical information was statistically analyzed. RESULTS In this review, we highlighted extracts of 8 single herbs, a total of 41 single active ingredients, 20 TCM formulae, and 25 patented drugs, which were described with chemical structure, source, model, efficacy and potential mechanism. CONCLUSION We summarized the cytopathological basis for the development of atherosclerosis involving vascular endothelial cells, macrophages and vascular smooth muscle cells, and categorically elaborated the medicinal TCM used for AS, all of which provide the current evidence on the better management of atherosclerosis by TCM.
Collapse
Affiliation(s)
- Wenbing Zhi
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Traditional Chinese Medicine Hospital), Xi'an, 710003, PR China.
| | - Yang Liu
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Traditional Chinese Medicine Hospital), Xi'an, 710003, PR China
| | - Xiumei Wang
- The Second Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi, China.
| | - Hong Zhang
- Shaanxi Academy of Traditional Chinese Medicine (Shaanxi Traditional Chinese Medicine Hospital), Xi'an, 710003, PR China.
| |
Collapse
|
10
|
Circ_0005699 participates in ox-LDL-induced human umbilical vein endothelial cell injury via targeting the miR-636/TLR4/NF-κB pathway. Biochem Eng J 2022. [DOI: 10.1016/j.bej.2022.108579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
11
|
Correlation between decreased plasma miR-29a and vascular endothelial injury induced by hyperlipidemia. Herz 2022:10.1007/s00059-022-05121-x. [PMID: 35674773 DOI: 10.1007/s00059-022-05121-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 02/14/2022] [Accepted: 04/22/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND Hyperlipidemia is a major risk factor for vascular endothelial injury and atherosclerosis leading to cardiovascular diseases. Early diagnosis of vascular endothelial injury is important for the prevention and prognosis of cardiovascular diseases. This study aimed to investigate sensitive circulating microRNA (miRNA) as a potential diagnostic biomarker of vascular endothelial injury in a hyperlipidemic rat model. METHODS The miRNA expression profile was detected by miRNA microarray. The hyperlipidemic rat model was established by intraperitoneal injection of vitamin D3 combined with a high-fat diet. Plasma miRNA levels were measured by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). RESULTS No significant difference was found in the types of highly expressed miRNAs between human umbilical artery endothelial cells (HUAEC) and human umbilical vein endothelial cells (HUVEC). A total of 10 highly expressed miRNAs in endothelial cells were selected as candidate miRNAs, including miR-21, miR-126, let-7a, miR-23a, miR-221, miR-125b, miR-26a, miR-29a, miR-16, and miR-100. The plasma levels of let-7a, miR-126, miR-21, and miR-26a were significantly elevated in hyperlipidemic rats at 30 and 50 days after modeling, while the plasma level of miR-29a was significantly decreased. No significant change was found in the plasma levels of miR-125b, miR-23a, miR-221, miR-100, and miR-16. Interestingly, a significant reduction in plasma miR-29 level was detected as early as 20 days after modeling, which was earlier than for soluble intercellular adhesion molecule‑1 (sICAM-1). CONCLUSION The plasma levels of endothelial cell-enriched miRNAs were correlated with vascular endothelial injury induced by hyperlipidemia. miR-29a might serve as a potential early diagnostic biomarker of endothelial injury-related diseases.
Collapse
|
12
|
Yuan Y, Zhang C, He Y, Yuan L, Zhao Q, Liu Y, Long S. Curcumin improves the function of umbilical vein endothelial cells by inhibiting H 2O 2‑induced pyroptosis. Mol Med Rep 2022; 25:214. [PMID: 35543146 PMCID: PMC9133960 DOI: 10.3892/mmr.2022.12730] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/19/2022] [Indexed: 11/05/2022] Open
Abstract
Endothelial cell (EC) dysfunction is one of the initiating factors of atherosclerosis. EC dysfunction is primarily caused by oxidative damage and inflammation. As a classic non-specific antioxidant and anti-inflammatory drug, curcumin has been widely used in studies of lipid metabolism disorders. However, whether curcumin is able to alleviate H2O2-induced EC damage and its related mechanisms has remained to be elucidated. The present study confirmed the protective effects of curcumin on human umbilical vein endothelial cells (HUVECs). A HUVEC injury model was established using H2O2 and the optimal concentrations and time of curcumin to achieve therapeutic effects were explored. Curcumin was observed to inhibit H2O2-induced pyroptosis by inhibiting the activation of NOD-, LRR- and pyrin domain-containing protein 3. In addition, curcumin improved HUVEC function by restoring αvβ3 and reducing endothelin-1 expression. In conclusion, the results of the present study revealed the mechanism through which curcumin inhibits pyroptosis and indicated that curcumin may have a potential utility in treating diseases of EC dysfunction.
Collapse
Affiliation(s)
- Yulin Yuan
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, Hengyang, Hunan 421001, P.R. China
| | - Caiping Zhang
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, Hengyang, Hunan 421001, P.R. China
| | - Yunwu He
- Department of Pain, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Lingzhi Yuan
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, Hengyang, Hunan 421001, P.R. China
| | - Qian Zhao
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, Hengyang, Hunan 421001, P.R. China
| | - Yuhe Liu
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, Hengyang, Hunan 421001, P.R. China
| | - Shiyin Long
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
13
|
Xu J, Zheng Y, Zhao Y, Zhang Y, Li H, Zhang A, Wang X, Wang W, Hou Y, Wang J. Succinate/IL-1β Signaling Axis Promotes the Inflammatory Progression of Endothelial and Exacerbates Atherosclerosis. Front Immunol 2022; 13:817572. [PMID: 35273600 PMCID: PMC8901997 DOI: 10.3389/fimmu.2022.817572] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/27/2022] [Indexed: 12/14/2022] Open
Abstract
Inflammation is an important driver of atherosclerosis. Succinate is a new extracellular inflammatory alarm released by activated macrophages. Succinate is sensed by succinate receptor 1 (Sucnr1) and then transferred to effector cells. It is worth exploring whether succinate is capable of facilitating the inflammatory response in atherosclerosis. In this study, we firstly found that arterial serum of Coronary Heart Disease (CHD) patients contained significantly higher succinate and interleukin (IL)-1β than Health control (HC) subjects, and succinate was positively correlated with IL-1β. As demonstrated by the in vitro study, succinate/hypoxia-inducible factor 1α (Hif)-1α/IL-1β signal axis existed and significantly facilitated the inflammatory program in human umbilical vein endothelial cells (HUVECs). Under the coculture, activated macrophages released succinate, which would be transferred to HUVECs via Sucnr1 and then activate Hif-1α to produce a greater amount of IL-1β. Likewise, the aortic sinus’s inflammatory phenotype was found to be more significant within Apoe-/- mice that were injected with succinate. Furthermore, Sucnr1 inhibitor (NF-56-EJ40) could significantly interrupt succinate/IL-1β signal in HUVECs and macrophages. As revealed by this study, glycolytic metabolism following the release of succinate could be found in atherosclerotic pathology, and succinate would drive succinate/IL-1β signal dependent on Sucnr1 and then exacerbate inflammatory responses. Sucnr1 might be a novel target for cutting off the transduction of succinate signal to prevent the inflammation of atherosclerosis.
Collapse
Affiliation(s)
- Jingwen Xu
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Yabing Zheng
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Yaqing Zhao
- College of Second Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yujiao Zhang
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Huilin Li
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - An Zhang
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xuehan Wang
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Weizong Wang
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Yinglong Hou
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Jiangrong Wang
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| |
Collapse
|
14
|
Kutikhin AG, Shishkova DK, Velikanova EA, Sinitsky MY, Sinitskaya AV, Markova VE. Endothelial Dysfunction in the Context of Blood–Brain Barrier Modeling. J EVOL BIOCHEM PHYS+ 2022; 58:781-806. [PMID: 35789679 PMCID: PMC9243926 DOI: 10.1134/s0022093022030139] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 01/04/2023]
Abstract
Here, we discuss pathophysiological approaches to the defining
of endothelial dysfunction criteria (i.e., endothelial activation,
impaired endothelial mechanotransduction, endothelial-to-mesenchymal
transition, reduced nitric oxide release, compromised endothelial
integrity, and loss of anti-thrombogenic properties) in different
in vitro and in vivo models. The canonical definition of endothelial
dysfunction includes insufficient production of vasodilators, pro-thrombotic
and pro-inflammatory activation of endothelial cells, and pathologically
increased endothelial permeability. Among the clinical consequences
of endothelial dysfunction are arterial hypertension, macro- and
microangiopathy, and microalbuminuria. We propose to extend the definition
of endothelial dysfunction by adding altered endothelial mechanotransduction
and endothelial-to-mesenchymal transition to its criteria. Albeit
interleukin-6, interleukin-8, and MCP-1/CCL2 dictate the pathogenic
paracrine effects of dysfunctional endothelial cells and are therefore
reliable endothelial dysfunction biomarkers in vitro, they are non-specific
for endothelial cells and cannot be used for the diagnostics of
endothelial dysfunction in vivo. Conceptual improvements in the
existing methods to model endothelial dysfunction, specifically,
in relation to the blood–brain barrier, include endothelial cell
culturing under pulsatile flow, collagen IV coating of flow chambers,
and endothelial lysate collection from the blood vessels of laboratory
animals in situ for the subsequent gene and protein expression profiling.
Combined with the simulation of paracrine effects by using conditioned
medium from dysfunctional endothelial cells, these flow-sensitive
models have a high physiological relevance, bringing the experimental
conditions to the physiological scenario.
Collapse
Affiliation(s)
- A. G. Kutikhin
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - D. K. Shishkova
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - E. A. Velikanova
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - M. Yu. Sinitsky
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - A. V. Sinitskaya
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| | - V. E. Markova
- Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russia
| |
Collapse
|
15
|
Reducing the Damage of Ox-LDL/LOX-1 Pathway to Vascular Endothelial Barrier Can Inhibit Atherosclerosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7541411. [PMID: 35391927 PMCID: PMC8983252 DOI: 10.1155/2022/7541411] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 03/11/2022] [Indexed: 11/17/2022]
Abstract
Aim The destruction of the vascular endothelial barrier mediated by Ox-LDL is the initial link to atherosclerosis. Here, we aimed to determine whether the immunological intervention with Ox-ApoB polypeptide fragment (Ox-ApoB-PF) can block the deposition of Ox-LDL in vascular endothelial cells through LOX-1 receptors, thereby protecting the barrier function and survival status of vascular endothelial cells and inhibiting the progression of atherosclerosis. Methods and Results In order to determine the harm of Ox-LDL to vascular endothelial cells and the protective effect of immune intervention with Ox-ApoB-PF, we conducted a series of corresponding experiments in vitro and in vivo. The in vitro results showed that Ox-LDL can activate endothelial cell apoptosis pathway; reduce the expression of endothelial junction proteins; affect the migration, deformation, and forming ability; and ultimately destroy the vascular endothelial barrier function. The increased permeability of endothelial cells led to a sharp increase in the phagocytosis of Ox-LDL by macrophages under the endothelial layer. Meanwhile, Ox-LDL stimulation induced a significant upregulation of LOX-1 in endothelial cells and increased the expression of endothelial cell chemokines and adhesion factors. Ox-ApoB-PF antibodies can significantly reduce the abovementioned harmful effects. The in vivo results showed that active immune intervention through Ox-ApoB-PF can protect the endothelial barrier function; reduce macrophage deposition and the inflammatory response in plaques; alleviate lipid deposition in the plaques, as well as apoptosis and necrosis; and increase the ability of liver macrophages to clear Ox-LDL. Eventually, the progression of plaque and the formation of necrotic cores in plaques can be inhibited. Conclusions An Ox-ApoB-PF antibody may protect the endothelial cell physiological function and survival status by blocking the combination of Ox-LDL/LOX-1 in vascular endothelial cells. Immune intervention with Ox-ApoB-PF inhibits the occurrence and development of atherosclerotic lesions by protecting the vascular endothelial barrier function.
Collapse
|
16
|
Gao F, Zhao Y, Zhang B, Xiao C, Sun Z, Gao Y, Dou X. SESN1 attenuates the Ox‑LDL‑induced inflammation, apoptosis and endothelial‑mesenchymal transition of human umbilical vein endothelial cells by regulating AMPK/SIRT1/LOX1 signaling. Mol Med Rep 2022; 25:161. [PMID: 35293601 PMCID: PMC8941522 DOI: 10.3892/mmr.2022.12678] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/10/2022] [Indexed: 11/06/2022] Open
Abstract
Endothelial cells are an important component of the heart and vasculature and form a crucial link between the cardiovascular system and the immune system. Sestrin 1 (SESN1) has an important role in atherosclerosis by inhibiting NOD-like receptor family pyrin domain containing 3 inflammasome activation. However, whether SESN1 is involved in human umbilical vein endothelial cell (HUVEC) injury caused by atherosclerosis has remained to be elucidated. The present study aimed to investigate the functions of SESN1 in the inflammatory response, apoptosis and endothelial-mesenchymal transition (EndMT) of HUVECs following stimulation with oxidized low-density lipoprotein (Ox-LDL). SESN1 expression at the mRNA and protein levels was detected using reverse transcription-quantitative PCR (RT-qPCR) and western blot analysis. Following SESN1 overexpression in Ox-LDL-stimulated HUVECs, cell viability was determined using a Cell Counting Kit-8 assay. Terminal deoxynucleotidyl transferase-mediated nick-end labeling staining was employed to detect cell apoptosis and western blot analysis was used to determine the levels of apoptosis-related proteins. RT-qPCR, ELISA and western blot were utilized to determine the levels of inflammatory factors. Immunofluorescence staining, RT-qPCR and western blot analysis were employed to assess the EndMT of Ox-LDL-stimulated HUVECs. The results revealed that SESN1 exhibited a low expression in HUVECs following Ox-LDL stimulation. SESN1 overexpression suppressed inflammation, apoptosis and EndMT in Ox-LDL-induced HUVECs. In addition, SESN1 stimulated adenosine monophosphate-activated protein kinase catalytic subunit α1/sirtuin 1 signaling to suppress Ox-LDL receptor-1 expression. An AMPK and SIRT1 inhibitor reversed the effects of SESN1 overexpression on the inflammatory response, apoptosis and EndMT of HUVECs exposed to Ox-LDL. Taken together, the present study demonstrated that SENS1 exerts a suppressive effect on Ox-LDL-induced inflammation, apoptosis and EndMT of HUVECs, suggesting that SENS1 may be used as a novel biomarker for endothelial injury-related disorders.
Collapse
Affiliation(s)
- Feng Gao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, Jiangsu 221005, P.R. China
| | - Yongcheng Zhao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, Jiangsu 221005, P.R. China
| | - Bin Zhang
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, Jiangsu 221005, P.R. China
| | - Chunwei Xiao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, Jiangsu 221005, P.R. China
| | - Zhanfa Sun
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, Jiangsu 221005, P.R. China
| | - Yuan Gao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, Jiangsu 221005, P.R. China
| | - Xueyong Dou
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, Xuzhou, Jiangsu 221005, P.R. China
| |
Collapse
|
17
|
Capsaicin and TRPV1 Channels in the Cardiovascular System: The Role of Inflammation. Cells 2021; 11:cells11010018. [PMID: 35011580 PMCID: PMC8750852 DOI: 10.3390/cells11010018] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 12/24/2022] Open
Abstract
Capsaicin is a potent agonist of the Transient Receptor Potential Vanilloid type 1 (TRPV1) channel and is a common component found in the fruits of the genus Capsicum plants, which have been known to humanity and consumed in food for approximately 7000-9000 years. The fruits of Capsicum plants, such as chili pepper, have been long recognized for their high nutritional value. Additionally, capsaicin itself has been proposed to exhibit vasodilatory, antimicrobial, anti-cancer, and antinociceptive properties. However, a growing body of evidence reveals a vasoconstrictory potential of capsaicin acting via the vascular TRPV1 channel and suggests that unnecessary high consumption of capsaicin may cause severe consequences, including vasospasm and myocardial infarction in people with underlying inflammatory conditions. This review focuses on vascular TRPV1 channels that are endogenously expressed in both vascular smooth muscle and endothelial cells and emphasizes the role of inflammation in sensitizing the TRPV1 channel to capsaicin activation. Tilting the balance between the beneficial vasodilatory action of capsaicin and its unwanted vasoconstrictive effects may precipitate adverse outcomes such as vasospasm and myocardial infarction, especially in the presence of proinflammatory mediators.
Collapse
|
18
|
Guo J, Chen W, Bao B, Zhang D, Pan J, Zhang M. Protective effect of berberine against LPS-induced endothelial cell injury via the JNK signaling pathway and autophagic mechanisms. Bioengineered 2021; 12:1324-1337. [PMID: 33896366 PMCID: PMC8806223 DOI: 10.1080/21655979.2021.1915671] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 04/07/2021] [Indexed: 12/14/2022] Open
Abstract
The role of autophagic mechanisms in the protective effect of berberine (BBR) on lipopolysaccharide (LPS)-induced injury in the endothelial cells human umbilical vein endothelial cells (HUVECs) and human pulmonary microvascular endothelial cells (HPMECs) was investigated. Cell viability, proliferation, and apoptosis were detected by the CCK-8 assay, the EdU kit, and flow cytometry, respectively, and autophagy-related protein expression, the number of autophagic vacuoles, and LC3 double-fluorescence were examined using western blot analysis, transmission electron microscopy, and confocal microscopy, respectively. LPS resulted in a decrease in the cell viability and proliferation of HUVECs and HPMECs and an increase in the number of apoptotic cells, while BBR treatment resulted in an increase in cell viability and proliferation, as well as a decrease in cell apoptosis. Furthermore, BBR could inhibit LPS-induced autophagy, as demonstrated by its inhibitory effects on the LC3-II/LC3-I ratio and Beclin-1 levels and its promotive effect on p62 expression. Addition of the autophagy inducer rapamycin (RAPA) aggravated LPS-induced injury, while treatment with the autophagy blocker 3-methyladenine (3-MA) attenuated the injury. Further, the protective effect of BBR was inhibited by rapamycin. JNK inhibition by SP600125 inhibited LPS-induced autophagy, and BBR could not alter the LPS-induced autophagy in HUVECs and HPMECs that were pretreated with SP600125. The present data indicate that BBR attenuated LPS-induced cell apoptosis by blocking JNK-mediated autophagy in HUVECs and HPMECs. Therefore, the JNK-mediated autophagy pathway could be a potential target for the prevention and treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Junping Guo
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Clinical Medicine, Zhejiang University City College, School of Medicine, Hangzhou, China
| | - Wei Chen
- Cancer Institute of Integrated Traditional Chinese and Western Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Beibei Bao
- Department of Clinical Medicine, Zhejiang University City College, School of Medicine, Hangzhou, China
| | - Dayong Zhang
- Department of Clinical Medicine, Zhejiang University City College, School of Medicine, Hangzhou, China
| | - Jianping Pan
- Department of Clinical Medicine, Zhejiang University City College, School of Medicine, Hangzhou, China
| | - Mao Zhang
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institue of Emergency Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
19
|
Kai H, Wu Q, Yin R, Tang X, Shi H, Wang T, Zhang M, Pan C. LncRNA NORAD Promotes Vascular Endothelial Cell Injury and Atherosclerosis Through Suppressing VEGF Gene Transcription via Enhancing H3K9 Deacetylation by Recruiting HDAC6. Front Cell Dev Biol 2021; 9:701628. [PMID: 34307380 PMCID: PMC8301222 DOI: 10.3389/fcell.2021.701628] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/08/2021] [Indexed: 11/15/2022] Open
Abstract
Coronary artery disease (CAD) is a major atherosclerotic cardiovascular disease and the leading cause of mortality globally. Long non-coding RNAs (lncRNAs) play crucial roles in CAD development. To date, the effect of lncRNA non-coding RNA activated by DNA damage (NORAD) on atherosclerosis in CAD remains unclear. The primary aim of this study was to investigate the effect of lncRNA NORAD on vascular endothelial cell injury and atherosclerosis. Here, ox-LDL-treated human umbilical vein endothelial cells (HUVECs) and high-fat-diet (HFD)-fed ApoE–/– mice were utilized as in vitro and in vivo models. The present study found that lncRNA NORAD expression was increased in ox-LDL-treated HUVECs and thoracic aorta of atherosclerotic mice, and knockdown of lncRNA NORAD alleviated vascular endothelial cell injury and atherosclerosis development in vitro and in vivo. Knockdown of lncRNA NORAD aggravated ox-LDL-reduced or atherosclerosis-decreased vascular endothelial growth factor (VEGF) expression in HUVECs and thoracic aorta of mice to ameliorate vascular endothelial cell injury and atherosclerosis development. Moreover, nucleus lncRNA NORAD suppressed VEGF gene transcription through enhancing H3K9 deacetylation via recruiting HDAC6 to the VEGF gene promoter in ox-LDL-treated HUVECs. In addition, VEGF reduced FUS (FUS RNA binding protein) expression by a negative feedback regulation in HUVECs. In summary, lncRNA NORAD enhanced vascular endothelial cell injury and atherosclerosis through suppressing VEGF gene transcription via enhancing H3K9 deacetylation by recruiting HDAC6. The findings could facilitate discovering novel diagnostic markers and therapeutic targets for CAD.
Collapse
Affiliation(s)
- Huihua Kai
- Department of Radiology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Qiyong Wu
- Department of Thoracic and Cardiac Surgery, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Ruohan Yin
- Department of Radiology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Xiaoqiang Tang
- Department of Radiology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Haifeng Shi
- Department of Radiology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Tao Wang
- Department of Radiology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Ming Zhang
- Department of Radiology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| | - Changjie Pan
- Department of Radiology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, China
| |
Collapse
|
20
|
Driscoll K, Cruz AD, Butcher JT. Inflammatory and Biomechanical Drivers of Endothelial-Interstitial Interactions in Calcific Aortic Valve Disease. Circ Res 2021; 128:1344-1370. [PMID: 33914601 DOI: 10.1161/circresaha.121.318011] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Calcific aortic valve disease is dramatically increasing in global burden, yet no therapy exists outside of prosthetic replacement. The increasing proportion of younger and more active patients mandates alternative therapies. Studies suggest a window of opportunity for biologically based diagnostics and therapeutics to alleviate or delay calcific aortic valve disease progression. Advancement, however, has been hampered by limited understanding of the complex mechanisms driving calcific aortic valve disease initiation and progression towards clinically relevant interventions.
Collapse
Affiliation(s)
| | - Alexander D Cruz
- Meinig School of Biomedical Engineering, Cornell University, Ithaca NY
| | | |
Collapse
|
21
|
Yamagata K. Prevention of Endothelial Dysfunction and Cardiovascular Disease by n-3 Fatty Acids-Inhibiting Action on Oxidative Stress and Inflammation. Curr Pharm Des 2021; 26:3652-3666. [PMID: 32242776 DOI: 10.2174/1381612826666200403121952] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/11/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND Prospective cohort studies and randomized controlled trials have shown the protective effect of n-3 fatty acids against cardiovascular disease (CVD). The effect of n-3 fatty acids on vascular endothelial cells indicates their possible role in CVD prevention. OBJECTIVE Here, we describe the effect of docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) on endothelial dysfunction-caused by inflammation and oxidative stress-and their role in the development of CVD. METHODS We reviewed epidemiological studies done on n-3 fatty acids in CVD. The effect of DHA and EPA on vascular endothelial cells was examined with regard to changes in various markers, such as arteriosclerosis, inflammation, and oxidative stress, using cell and animal models. RESULTS Epidemiological studies revealed that dietary intake of EPA and DHA was associated with a reduced risk of various CVDs. EPA and DHA inhibited various events involved in arteriosclerosis development by preventing oxidative stress and inflammation associated with endothelial cell damage. In particular, EPA and DHA prevented endothelial cell dysfunction mediated by inflammatory responses and oxidative stress induced by events related to CVD. DHA and EPA also increased eNOS activity and induced nitric oxide production. CONCLUSION The effects of DHA and EPA on vascular endothelial cell damage and dysfunction may involve the induction of nitric oxide, in addition to antioxidant and anti-inflammatory effects. n-3 fatty acids inhibit endothelial dysfunction and prevent arteriosclerosis. Therefore, the intake of n-3 fatty acids may prevent CVDs, like myocardial infarction and stroke.
Collapse
Affiliation(s)
- Kazuo Yamagata
- College of Bioresource Science, Nihon University (UNBS), Kanagawa, Japan
| |
Collapse
|
22
|
Calciprotein Particles Cause Endothelial Dysfunction under Flow. Int J Mol Sci 2020; 21:ijms21228802. [PMID: 33233811 PMCID: PMC7699979 DOI: 10.3390/ijms21228802] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/11/2020] [Accepted: 11/19/2020] [Indexed: 12/20/2022] Open
Abstract
Calciprotein particles (CPPs), which increasingly arise in the circulation during the disorders of mineral homeostasis, represent a double-edged sword protecting the human organism from extraskeletal calcification but potentially causing endothelial dysfunction. Existing models, however, failed to demonstrate the detrimental action of CPPs on endothelial cells (ECs) under flow. Here, we applied a flow culture system, where human arterial ECs were co-incubated with CPPs for 4 h, and a normolipidemic and normotensive rat model (10 daily intravenous injections of CPPs) to simulate the scenario occurring in vivo in the absence of confounding cardiovascular risk factors. Pathogenic effects of CPPs were investigated by RT-qPCR and Western blotting profiling of the endothelial lysate. CPPs were internalised within 1 h of circulation, inducing adhesion of peripheral blood mononuclear cells to ECs. Molecular profiling revealed that CPPs stimulated the expression of pro-inflammatory cell adhesion molecules VCAM1 and ICAM1 and upregulated transcription factors of endothelial-to-mesenchymal transition (Snail, Slug and Twist1). Furthermore, exposure to CPPs reduced the production of atheroprotective transcription factors KLF2 and KLF4 and led to YAP1 hypophosphorylation, potentially disturbing the mechanisms responsible for the proper endothelial mechanotransduction. Taken together, our results suggest the ability of CPPs to initiate endothelial dysfunction at physiological flow conditions.
Collapse
|
23
|
Co-Culture of Primary Human Coronary Artery and Internal Thoracic Artery Endothelial Cells Results in Mutually Beneficial Paracrine Interactions. Int J Mol Sci 2020; 21:ijms21218032. [PMID: 33126651 PMCID: PMC7663246 DOI: 10.3390/ijms21218032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/21/2020] [Accepted: 10/27/2020] [Indexed: 12/12/2022] Open
Abstract
Although saphenous veins (SVs) are commonly used as conduits for coronary artery bypass grafting (CABG), internal thoracic artery (ITA) grafts have significantly higher long-term patency. As SVs and ITA endothelial cells (ECs) have a considerable level of heterogeneity, we suggested that synergistic paracrine interactions between CA and ITA ECs (HCAECs and HITAECs, respectively) may explain the increased resistance of ITA grafts and adjacent CAs to atherosclerosis and restenosis. In this study, we measured the gene and protein expression of the molecules responsible for endothelial homeostasis, pro-inflammatory response, and endothelial-to-mesenchymal transition in HCAECs co-cultured with either HITAECs or SV ECs (HSaVECs) for an ascending duration. Upon the co-culture, HCAECs and HITAECs showed augmented expression of endothelial nitric oxide synthase (eNOS) and reduced expression of endothelial-to-mesenchymal transition transcription factors Snail and Slug when compared to the HCAEC–HSaVEC model. HCAECs co-cultured with HITAECs demonstrated an upregulation of HES1, a master regulator of arterial specification, of which the expression was also exclusively induced in HSaVECs co-cultured with HCAECs, suggestive of their arterialisation. In addition, co-culture of HCAECs and HITAECs promoted the release of pro-angiogenic molecules. To conclude, co-culture of HCAECs and HITAECs results in reciprocal and beneficial paracrine interactions that might contribute to the better performance of ITA grafts upon CABG.
Collapse
|
24
|
Cartilage oligomeric matrix protein fine-tunes disturbed flow-induced endothelial activation and atherogenesis. Matrix Biol 2020; 95:32-51. [PMID: 33068727 DOI: 10.1016/j.matbio.2020.10.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/15/2020] [Accepted: 10/11/2020] [Indexed: 12/27/2022]
Abstract
Disturbed flow leads to increased inflammatory responses of endothelial cells (ECs) prone to atherogenic state. Currently, little is known about the physiological mechanisms protecting vasculature against disturbed flow-activated ECs leading to atherosclerosis. Understanding the protective mediators involved in EC activation could provide novel therapeutic strategies for atherosclerosis. The extracellular matrix microenvironment profoundly regulates cellular homeostasis. A non-EC resident ECM protein, cartilage oligomeric matrix protein (COMP), has diverse protective roles in the cardiovascular system. To determine whether COMP could protect against disturbed flow-activated EC and atherosclerosis, we compared oscillatory shear stress (OSS) induced EC activation coated with various ECM proteins. Purified COMP inhibited EC activation caused by OSS. EC activation was upregulated in the aortic arch where the flow is disturbed in COMP-/- mice as compared with wild-type mice under physiological conditions or pathologically in partially ligated mouse carotid arteries. Mechanistically, co-immunoprecipitation, mammalian two-hybrid and FRET assay results suggest that COMP bound directly to integrin α5 via its C-terminus. We next synthesized a COMP-derived peptidomimetics (CCPep24) mimicking a specific COMP-integrin α5 interaction and found that CCPep24 protected against EC activation and atherogenesis in vivo. This study extends our current understanding of how ECM and flow coordinately fine-tune EC homeostasis and reveals the potential therapeutic effect of COMP or COMP-derived peptidomimetics on blocking aberrant integrin α5 activation, inflammatory EC activation and atherosclerosis pathogenesis.
Collapse
|
25
|
Vasilieva E, Gianella S, Freeman ML. Novel Strategies to Combat CMV-Related Cardiovascular Disease. Pathog Immun 2020; 5:240-274. [PMID: 33089035 PMCID: PMC7556413 DOI: 10.20411/pai.v5i1.382] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/08/2020] [Indexed: 12/20/2022] Open
Abstract
Cytomegalovirus (CMV), a ubiquitous human pathogen that is never cleared from the host, has long been thought to be relatively innocuous in immunocompetent adults, but causes severe complications including blindness, end-organ disease, and death in newborns and in immuno-compromised individuals, such as organ transplant recipients and those suffering from AIDS. Yet even in persons with intact immunity, CMV infection is associated with profound stimulation of immune and inflammatory pathways. Carriers of CMV infection also have an elevated risk of developing cardiovascular complications. In this review, we define the proposed mechanisms of how CMV contributes to cardiovascular disease (CVD), describe current approaches to target CMV, and discuss how these strategies may or may not alleviate cardiovascular complications in those with CMV infection. In addition, we discuss the special situation of CMV coinfection in people with HIV infection receiving antiretroviral therapy, and describe how these 2 viral infections may interact to potentiate CVD in this especially vulnerable population.
Collapse
Affiliation(s)
- Elena Vasilieva
- Laboratory of Atherothrombosis, Moscow State University of Medicine and Dentistry, Moscow 127473, Russia
| | - Sara Gianella
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Michael L. Freeman
- Division of Infectious Diseases and HIV Medicine; Department of Medicine; Case Western Reserve University, Cleveland, Ohio, United States
| |
Collapse
|
26
|
Dubchenko E, Ivanov A, Spirina N, Smirnova N, Melnikov M, Boyko A, Gusev E, Kubatiev A. Hyperhomocysteinemia and Endothelial Dysfunction in Multiple Sclerosis. Brain Sci 2020; 10:brainsci10090637. [PMID: 32947812 PMCID: PMC7564574 DOI: 10.3390/brainsci10090637] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/07/2020] [Accepted: 09/14/2020] [Indexed: 12/24/2022] Open
Abstract
Endothelial dysfunction is recognized as one of the leading factors in the pathogenesis of diseases of the central nervous system of various etiologies. Numerous studies have shown the role of hyperhomocysteinemia in the development of endothelial dysfunction and the prothrombogenic state. The most important condition in the development of multiple sclerosis (MS) is a dysregulation of the blood-brain barrier (BBB) and transendothelial leukocyte migration. It has been proven that homocysteine also contributes to the damage of neurons by the mechanism of excitotoxicity and the induction of the apoptosis of neurons. These processes can be one of the factors of neurodegenerative brain damage, which plays a leading role in the progression of MS. This review describes the pleiotropic effect of homocysteine on these processes and its role in MS pathogenesis.
Collapse
Affiliation(s)
- Ekaterina Dubchenko
- Department of Neuroimmunology of Federal Center of Brain and Neurotechnology of the Federal Medical-Biological Agency of Russia, 117997 Moscow, Russia; (E.D.); (N.S.); (A.B.)
- Interdistrict Department of Multiple Sclerosis at the State Clinical Hospital VV Veresaeva, 127644 Moscow, Russia
| | - Alexander Ivanov
- Federal State Budgetary Scientific Institution “Institute of General Pathology and Pathophysiology”, 125315 Moscow, Russia; (A.I.); (A.K.)
| | - Natalia Spirina
- Department of Nervous Diseases with Medical Genetics and Neurosurgery Yaroslavl State Medical University, 150000 Yaroslavl, Russia;
| | - Nina Smirnova
- Department of Neuroimmunology of Federal Center of Brain and Neurotechnology of the Federal Medical-Biological Agency of Russia, 117997 Moscow, Russia; (E.D.); (N.S.); (A.B.)
- Department of Neurology, Neurosurgery and Medical Genetic of Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
| | - Mikhail Melnikov
- Department of Neuroimmunology of Federal Center of Brain and Neurotechnology of the Federal Medical-Biological Agency of Russia, 117997 Moscow, Russia; (E.D.); (N.S.); (A.B.)
- Department of Neurology, Neurosurgery and Medical Genetic of Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
- Laboratory of Clinical Immunology, National Research Center Institute of Immunology of the Federal Medical-Biological Agency of Russia, 115478 Moscow, Russia
- Correspondence: ; Tel.: +7-926-331-8946
| | - Alexey Boyko
- Department of Neuroimmunology of Federal Center of Brain and Neurotechnology of the Federal Medical-Biological Agency of Russia, 117997 Moscow, Russia; (E.D.); (N.S.); (A.B.)
- Department of Neurology, Neurosurgery and Medical Genetic of Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
| | - Evgeniy Gusev
- Department of Neurology, Neurosurgery and Medical Genetic of Pirogov Russian National Research Medical University, 117997 Moscow, Russia;
| | - Aslan Kubatiev
- Federal State Budgetary Scientific Institution “Institute of General Pathology and Pathophysiology”, 125315 Moscow, Russia; (A.I.); (A.K.)
| |
Collapse
|
27
|
Sun S, Ji Z, Fu J, Wang XF, Zhang LS. Endosulfan induces endothelial inflammation and dysfunction via IRE1α/NF-κB signaling pathway. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:26163-26171. [PMID: 32361974 DOI: 10.1007/s11356-020-09023-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 04/22/2020] [Indexed: 05/23/2023]
Abstract
Cardiovascular diseases are related to vascular endothelial cell injury; our previous studies showed that endosulfan could cause hypercoagulation of blood by inducing endothelial cell injury. To clarify the mechanism of it, we treated human umbilical vein endothelial cells (HUVECs) with 0, 1, 5, and 10 μg/mL endosulfan, while in the inhibition groups, reactive oxygen species (ROS) inhibitor N-acetylcysteine (NAC, 3 mmol) and endoplasmic reticulum (ER) stress inhibitor (STF-083010, 10 μmol) were incubated prior to endosulfan. The results showed that endosulfan could induce inflammatory response and dysfunction by increasing the release of inflammatory cytokines such as interleukin-1β (IL-1β), interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), and adhesion molecules such as vascular cell adhesion molecule 1 (VCAM-1) and endothelin-1 (ET-1), and inducing ROS production in HUVECs. We also found that endosulfan could cause ER damage, remarkably increase the expressions of inositol-requiring enzyme 1α (IRE1α), phosphorylated IRE1α (p-IRE1α), GRP78, XBP1, nuclear factor-kappa B (NF-κB), and phosphorylated NF-κB (p-NF-κB) in HUVECs. The presence of NAC antagonized the ROS production, expressions of IRE1α and p-IRE1α; however, STF-083010 could decrease the expression levels of GRP78, XBP1, NF-κB, and p-NF-κB and attenuate IL-1β, IL-6, TNF-α, VCAM-1, and ET-1 release induced by endosulfan. These results demonstrated that endosulfan-induced endothelial inflammation and dysfunction through the IRE1α/NF-κB signaling pathway may be triggered by oxidative stress. The study provided experimental basis for the correlation between environmental pollutants (endosulfan) and cardiovascular diseases.
Collapse
Affiliation(s)
- ShiTian Sun
- College of Clinical Medicine, Binzhou Medical University, Yantai, People's Republic of China
| | - ZhengGuo Ji
- Department of Urology, Beijing Friendship Hospital, Capital medical University, Beijing, China
| | - JiaRong Fu
- College of Clinical Medicine, Binzhou Medical University, Yantai, People's Republic of China
| | - Xi-Feng Wang
- Department of Critical Care Medicine, Yu Huang Ding Hospital, Qingdao University, Yantai, 264000, People's Republic of China.
| | - Lian-Shuang Zhang
- Department of Histology and Embryology, Binzhou Medical University, Yantai, 264003, People's Republic of China.
| |
Collapse
|
28
|
Bian W, Jing X, Yang Z, Shi Z, Chen R, Xu A, Wang N, Jiang J, Yang C, Zhang D, Li L, Wang H, Wang J, Sun Y, Zhang C. Downregulation of LncRNA NORAD promotes Ox-LDL-induced vascular endothelial cell injury and atherosclerosis. Aging (Albany NY) 2020; 12:6385-6400. [PMID: 32267831 PMCID: PMC7185106 DOI: 10.18632/aging.103034] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 02/25/2020] [Indexed: 12/17/2022]
Abstract
Long noncoding RNAs (lncRNAs) play important roles in the development of vascular diseases. However, the effect of lncRNA NORAD on atherosclerosis remains unknown. This study aimed to investigate the effect NORAD on endothelial cell injury and atherosclerosis. Ox-LDL-treated human umbilical vein endothelial cells (HUVECs) and high-fat-diet (HFD)-fed ApoE−/− mice were used as in vitro and in vivo models. Results showed that NORAD-knockdown induced cell cycle arrest in G0/G1 phase, aggravated ox-LDL-induced cell viability reduction, cell apoptosis, and cell senescence along with the increased expression of Bax, P53, P21 and cleaved caspase-3 and the decreased expression of Bcl-2. The effect of NORAD on cell viability was further verified via NORAD-overexpression. NORAD- knockdown increased ox-LDL-induced reactive oxygen species, malondialdehyde, p-IKBα expression levels and NF-κB nuclear translocation. Proinflammatory molecules ICAM, VCAM, and IL-8 were also increased by NORAD- knockdown. Additionally, we identified the strong interaction of NORAD and IL-8 transcription repressor SFPQ in HUVECs. In ApoE−/− mice, NORAD-knockdown increased the lipid disorder and atherosclerotic lesions. The results have suggested that lncRNA NORAD attenuates endothelial cell senescence, endothelial cell apoptosis, and atherosclerosis via NF-κB and p53–p21 signaling pathways and IL-8, in which NORAD-mediated effect on IL-8 might through the direct interaction with SFPQ.
Collapse
Affiliation(s)
- Weihua Bian
- Department of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Xiaohong Jing
- Department of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Zhiyu Yang
- Department of Gastroenterology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Zhen Shi
- Department of Basic Medicine, Binzhou Medical University, Yantai 264003, China
| | - Ruiyao Chen
- Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou 325027, China
| | - Aili Xu
- Department of Basic Medicine, Binzhou Medical University, Yantai 264003, China
| | - Na Wang
- Department of Basic Medicine, Binzhou Medical University, Yantai 264003, China
| | - Jing Jiang
- Department of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Cheng Yang
- Department of Basic Medicine, Binzhou Medical University, Yantai 264003, China
| | - Daolai Zhang
- Department of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Lan Li
- Department of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Haiyan Wang
- Department of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Juan Wang
- Department of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Yeying Sun
- Department of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Chunxiang Zhang
- Department of Pharmacy, Binzhou Medical University, Yantai 264003, China.,Children's Heart Center, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou 325027, China
| |
Collapse
|
29
|
Yu QQ, Cheng DX, Xu LR, Li YK, Zheng XY, Liu Y, Li YF, Liu HL, Bai L, Wang R, Fan JL, Liu EQ, Zhao SH. Urotensin II and urantide exert opposite effects on the cellular components of atherosclerotic plaque in hypercholesterolemic rabbits. Acta Pharmacol Sin 2020; 41:546-553. [PMID: 31685976 PMCID: PMC7468446 DOI: 10.1038/s41401-019-0315-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 09/30/2019] [Indexed: 12/26/2022]
Abstract
Increasing levels of plasma urotensin II (UII) are positively associated with atherosclerosis. In this study we investigated the role of macrophage-secreted UII in atherosclerosis progression, and evaluated the therapeutic value of urantide, a potent competitive UII receptor antagonist, in atherosclerosis treatment. Macrophage-specific human UII-transgenic rabbits and their nontransgenic littermates were fed a high cholesterol diet for 16 weeks to induce atherosclerosis. Immunohistochemical staining of the cellular components (macrophages and smooth muscle cells) of aortic atherosclerotic lesions revealed a significant increase (52%) in the macrophage-positive area in only male transgenic rabbits compared with that in the nontransgenic littermates. However, both male and female transgenic rabbits showed a significant decrease (45% in males and 31% in females) in the smooth muscle cell-positive area compared with that of their control littermates. The effects of macrophage-secreted UII on the plaque cellular components were independent of plasma lipid level. Meanwhile the wild-type rabbits were continuously subcutaneously infused with urantide (5.4 µg· kg-1· h-1) using osmotic mini-pumps. Infusion of urantide exerted effects opposite to those caused by UII, as it significantly decreased the macrophage-positive area in male wild-type rabbits compared with that of control rabbits. In cultured human umbilical vein endothelial cells, treatment with UII dose-dependently increased the expression of the adhesion molecules VCAM-1 and ICAM-1, and this effect was partially reversed by urantide. The current study provides direct evidence that macrophage-secreted UII plays a key role in atherogenesis. Targeting UII with urantide may promote plaque stability by decreasing macrophage-derived foam cell formation, which is an indicator of unstable plaque.
Collapse
|
30
|
Dubchenko EA, Ivanov AV, Boiko AN, Spirina NN, Gusev EI, Kubatiev AA. [Hyperhomocysteinemia and endothelial dysfunction in patients with cerebral vascular and autoimmune diseases]. Zh Nevrol Psikhiatr Im S S Korsakova 2020; 119:133-138. [PMID: 31851185 DOI: 10.17116/jnevro2019119111133] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Endothelial dysfunction today is recognized as one of the leading factors in the pathogenesis of diseases of the central nervous system of various etiologies. Numerous studies have shown the role of hyperhomocysteinemia in the development of endothelial dysfunction and prothrombogenic state. The most important condition in the development of multiple sclerosis (MS) is dysregulation of the blood-brain barrier (BBB) and transendothelial leukocyte migration. It has been proven that homocysteine also contributes to the damage of neurons by the mechanism of excitotoxicity and induction of apoptosis of neurons. These processes can be one of the factors of neurodegenerative brain damage, which plays a leading role in the progression of MS. This review describes the pleiotropic effect of homocysteine on these processes and its role in the pathogenesis of MS.
Collapse
Affiliation(s)
- E A Dubchenko
- Pirogov Russian National Research Medical University, Moscow, Russia; Interdistrict Department of Multiple Sclerosis Veresaev Clinical Hospital Department of Health of Moscow, Russia, Moscow; Federal Institute of Cerebrovascular Pathology and Stroke, Russia, Moscow
| | - A V Ivanov
- Institute of General Pathology and Pathophysiology, Russia, Moscow
| | - A N Boiko
- Pirogov Russian National Research Medical University, Moscow, Russia; Federal Institute of Cerebrovascular Pathology and Stroke, Russia, Moscow
| | - N N Spirina
- Yaroslavl State Medical University, Yaroslavl, Russia
| | - E I Gusev
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - A A Kubatiev
- Institute of General Pathology and Pathophysiology, Russia, Moscow
| |
Collapse
|
31
|
Caradu C, Couffinhal T, Chapouly C, Guimbal S, Hollier PL, Ducasse E, Bura-Rivière A, Dubois M, Gadeau AP, Renault MA. Restoring Endothelial Function by Targeting Desert Hedgehog Downstream of Klf2 Improves Critical Limb Ischemia in Adults. Circ Res 2019; 123:1053-1065. [PMID: 30355159 DOI: 10.1161/circresaha.118.313177] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
RATIONALE Klf (kruppel-like factor) 2 is critical to establish and maintain endothelial integrity. OBJECTIVE Therefore, determining upstream and downstream mediators of Klf2 would lead to alternative therapeutic targets in cardiovascular disease management. METHODS AND RESULTS Here we identify Dhh (desert hedgehog) as a downstream effector of Klf2, whose expression in endothelial cells (ECs) is upregulated by shear stress and decreased by inflammatory cytokines. Consequently, we show that Dhh knockdown in ECs promotes endothelial permeability and EC activation and that Dhh agonist prevents TNF-α (tumor necrosis factor alpha) or glucose-induced EC dysfunction. Moreover, we demonstrate that human critical limb ischemia, a pathological condition linked to diabetes mellitus and inflammation, is associated to major EC dysfunction. By recreating a complex model of critical limb ischemia in diabetic mice, we found that Dhh-signaling agonist significantly improved EC function without promoting angiogenesis, which subsequently improved muscle perfusion. CONCLUSION Restoring EC function leads to significant critical limb ischemia recovery. Dhh appears to be a promising target, downstream of Klf2, to prevent the endothelial dysfunction involved in ischemic vascular diseases.
Collapse
Affiliation(s)
- Caroline Caradu
- From the Inserm U1034, Biology of Cardiovascular Diseases, CHU de Bordeaux, Pessac, France (C. Caradu, T.C., C. Chapouly, S.G., P.-L.H., E.D., A.-P.G., M.-A.R.)
| | - Thierry Couffinhal
- From the Inserm U1034, Biology of Cardiovascular Diseases, CHU de Bordeaux, Pessac, France (C. Caradu, T.C., C. Chapouly, S.G., P.-L.H., E.D., A.-P.G., M.-A.R.)
| | - Candice Chapouly
- From the Inserm U1034, Biology of Cardiovascular Diseases, CHU de Bordeaux, Pessac, France (C. Caradu, T.C., C. Chapouly, S.G., P.-L.H., E.D., A.-P.G., M.-A.R.)
| | - Sarah Guimbal
- From the Inserm U1034, Biology of Cardiovascular Diseases, CHU de Bordeaux, Pessac, France (C. Caradu, T.C., C. Chapouly, S.G., P.-L.H., E.D., A.-P.G., M.-A.R.)
| | - Pierre-Louis Hollier
- From the Inserm U1034, Biology of Cardiovascular Diseases, CHU de Bordeaux, Pessac, France (C. Caradu, T.C., C. Chapouly, S.G., P.-L.H., E.D., A.-P.G., M.-A.R.)
| | - Eric Ducasse
- From the Inserm U1034, Biology of Cardiovascular Diseases, CHU de Bordeaux, Pessac, France (C. Caradu, T.C., C. Chapouly, S.G., P.-L.H., E.D., A.-P.G., M.-A.R.)
| | | | - Mathilde Dubois
- Inserm U1045, Centre de recherche Cardio-thoracique, University of Bordeaux, France (M.D.)
| | - Alain-Pierre Gadeau
- From the Inserm U1034, Biology of Cardiovascular Diseases, CHU de Bordeaux, Pessac, France (C. Caradu, T.C., C. Chapouly, S.G., P.-L.H., E.D., A.-P.G., M.-A.R.)
| | - Marie-Ange Renault
- From the Inserm U1034, Biology of Cardiovascular Diseases, CHU de Bordeaux, Pessac, France (C. Caradu, T.C., C. Chapouly, S.G., P.-L.H., E.D., A.-P.G., M.-A.R.)
| |
Collapse
|
32
|
Using Yoda-1 to mimic laminar flow in vitro: A tool to simplify drug testing. Biochem Pharmacol 2019; 168:473-480. [PMID: 31437459 PMCID: PMC6852096 DOI: 10.1016/j.bcp.2019.08.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 08/14/2019] [Indexed: 01/24/2023]
Abstract
The endothelium is an attractive drug target and an important site of adverse drug reactions. Endothelial dysfunction is strongly associated with inflammation and contributes to drug-induced cardiovascular toxicity. Endothelial cells in the circulation are exposed to haemodynamic forces including shear stress. Including shear stress may improve future endothelial cell drug discovery or toxicity screening. Piezo-1 is required for endothelial cells to respond to shear stress. In this study, we investigated whether a small molecule activator of Piezo-1, Yoda-1, can mimic the effect of laminar flow-induced shear stress on endothelial cell inflammation, and endothelial cytotoxicity in response to the chemotherapy agent, doxorubicin. First, we tested whether Yoda-1 could mimic the effects of shear stress of expression of the endothelial adhesion molecules, ICAM-1 and VCAM-1. Human umbilical vein endothelial cells (HUVEC) were cultured in static conditions (with or without Yoda-1) or under laminar flow-induced shear stress (5 dyn/cm2). Yoda-1 and laminar flow had similar anti-inflammatory effects, reducing the ability of TNF-α to induce ICAM-1 and VCAM-1 expression. We then tested whether Yoda-1 could mimic the effect of shear stress on doxorubicin-induced cytotoxicity. Both laminar flow and Yoda-1 treatment of static cultures increased the cytotoxicity of doxorubicin. These findings show that Piezo-1 activation with Yoda-1 in static culture leads to an endothelial cell phenotype that mimics endothelial cells under laminar flow. Pharmacological activation of Piezo-1 may be a useful approach to mimic constant shear stress in static cultures, which may improve endothelial drug discovery and toxicity testing.
Collapse
|
33
|
Protective effect of Ziziphora clinopodioides flavonoids against H 2O 2-induced oxidative stress in HUVEC cells. Biomed Pharmacother 2019; 117:109156. [PMID: 31387192 DOI: 10.1016/j.biopha.2019.109156] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/15/2019] [Accepted: 06/19/2019] [Indexed: 02/07/2023] Open
Abstract
The present study was designed to study the protective effect of Ziziphora clinopodioides flavonoids (ZCF) against H2O2-induced oxidative stress in HUVEC cells. MTT assay was carried out to determine the cell viability of HUVEC cells following pretreatment with ZCF. Fluorescent microscopy measurements were performed to evaluate apoptosis of HUVEC cells. Furthermore, the effects of ZCF on the activities of antioxidants superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), catalase (CAT), malondialdehyde production (MDA) and lactic dehydrogenase (LDH) levels were analyzed. Apoptosis was observed by Hoechst33258 staining and AO staining. Real-time fluorescence quantitative polymerase chain reaction (Real-time PCR) was used to detect the expression of B-cell lymphoma/leukemia-2 (Bcl-2), Bcl-2-associated X protein (Bax) and aspartate proteolytic enzyme-3 (Caspase-3) mRNA. The expression of vascular endothelial growth factor receptor 2 (VEGFR2), protein kinase B (Akt), phosphorylated protein kinase B (p-Akt), Bax, Bcl-2 and Caspase-3 were detected by western blot. ZCF attenuated H2O2-induced cell death, as determined by the MTT assay. ZCF decreased malondialdehyde and lactic dehydrogenase levels, increased superoxide dismutase, glutathione peroxidase, catalase activities and inhibited apoptosis. Moreover, pretreatment with ZCF decreased the expression of Bax and Caspase-3 at mRNA level, increased the expression of Bcl-2 mRNA level, decreased the levels of VEGFR2, Bax and Caspase-3 protein, and increased the level of p-Akt / Akt and Bcl-2 protein in HUVEC cells. These results suggested that ZCF protected against H2O2-induced injury in HUVEC cells. The mechanism for this effect is related to the enhancement of antioxidant capacity, suppression of angiogenesis and apoptosis.
Collapse
|
34
|
Emamalipour M, Seidi K, Jahanban‐Esfahlan A, Jahanban‐Esfahlan R. Implications of resistin in type 2 diabetes mellitus and coronary artery disease: Impairing insulin function and inducing pro‐inflammatory cytokines. J Cell Physiol 2019; 234:21758-21769. [DOI: 10.1002/jcp.28913] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 05/13/2019] [Accepted: 05/20/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Melissa Emamalipour
- Department of Medical Biotechnology Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences Tabriz Iran
- Student Research Committee, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences Tabriz Iran
| | - Khaled Seidi
- Department of Medical Biotechnology Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences Tabriz Iran
- Student Research Committee, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences Tabriz Iran
| | - Ali Jahanban‐Esfahlan
- Department of Medical Biotechnology Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences Tabriz Iran
- Student Research Committee, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences Tabriz Iran
| | - Rana Jahanban‐Esfahlan
- Department of Medical Biotechnology Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences Tabriz Iran
- Student Research Committee, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
35
|
Haybar H, Shahrabi S, Rezaeeyan H, Shirzad R, Saki N. Endothelial Cells: From Dysfunction Mechanism to Pharmacological Effect in Cardiovascular Disease. Cardiovasc Toxicol 2019; 19:13-22. [PMID: 30506414 DOI: 10.1007/s12012-018-9493-8] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Endothelial cells (ECs) are the innermost layer of blood vessels that play important roles in homeostasis and vascular function. However, recent evidence suggests that the onset of inflammation and the production of reactive oxygen species impair the function of ECs and are a main factor in the development of cardiovascular disease (CVD). In this study, we investigated the effects of inflammatory markers, oxidative stress, and treatment on ECs in CVD patients. This review article is based on the material obtained from PubMed up to 2018. The key search terms used were "Cardiovascular Disease," "Endothelial Cell Dysfunction," "Inflammation," "Treatment," and "Oxidative Stress." The generation of reactive oxygen species (ROS) as well as reduced nitric oxide (NO) production by ECs impairs the function of blood vessels. Therefore, treatment of CVD patients leads to the expression of transcription factors activating anti-oxidant mechanisms and NO production. In contrast, NO production by inflammatory agents can cause ECs repair due to differentiation of endothelial progenitor cells (EPCs). Therefore, identifying the molecular pathways leading to the differentiation of EPCs through mediation of factors induced by inflammatory factors can be effective in regenerative medicine for ECs repair.
Collapse
Affiliation(s)
- Habib Haybar
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeid Shahrabi
- Department of Biochemistry and Hematology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Hadi Rezaeeyan
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Reza Shirzad
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
36
|
Gu L, Ye P, Li H, Wang Y, Xu Y, Tian Q, Lei G, Zhao C, Gao Z, Zhao W, Tan S. Lunasin attenuates oxidant-induced endothelial injury and inhibits atherosclerotic plaque progression in ApoE -/- mice by up-regulating heme oxygenase-1 via PI3K/Akt/Nrf2/ARE pathway. FASEB J 2019; 33:4836-4850. [PMID: 30601695 DOI: 10.1096/fj.201802251r] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
Oxidative stress-induced vascular endothelial cell (VEC) injury is a major mechanism in the initiation and development of atherosclerosis. Lunasin, a soybean-derived 43-aa peptide, has been previously shown to possess potent antioxidant and anti-inflammatory activities other than its established anticancer activities. This study investigated the effects of lunasin on protecting VECs from oxidative damage and inhibiting atherosclerotic plaque progression in apolipoprotein E-deficient (ApoE-/-) mice and explored its underlying mechanism. Biochemical and histologic analyses were performed by using EA.hy926 human VECs and a high-fat diet (HFD) ApoE-/- mouse atherosclerosis model. Our data indicated that lunasin attenuated H2O2-induced, mitochondria-dependent endothelial apoptosis via down-regulating Bax and up-regulating Bcl-2, inhibiting the mitochondrial depolarization, and reducing the release of cytochrome c, as well as decreasing the activation of caspase-9 and caspase-3 in vitro and in vivo. Mechanic studies showed that lunasin significantly up-regulated heme oxygenase-1 via the PI3K/Akt/nuclear factor erythroid 2-related factor 2/antioxidant response element pathway, and reduced H2O2-induced ROS production in VECs, thereby attenuating oxidant-induced endothelial injury and inhibiting atherosclerotic plaque progression in ApoE-/- mice. In conclusion, our in vitro and in vivo data suggest that lunasin protects VECs from oxidative damage by enhancing heme oxygenase-1 expression via activation of the PI3K/Akt/nuclear factor erythroid 2-related factor 2/antioxidant response element pathway and inhibiting mitochondria-dependent apoptosis, thereby effectively attenuating atherosclerosis in HFD-fed ApoE-/- mice. Lunasin may act as a potential therapeutic agent for the prevention and treatment of atherosclerosis.-Gu, L., Ye, P., Li, H., Wang, Y., Xu, Y., Tian, Q., Lei, G., Zhao, C., Gao, Z., Zhao, W., Tan, S. Lunasin attenuates oxidant-induced endothelial injury and inhibits atherosclerotic plaque progression in ApoE-/- mice by up-regulating heme oxygenase-1 via PI3K/Akt/Nrf2/ARE pathway.
Collapse
Affiliation(s)
- Lili Gu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Pei Ye
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Hengli Li
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yue Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yaqiong Xu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Qinghua Tian
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Gaoxin Lei
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Cheng Zhao
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Zhan Gao
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Wenfeng Zhao
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Shuhua Tan
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
37
|
Lebedeva AM, Shpektor AV, Vasilieva EY, Margolis LB. Cytomegalovirus Infection in Cardiovascular Diseases. BIOCHEMISTRY (MOSCOW) 2019; 83:1437-1447. [PMID: 30878019 DOI: 10.1134/s0006297918120027] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Atherosclerosis underlies the development of many cardiovascular diseases that continue to hold a leading place among the causes of death in developed countries. The role of activated immune cells in atherosclerosis progression has been convincingly demonstrated, but the mechanism of their action remains poorly investigated. Since atherosclerosis is associated with chronic inflammatory response, involvement of viral and bacterial infections in atherogenesis has been examined. A special place among the infectious agents is held by human herpesviruses as the most common persistent viruses in human population coupled to chronic inflammation during atherosclerosis. We found that activation of cytomegalovirus (CMV, human herpesvirus 5) infection is associated with the emergence of acute coronary syndrome, which is in a good agreement with the data on productive CMV infection published elsewhere. In this review, we discuss the data obtained by us and other researchers regarding the role of cytomegalovirus infection and related potential mechanisms resulting in the expansion of atherosclerotic plaques during ischemic heart disease and stroke, including virus transfer to immune and endothelial cells via extracellular vesicles. In particular, the data presented in the review demonstrate that virus spreading in the vascular wall triggers immune system activation in atherosclerotic plaques and causes endothelial dysfunction. Moreover, productive CMV infection in patients with acute myocardial infarction correlates with the extent of endothelial dysfunction. The mechanisms described by us and other researchers may explain the role of CMV infection in atherosclerosis and development of ischemic heart disease.
Collapse
Affiliation(s)
- A M Lebedeva
- Department of Cardiology and Laboratory of Atherothrombosis, A. I. Evdokimov Moscow State University of Medicine and Dentistry, Ministry of Health of the Russian Federation, Moscow, 127473, Russia.
| | - A V Shpektor
- Department of Cardiology and Laboratory of Atherothrombosis, A. I. Evdokimov Moscow State University of Medicine and Dentistry, Ministry of Health of the Russian Federation, Moscow, 127473, Russia
| | - E Yu Vasilieva
- Department of Cardiology and Laboratory of Atherothrombosis, A. I. Evdokimov Moscow State University of Medicine and Dentistry, Ministry of Health of the Russian Federation, Moscow, 127473, Russia
| | - L B Margolis
- Section on Intercellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
38
|
Agouni A, Parray AS, Akhtar N, Mir FA, Bourke PJ, Joseph S, Morgan DM, Santos MD, Wadiwala MF, Kamran S, Sivaraman SK, Shuaib A. There Is Selective Increase in Pro-thrombotic Circulating Extracellular Vesicles in Acute Ischemic Stroke and Transient Ischemic Attack: A Study of Patients From the Middle East and Southeast Asia. Front Neurol 2019; 10:251. [PMID: 30941096 PMCID: PMC6434679 DOI: 10.3389/fneur.2019.00251] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 02/25/2019] [Indexed: 12/14/2022] Open
Abstract
Stroke attacks were found to be present at a younger age in patients from Southeast Asia (SE) and the Middle East (ME) resident in the state of Qatar. Extracellular vesicles (EVs), which are small membrane vesicles with pro-thrombotic properties, may contribute to the high risk of stroke in this population. Thus, total and cell-specific medium size EVs were counted by flow cytometry in platelet-free plasma from healthy volunteers and patients with transient ischemic attacks (TIA) and acute ischemic stroke (AIS) from SE and ME. Acutely, within 48 h of attacks, there was an increase in total endothelial EVs in TIA (6.73 ± 1.77; P = 0.0156; n = 21) and AIS (11.23 ± 1.95; P = 0.0007; n = 66) patients compared to controls (2.04 ± 0.78; n = 24). Similar increases were also evident in EVs originating from platelets, erythrocytes, granulocytes, and leukocytes. Compared to controls, there was also an increase in EVs derived from activated endothelial cells, platelets, granulocytes, leukocytes, and pro-coagulant EVs (Annexin V+) at 5 and 30-days following the acute events, while a decrease was observed in erythrocyte-derived EVs. This is the first study characterizing EVs in TIA and AIS patients from ME and SE showing an increase in EVs associated with endothelial and platelet cell activation, which may contribute to the elevated risk of stroke at a younger age in this population.
Collapse
Affiliation(s)
- Abdelali Agouni
- Department of Pharmaceutical Sciences, College of Pharmacy, Qatar University, Doha, Qatar
| | - Aijaz S Parray
- The Stroke Program, The Neuroscience Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Naveed Akhtar
- The Stroke Program, The Neuroscience Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Fayaz A Mir
- Interim Translational Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Paula J Bourke
- The Stroke Program, The Neuroscience Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Sujata Joseph
- The Stroke Program, The Neuroscience Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Deborah M Morgan
- The Stroke Program, The Neuroscience Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Mark D Santos
- The Stroke Program, The Neuroscience Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Muhammad F Wadiwala
- The Stroke Program, The Neuroscience Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Saadat Kamran
- The Stroke Program, The Neuroscience Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Siveen K Sivaraman
- Interim Translational Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Ashfaq Shuaib
- The Stroke Program, The Neuroscience Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar.,Department of Medicine (Neurology), University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
39
|
Song Y, Yang L, Guo R, Lu N, Shi Y, Wang X. Long noncoding RNA MALAT1 promotes high glucose-induced human endothelial cells pyroptosis by affecting NLRP3 expression through competitively binding miR-22. Biochem Biophys Res Commun 2018; 509:359-366. [PMID: 30591217 DOI: 10.1016/j.bbrc.2018.12.139] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 12/18/2018] [Indexed: 11/28/2022]
Abstract
Cell death and inflammation play critical roles in atherosclerosis. Pyroptosis, a novel proinflammatory programmed cell death process, participates in atherosclerosis pathogenesis. Recently, MALAT1 was identified as a pyroptosis-related long noncoding RNA (lncRNA). Here, we investigated the potential role and underlying mechanism of lncRNA MALAT1 in endothelial cells pyroptosis. We first established an endothelial cell pyroptosis model by stimulating EA.hy926 human endothelial cells (EA.hy926 cells) with high glucose. Then, we investigated lncRNA MALAT1 expression and found that it was upregulated in high glucose-treated EA.hy926 cells. Furthermore, lncRNA MALAT1 knockdown significantly inhibited high glucose-induced pyroptosis in EA.hy926 cells, which may critically influence atherosclerosis. Moreover, miR-22 was a target of lncRNA MALAT1 and was negatively correlated with lncRNA MALAT1. NLRP3 expression was significantly suppressed by transfection with a MALAT1-targeting antisense oligonucleotide (ASO). Ultimately, miR-22 overexpression abrogated the effect of MALAT1 on high glucose-induced EA.hy926 cells pyroptosis. Together, our results suggest that lncRNA MALAT1 promotes high glucose-induced pyroptosis of endothelial cells partly by affecting NLRP3 expression through competitively binding miR-22. Our findings indicate a new regulatory mechanism for endothelial cells pyroptosis under high-glucose stress, providing a novel therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Yaxian Song
- Department of Postgraduate, Kunming Medical University, Yunnan, 650500, China
| | - Lixia Yang
- Department of Cardiology, The 920th Hospital of Joint Logistics Support Force of the Chinese People's Liberation Army, Yunnan, China.
| | - Ruiwei Guo
- Department of Cardiology, The 920th Hospital of Joint Logistics Support Force of the Chinese People's Liberation Army, Yunnan, China
| | - Nihong Lu
- Department of Postgraduate, Kunming Medical University, Yunnan, 650500, China
| | - Yankun Shi
- Department of Cardiology, The 920th Hospital of Joint Logistics Support Force of the Chinese People's Liberation Army, Yunnan, China
| | - Xianmei Wang
- Department of Cardiology, The 920th Hospital of Joint Logistics Support Force of the Chinese People's Liberation Army, Yunnan, China
| |
Collapse
|
40
|
Kong Y, Gao Y, Lan D, Zhang Y, Zhan R, Liu M, Zhu Z, Zeng G, Huang Q. Trans-repression of NFκB pathway mediated by PPARγ improves vascular endothelium insulin resistance. J Cell Mol Med 2018; 23:216-226. [PMID: 30398029 PMCID: PMC6307800 DOI: 10.1111/jcmm.13913] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 07/23/2018] [Accepted: 08/20/2018] [Indexed: 02/07/2023] Open
Abstract
Previous study has shown that thiazolidinediones (TZDs) improved endothelium insulin resistance (IR) induced by high glucose concentration (HG)/hyperglycaemia through a PPARγ‐dependent‐NFκB trans‐repression mechanism. However, it is unclear, whether changes in PPARγ expression affect the endothelium IR and what the underlying mechanism is. In the present study, we aimed to address this issue. HG‐treated human umbilical vascular endothelial cells (HUVEC) were transfected by either PPARγ‐overexpressing (Ad‐PPARγ) or PPARγ‐shRNA‐containing (Ad‐PPARγ‐shRNA) adenoviral vectors. Likewise, the rats fed by high‐fat diet (HFD) were infected by intravenous administration of Ad‐PPARγ or Ad‐PPARγ‐shRNA. The levels of nitric oxide (NO), endothelin‐1 (ET‐1) and cytokines (TNFα, IL‐6, sICAM‐1 and sVCAM‐1) and the expression levels of PPARγ, eNOS, AKT, p‐AKT, IKKα/β and p‐IKKα/β and IκBα were examined; and the interaction between PPARγ and NFκB‐P65 as well as vascular function were evaluated. Our present results showed that overexpression of PPARγ notably increased the levels of NO, eNOS, p‐AKT and IκBα as well as the interaction of PPARγ and NFκB‐P65, and decreased the levels of ET‐1, p‐IKKα/β, TNFα, IL‐6, sICAM‐1 and sVCAM‐1. In contrast, down‐expression of PPARγ displayed the opposite effects. The results demonstrate that the overexpression of PPARγ improves while the down‐expression worsens the endothelium IR via a PPARγ‐mediated NFκB trans‐repression dependent manner. The findings suggest PPARγ is a potential therapeutic target for diabetic vascular complications.
Collapse
Affiliation(s)
- Ying Kong
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, China.,Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| | - Yan Gao
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, China.,Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| | - Dongyi Lan
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Ying Zhang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, China.,Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| | - Rixin Zhan
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, China.,Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| | - Meiqi Liu
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, China.,Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| | - Zhouan Zhu
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Guohua Zeng
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, China.,Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| | - Qiren Huang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi, China.,Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
41
|
The Yin and Yang of carbon nanomaterials in atherosclerosis. Biotechnol Adv 2018; 36:2232-2247. [PMID: 30342084 DOI: 10.1016/j.biotechadv.2018.10.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 10/06/2018] [Accepted: 10/15/2018] [Indexed: 02/06/2023]
Abstract
With unique characteristics such as high surface area, capacity of various functionalization, low weight, high conductivity, thermal and chemical stability, and free radical scavenging, carbon nanomaterials (CNMs) such as carbon nanotubes (CNTs), fullerene, graphene (oxide), carbon nanohorns (CNHs), and their derivatives have increasingly been utilized in nanomedicine and biomedicine. On the one hand, owing to ever-increasing applications of CNMs in technological and industrial fields as well as presence of combustion-derived CNMs in the ambient air, the skepticism has risen over the adverse effects of CNMs on human being. The influences of CNMs on cardiovascular system and cardiovascular diseases (CVDs) such as atherosclerosis, of which consequences are ischemic heart disease and ischemic stroke, as the main causes of death, is of paramount importance. In this regard, several studies have been devoted to specify the biomedical applications and cardiovascular toxicity of CNMs. Therefore, the aim of this review is to specify the roles and applications of various CNMs in atherosclerosis, and also identify the key role playing parameters in cardiovascular toxicity of CNMs so as to be a clue for prospective deployment of CNMs.
Collapse
|
42
|
Chen J, Zhang M, Zhu M, Gu J, Song J, Cui L, Liu D, Ning Q, Jia X, Feng L. Paeoniflorin prevents endoplasmic reticulum stress-associated inflammation in lipopolysaccharide-stimulated human umbilical vein endothelial cells via the IRE1α/NF-κB signaling pathway. Food Funct 2018; 9:2386-2397. [PMID: 29594285 DOI: 10.1039/c7fo01406f] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Endoplasmic reticulum (ER) stress-associated inflammation is a critical molecular mechanism involved in the pathogenesis of endothelial dysfunction (ED). Hence, strategies for alleviating ER stress-induced inflammation may be essential for the prevention of cardiovascular diseases. Paeoniflorin (PF), a bioactive compound from Paeonia lactiflora Pallas is known for its functional properties against vascular inflammation. However, to date, PF-mediated protection against ER stress-dependent inflammation has not been identified. Herein, we investigate the protective effect of PF on lipopolysaccharide (LPS)-stimulated human umbilical vein endothelial cell (HUVEC) injury and explore its underlying mechanism. The result of the cell viability assay indicates that PF promotes the cell survival rate in LPS-stimulated HUVECs. In addition, the LPS-induced over-production of inflammatory cytokines (interleukin-6 (IL-6) and monocyte chemotactic protein 1 (MCP-1)) and ER stress markers (78 kDa glucose regulated protein (GRP78) and CCAAT/enhancer binding protein homologous protein (CHOP)) are significantly decreased by PF and the ER stress inhibitor 4-phenylbutric acid (4-PBA). The transmission electron microscopy (TEM) assay implies that the ultrastructural abnormalities in ER are reversed by PF treatment, which is similar to the protective effect of 4-PBA. Impressively, we find that the inositol-requiring enzyme 1α (IRE1α)/nuclear factor-kappa B (NF-κB) pathway is significantly activated and contributes to the progress of LPS-induced HUVEC injury by promoting inflammatory cytokine production. IRE1α siRNA, AEBSF (ATF6 inhibitor), GSK2656157 (PERK inhibitor), PDTC (NF-κB inhibitor) and thapsigargin (TG, IRE1 activator) are used to confirm the role of the IRE1α/NF-κB pathway in PF-mediated protection against LPS-induced HUVEC injury. Our findings indicate that PF has an inhibitory effect on endothelial injury. To summarize, PF might be a potential therapeutic agent to inhibit ER stress-associated vascular inflammation.
Collapse
Affiliation(s)
- Juan Chen
- School of Life Sciences, Anhui University, Hefei 230601, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Xian T, Gan Y, Lu Y, Wang M, Yuan W, Zhou Y, Chen J, Wang K, Xiong S, Huang Q. Ameliorative effects of indomethacin at different concentrations on endothelial insulin resistance through two distinct pathways. Biomed Pharmacother 2018; 106:1161-1168. [PMID: 30119183 DOI: 10.1016/j.biopha.2018.07.069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/11/2018] [Accepted: 07/13/2018] [Indexed: 10/28/2022] Open
Abstract
Indomethacin (IDMT), a non-selective inhibitor of cycloxygenase-2 (COX-2), plays important roles in anti-inflammation and analgesia and it is commonly used to treat the patients with rheumatic and rheumatoid arthritis. Besides, various literatures reported that IDMT is a synthetic ligand of peroxisome proliferator activated receptor gamma (PPARγ). Rosiglitazone (RSG), an insulin-sensitizer, is also a synthetic ligand and applied clinically to cure the patients with type 2 diabetes mellitus. However, up to date little is known about whether IDMT ameliorates endothelial insulin resistance (IR). Accordingly, the purpose of this study is to investigate the effects of IDMT on endothelial IR and its underlying mechanism. Our present results showed that IDMT improved the endothelial IR induced by high glucose and fat concentration (HG/HF) in a concentration and time-dependent manner. Intriguingly, we further identified that 0.25 mM of IDMT noticeably induced the expression levels of PPARγ, AKT and endothelial nitric oxide synthase (eNOS) but failed to notably reverse the increases in expression levels of COX-2, inhibitory κB kinase (IKK) and tumor necrosis factor alpha (TNFα) induced by HG/HF; whereas 1.0 mM of IDMT exerted opposite effects compared with 0.25 mM of IDMT. Therefore, we conclude that IDMT ameliorates the endothelial IR induced by HG/HF through two distinct pathways, i.e., a lower concentration of IDMT through a PPARγ-AKT-eNOS pathway while a higher concentration mainly via an IKK-COX-2/TNFα pathway. The findings might provide a novel clinical use for IDMT to cure IR-related disorders.
Collapse
Affiliation(s)
- Tao Xian
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi Province, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Yuyang Gan
- Jiangxi Medical School, Nanchang University, Nanchang 330006, Jiangxi Province, PR China; Nanchang Joint Programme, Queen Mary University of London, London E1 4NS, UK
| | - Yi Lu
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi Province, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Mengxi Wang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi Province, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Wanwan Yuan
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi Province, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Yumeng Zhou
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi Province, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Junye Chen
- Jiangxi Medical School, Nanchang University, Nanchang 330006, Jiangxi Province, PR China; Nanchang Joint Programme, Queen Mary University of London, London E1 4NS, UK
| | - Kun Wang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi Province, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Shaofeng Xiong
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi Province, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, Jiangxi Province, PR China
| | - Qiren Huang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, Jiangxi Province, PR China; Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, Jiangxi Province, PR China.
| |
Collapse
|
44
|
Su G, Sun G, Liu H, Shu L, Liang Z. Downregulation of miR-34a promotes endothelial cell growth and suppresses apoptosis in atherosclerosis by regulating Bcl-2. Heart Vessels 2018; 33:1185-1194. [PMID: 29704100 DOI: 10.1007/s00380-018-1169-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 04/20/2018] [Indexed: 12/18/2022]
Abstract
Several miRNAs have been demonstrated to be involved in endothelial dysfunction during atherosclerosis (AS). However, the detailed roles and underlying mechanisms of miR-34a in AS-associated endothelial cell apoptosis are far from being addressed. Apolipoprotein E-deficient (ApoE-/-) mice fed with high-fat diet (HFD) were used as in vivo model of AS. Oxidized low-density lipoprotein (ox-LDL)-treated human aortic endothelial cells (HAECs) were applied as in vitro model of AS. The effects of miR-34a on atherosclerotic lesions were evaluated by hematoxylin-eosin (HE) and Oil Red O staining. Pecam-1+ endothelial cells were isolated from the aortic arch with flow cytometry. qRT-PCR and western blot were employed to measure gene and protein expression. The effects of miR-34a on cell viability, cell cycle distribution, and apoptosis were assessed by Cell counting kit (CCK)-8 and flow cytometry analysis. The relationship between miR-34a and Bcl-2 was confirmed by online softwares, luciferase reporter assay, and RNA immunoprecipitation (RIP). miR-34a was upregulated in HFD-induced ApoE-/- mice and ox-LDL-treated HAECs. Anti-miR-34a decreased atherosclerotic lesions and inhibited Pecam-1+ endothelial cells apoptosis in HFD-induced ApoE-/- mice. Moreover, anti-miR-34a significantly promoted cell viability, alleviated cell cycle arrest, and restrained apoptosis in ox-LDL-treated HAECs. Furthermore, Bcl-2 was identified as a target of miR-34a, and miR-34a inhibited Bcl-2 expression via binding to its 3'UTR. Rescue experiments demonstrated that Bcl-2 overexpression dramatically reversed miR-34a-mediated inhibition of cell growth and promotion of apoptosis in ox-LDL-exposed HAECs. Depletion of miR-34a facilitated endothelial cell growth and blocked apoptosis in AS by upregulating Bcl-2, offering a promising avenue for AS therapy.
Collapse
Affiliation(s)
- Gang Su
- Department of Cardiac Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 453100, China
| | - Guangli Sun
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 453100, China.
| | - Hai Liu
- Department of Cardiac Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 453100, China
| | - Liliang Shu
- Department of Cardiac Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 453100, China
| | - Zhenxing Liang
- Department of Cardiac Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 453100, China
| |
Collapse
|
45
|
Shear stress: An essential driver of endothelial progenitor cells. J Mol Cell Cardiol 2018; 118:46-69. [PMID: 29549046 DOI: 10.1016/j.yjmcc.2018.03.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/08/2018] [Accepted: 03/09/2018] [Indexed: 02/06/2023]
Abstract
The blood flow through vessels produces a tangential, or shear, stress sensed by their innermost layer (i.e., endothelium) and representing a major hemodynamic force. In humans, endothelial repair and blood vessel formation are mainly performed by circulating endothelial progenitor cells (EPCs) characterized by a considerable expression of vascular endothelial growth factor receptor 2 (VEGFR2), CD34, and CD133, pronounced tube formation activity in vitro, and strong reendothelialization or neovascularization capacity in vivo. EPCs have been proposed as a promising agent to induce reendothelialization of injured arteries, neovascularization of ischemic tissues, and endothelialization or vascularization of bioartificial constructs. A number of preconditioning approaches have been suggested to improve the regenerative potential of EPCs, including the use of biophysical stimuli such as shear stress. However, in spite of well-defined influence of shear stress on mature endothelial cells (ECs), articles summarizing how it affects EPCs are lacking. Here we discuss the impact of shear stress on homing, paracrine effects, and differentiation of EPCs. Unidirectional laminar shear stress significantly promotes homing of circulating EPCs to endothelial injury sites, induces anti-thrombotic and anti-atherosclerotic phenotype of EPCs, increases their capability to form capillary-like tubes in vitro, and enhances differentiation of EPCs into mature ECs in a dose-dependent manner. These effects are mediated by VEGFR2, Tie2, Notch, and β1/3 integrin signaling and can be abrogated by means of complementary siRNA/shRNA or selective pharmacological inhibitors of the respective proteins. Although the testing of sheared EPCs for vascular tissue engineering or regenerative medicine applications is still an unaccomplished task, favorable effects of unidirectional laminar shear stress on EPCs suggest its usefulness for their preconditioning.
Collapse
|
46
|
Whole-Transcriptome Sequencing: a Powerful Tool for Vascular Tissue Engineering and Endothelial Mechanobiology. High Throughput 2018; 7:ht7010005. [PMID: 29485616 PMCID: PMC5876531 DOI: 10.3390/ht7010005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/18/2018] [Accepted: 02/19/2018] [Indexed: 02/07/2023] Open
Abstract
Among applicable high-throughput techniques in cardiovascular biology, whole-transcriptome sequencing is of particular use. By utilizing RNA that is isolated from virtually all cells and tissues, the entire transcriptome can be evaluated. In comparison with other high-throughput approaches, RNA sequencing is characterized by a relatively low-cost and large data output, which permits a comprehensive analysis of spatiotemporal variation in the gene expression profile. Both shear stress and cyclic strain exert hemodynamic force upon the arterial endothelium and are considered to be crucial determinants of endothelial physiology. Laminar blood flow results in a high shear stress that promotes atheroresistant endothelial phenotype, while a turbulent, oscillatory flow yields a pathologically low shear stress that disturbs endothelial homeostasis, making respective arterial segments prone to atherosclerosis. Severe atherosclerosis significantly impairs blood supply to the organs and frequently requires bypass surgery or an arterial replacement surgery that requires tissue-engineered vascular grafts. To provide insight into patterns of gene expression in endothelial cells in native or bioartificial arteries under different biomechanical conditions, this article discusses applications of whole-transcriptome sequencing in endothelial mechanobiology and vascular tissue engineering.
Collapse
|
47
|
Perrin-Sarrado C, Dahboul F, Leroy P, Lartaud I. Aging and hypertension decrease endothelial NO-related dilating function and gamma-glutamyl transferase activity but notS-nitrosoglutathione-induced aortic vasodilation. Fundam Clin Pharmacol 2018; 32:134-140. [DOI: 10.1111/fcp.12347] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 11/10/2017] [Accepted: 12/22/2017] [Indexed: 12/25/2022]
Affiliation(s)
- Caroline Perrin-Sarrado
- EA3452 CITHEFOR ‘Drug Targets, Formulation and Preclinical Assessment’; Faculté de Pharmacie; Université de Lorraine; Nancy France
| | - Fatima Dahboul
- EA3452 CITHEFOR ‘Drug Targets, Formulation and Preclinical Assessment’; Faculté de Pharmacie; Université de Lorraine; Nancy France
| | - Pierre Leroy
- EA3452 CITHEFOR ‘Drug Targets, Formulation and Preclinical Assessment’; Faculté de Pharmacie; Université de Lorraine; Nancy France
| | - Isabelle Lartaud
- EA3452 CITHEFOR ‘Drug Targets, Formulation and Preclinical Assessment’; Faculté de Pharmacie; Université de Lorraine; Nancy France
| |
Collapse
|
48
|
Protective Effects of Methotrexate against Proatherosclerotic Cytokines: A Review of the Evidence. Mediators Inflamm 2017; 2017:9632846. [PMID: 29430085 PMCID: PMC5753000 DOI: 10.1155/2017/9632846] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 11/02/2017] [Accepted: 11/26/2017] [Indexed: 12/16/2022] Open
Abstract
There is good epidemiological evidence that patients with autoimmune rheumatic disease states, particularly rheumatoid arthritis, have an increased risk of cardiovascular morbidity and mortality when compared to the general population. The presence of a chronic systemic proinflammatory state in this patient group disrupts the structural and functional integrity of the endothelium and the arterial wall, favouring the onset and progression of atherosclerosis. A significant role in the detrimental effects of inflammation on endothelial function and vascular homeostasis is played by specific proatherosclerotic cytokines such as tumour necrosis factor-alpha (TNF-α), interleukin-1 (IL-1), and interleukin-6 (IL-6). Recent systematic reviews and meta-analyses have shown that treatment with methotrexate, a first-line disease-modifying antirheumatic drug (DMARD), is associated with a significant reduction in atherosclerosis-mediated cardiovascular events, such as myocardial infarction and stroke, and mortality, when compared to other DMARDs. This suggests that methotrexate might exert specific protective effects against vascular inflammation and atherosclerosis in the context of autoimmune rheumatic disease. This review discusses the available evidence regarding the potential antiatherosclerotic effects of methotrexate through the inhibition of TNF-α, IL-1, and IL-6 and provides suggestions for future experimental and human studies addressing this issue.
Collapse
|
49
|
Network pharmacology exploration reveals endothelial inflammation as a common mechanism for stroke and coronary artery disease treatment of Danhong injection. Sci Rep 2017; 7:15427. [PMID: 29133791 PMCID: PMC5684234 DOI: 10.1038/s41598-017-14692-3] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 10/09/2017] [Indexed: 12/17/2022] Open
Abstract
Although Danhong injection (DHI) is the most widely prescribed Chinese medicine for both stroke and coronary artery disease (CAD), its underlying common molecular mechanisms remain unclear. An integrated network pharmacology and experimental verification approach was used to decipher common pharmacological mechanisms of DHI on stroke and CAD treatment. A compound-target-disease & function-pathway network was constructed and analyzed, indicating that 37 ingredients derived from DH (Salvia miltiorrhiza Bge., Flos Carthami tinctorii and DHI) modulated 68 common targets shared by stroke and CAD. In-depth network analysis results of the top diseases, functions, pathways and upstream regulators implied that a common underlying mechanism linking DHI’s role in stroke and CAD treatment was inflammatory response in the process of atherosclerosis. Experimentally, DHI exerted comprehensive anti-inflammatory effects on LPS, ox-LDL or cholesterol crystal-induced NF-κB, c-jun and p38 activation, as well as IL-1β, TNF-α, and IL-10 secretion in vascular endothelial cells. Ten of 14 predicted ingredients were verified to have significant anti-inflammatory activities on LPS-induced endothelial inflammation. DHI exerts pharmacological efficacies on both stroke and CAD through multi-ingredient, multi-target, multi-function and multi-pathway mode. Anti-endothelial inflammation therapy serves as a common underlying mechanism. This study provides a new understanding of DHI in clinical application on cardiovascular and cerebrovascular diseases.
Collapse
|
50
|
VLDL Induced Modulation of Nitric Oxide Signalling and Cell Redox Homeostasis in HUVEC. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:2697364. [PMID: 29085553 PMCID: PMC5632467 DOI: 10.1155/2017/2697364] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/31/2017] [Accepted: 08/15/2017] [Indexed: 12/25/2022]
Abstract
High levels of circulating lipoprotein constitute a risk factor for cardiovascular diseases, and in this context, the specific role of the very-low-density lipoproteins (VLDL) is poorly understood. The response of human umbilical vein endothelial cells (HUVEC) to VLDL exposure was studied, especially focusing on the pathways involved in alteration of redox homeostasis and nitric oxide (NO) bioavailability. The results obtained by the analysis of the expression level of genes implicated in the NO metabolism and oxidative stress response indicated a strong activation of inducible NO synthase (iNOS) upon 24 h exposure to VLDL, particularly if these have been preventively oxidised. Simultaneously, both mRNA and protein expression of endothelial NO synthase (eNOS) were decreased and its phosphorylation pattern, at the key residues Tyr495 and Ser1177, strongly suggested the occurrence of the eNOS uncoupling. The results are consistent with the observed increased production of nitrites and nitrates (NOx), reactive oxygen species (ROS), 3-nitrotyrosine (3-NT), and, at mitochondrial level, a deficit in mitochondrial O2 consumption. Altogether, these data suggest that the VLDL, particularly if oxidised, when allowed to persist in contact with endothelial cells, strongly alter NO bioavailability, affecting redox homeostasis and mitochondrial function.
Collapse
|