1
|
Guha A, Sadeghi SA, Kunhiraman HH, Fang F, Wang Q, Rafieioskouei A, Grumelot S, Gharibi H, Saei AA, Sayadi M, Weintraub NL, Horibata S, Yang PCM, Bonakdarpour B, Ghassemi M, Sun L, Mahmoudi M. AI-Driven Prediction of Cardio-Oncology Biomarkers Through Protein Corona Analysis. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2025; 509:161134. [PMID: 40190726 PMCID: PMC11970620 DOI: 10.1016/j.cej.2025.161134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Protein corona, a layer predominantly composed of proteins and other biomolecules that forms on nanoparticle surfaces upon interaction with biological fluids, has recently been extensively utilized to enhance the depth of plasma proteomics and biomarker discovery. In this study, we integrate protein corona profiling with mass spectrometry (MS)-based bottom-up proteomics (BUP), machine learning, and causality analysis to identify potential biomarkers in the field of cardio-oncology. We selected prostate cancer (PC) and atherosclerosis as model cardio-oncology diseases, given that PC is the most prevalent cancer among men in the United States and frequently coexists with atherosclerotic cardiovascular disease (ASCVD), which contributes to the progression of metastatic PC (mPC). Protein corona profiles were generated from 35 plasma samples categorized into four groups: mPC with ASCVD, nonmetastatic PC (nmPC) with ASCVD, mPC without ASCVD, and nmPC without ASCVD. MS-based BUP analysis identified 887 unique proteins within the protein corona. Gene Ontology (GO) analysis of the 260 proteins common to all samples revealed key plasma proteomic pathways significantly associated with ASCVD and mPC. Using Least Absolute Shrinkage and Selection Operator (LASSO) regularization, we isolated 22 proteins strongly associated with ASCVD or mPC, including chaperonin containing TCP1 subunit 7 (CCT7), which was common to both conditions. Automated formal reasoning and causality analysis of these 22 proteins identified thromboxane-A synthase 1 (TBXAS1) as a primary causal factor linked to both ASCVD and mPC. TBXAS1 plays a critical role in promoting platelet aggregation, vascular smooth muscle cell proliferation, endothelial dysfunction, and thrombosis. In this proof-of-concept study, CCT7 and TBXAS1 emerged as potential biomarkers for both ASCVD and mPC, suggesting their utility as dual biomarkers for early detection and targeted therapeutic interventions. By combining nanomedicine with advanced analytical methods, our integrated approach provides a robust framework for uncovering causal relationships between biomarkers and disease states.
Collapse
Affiliation(s)
- Avirup Guha
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
- Cardio-Oncology Program, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | | | - Harikrishnan Hyma Kunhiraman
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
- Cardio-Oncology Program, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Fei Fang
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
| | - Qianyi Wang
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
| | - Arshia Rafieioskouei
- Department of Computer Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Shaun Grumelot
- Precision Health Program, Michigan State University, East Lansing, MI, United States
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| | - Hassan Gharibi
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Amir Ata Saei
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Maryam Sayadi
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, United States
| | - Neal L. Weintraub
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Sachi Horibata
- Precision Health Program, Michigan State University, East Lansing, MI, United States
- Cell and Molecular Biology Program, Michigan State University, East Lansing, MI, United States
- Department of Pharmacology and Toxicology, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| | - Phillip Chung-Ming Yang
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, United States
| | - Borzoo Bonakdarpour
- Department of Computer Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Mohammad Ghassemi
- Department of Computer Science and Engineering, Michigan State University, East Lansing, MI, United States
| | - Liangliang Sun
- Department of Chemistry, Michigan State University, East Lansing, MI, United States
| | - Morteza Mahmoudi
- Precision Health Program, Michigan State University, East Lansing, MI, United States
- Department of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
2
|
Qiu X, Feng Y. Echinacoside activates Nrf2/PPARγ signaling pathway to modulate mitochondrial fusion-fission balance to ameliorate ox-LDL-induced dysfunction of coronary artery endothelial cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:9767-9776. [PMID: 38916831 DOI: 10.1007/s00210-024-03233-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/08/2024] [Indexed: 06/26/2024]
Abstract
As a cardiovascular disease, coronary heart disease (CHD) is characterized by poor prognosis and increasing morbidity and mortality rates. Echinacoside (ECH) can protect against multiple cardiovascular diseases due to its antioxidant and anti-inflammatory properties. However, the role of ECH in CHD remains unclear. In ECH-treated human coronary artery endothelial cells (HCAECs), cell viability, NO production, endothelial nitric oxide synthase (eNOS) expression, and angiogenesis ability were detected using cell counting kit-8 (CCK-8) assay, diaminofluorescein-FM diacetate (DAF-FM DA) staining, western blot, and tube formation assay, respectively. The activities of oxidative stress markers were detected using dichloro-dihydro-fluorescein diacetate (DCFH-DA) assay and corresponding assay kits. Cell apoptosis was detected utilizing flow cytometry and caspase3 assay. Western blot was used to detect the expressions of Nrf2/PPARγ signaling pathway- and mitochondrial dynamics-related proteins. Mitochondrial membrane potential and mitochondrial fusion and fission were detected using JC-1 staining and immunofluorescence (IF) assay. In this study, ECH was found to revive the viability, ameliorate the endothelial dysfunction, suppress oxidative stress, and inhibit the apoptosis in ox-LDL-induced HCAECs via activating Nrf2/PPARγ signaling pathway, which were all abolished following the treatment of Nrf2 inhibitor ML385. It was also identified that ECH regulated mitochondrial fusion-fission balance in ox-LDL-induced HCAECs through the activation of Nrf2/PPARγ signaling pathway. In summary, ECH activated Nrf2/PPARγ signaling pathway to regulate mitochondrial fusion-fission balance, thereby improving ox-LDL-induced dysfunction of HCAECs.
Collapse
Affiliation(s)
- Xiandi Qiu
- Department of Cardiovascular Medicine, The Ninth People's Hospital of Chongqing, Chongqing, China
| | - Yuxing Feng
- Department of Neurology, The Ninth People's Hospital of Chongqing, No. 69 Jialing Village, Beibei District, Chongqing, 400700, China.
| |
Collapse
|
3
|
Liu S, Li Y, Wu C. Paeoniflorin suppresses the apoptosis and inflammation of human coronary artery endothelial cells induced by oxidized low-density lipoprotein by regulating the Wnt/β-catenin pathway. PHARMACEUTICAL BIOLOGY 2023; 61:1454-1461. [PMID: 37674320 PMCID: PMC10486282 DOI: 10.1080/13880209.2023.2220360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 04/24/2023] [Accepted: 05/27/2023] [Indexed: 09/08/2023]
Abstract
CONTEXT Paeoniflorin (PF) contributes to improving coronary artery disease (CAD). OBJECTIVE This study clarified the efficiency of PF in CAD and the molecular mechanism. MATERIALS AND METHODS Human coronary artery endothelial cells (HCAECs) were treated with oxidized low-density lipoprotein (ox-LDL; 20, 40, 80 and 160 μg/mL) and PF (0.05, 0.1 0.2 and 0.4 mM). To study cell phenotypes, HCAECs were treated with 80 μg/mL ox-LDL with or without 0.1 mM PF for 24 h, and cell viability and apoptosis were evaluated using the methyl thiazolyl tetrazolium (MTT) assay and flow cytometry, respectively. In addition, inflammatory cytokines levels were measured by enzyme-linked immunosorbent assay (ELISA). Western blot evaluated the Wnt/β-catenin pathway-related factors. RESULTS ox-LDL and PF (0.2 and 0.4 mM) suppressed cell viability in a dose-dependent manner. The IC50 value of PF was 722.9 nM. PF facilitated cell viability (115.76%), inhibited apoptosis (46.28%), reduced IL-6 (63.43%) and IL-8 (66.70%) levels and increased IL-10 levels (181.15%) of ox-LDL-treated HCAECs. Additionally, PF inactivated the Wnt/β-catenin pathway, and XAV939 treatment further promoted cell viability (120.54%), suppressed apoptosis (56.92%), reduced the levels of IL-6 (76.16%) and IL-8 (86.82%) and increased the IL-10 levels (120.22%) of ox-LDL-induced HCAECs after PF treatment. Moreover, PF alleviated plaque lesions of the aorta and aorta root and serum lipid of ApoE-/- mice with a high-fat diet. DISCUSSION AND CONCLUSIONS This study first revealed that PF inhibited ox-LDL-induced HCAECs apoptosis and inflammation via the Wnt/β-catenin pathway and alleviated CAD, suggesting the potential of PF as a drug for CAD treatment.
Collapse
Affiliation(s)
- Shasha Liu
- Department of Geriatrics, Sichuan People’s Hospital, Sichuan Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Ying Li
- Department of Geriatrics, Sichuan People’s Hospital, Sichuan Academy of Medical Sciences, Chengdu, Sichuan, China
| | - Caojie Wu
- Department of Geriatrics, Sichuan People’s Hospital, Sichuan Academy of Medical Sciences, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Yang Y, Wang Z, Xu Y, Liu X, Sun Y, Li W. Knockdown of lncRNA H19 alleviates ox-LDL-induced HCAECs inflammation and injury by mediating miR-20a-5p/HDAC4 axis. Inflamm Res 2022; 71:1109-1121. [PMID: 35854140 DOI: 10.1007/s00011-022-01604-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: 11/15/2021] [Revised: 04/22/2022] [Accepted: 06/23/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Coronary artery disease (CAD) seriously disturbs the life of people. LncRNA H19 is reported to promote the progression of CAD; Nevertheless, the detailed mechanism by which H19 modulates CAD development is unclear. METHODS Clinical samples of CAD patients were collected, meanwhile we established in vitro and in vivo models of CAD by treating HCAECs with ox-LDL and feeding ApoE-/- mice with high fat diets (HFD). MTT assay was adopted to assess the cell viability. Transwell detection was applied to test the migration, and apoptosis was tested by flow cytometry. The levels of inflammatory cytokines were examined by ELISA. The relation among H19, miR-20a-5p and HDAC4 was explored by dual luciferase reporter and RIP assay. RESULTS H19 and HDAC4 levels were elevated, while miR-20a-5p was reduced in plasma of CAD patients and ox-LDL-treated HCAECs. ox-LDL increased H19 level and induced apoptosis and inflammation in HCAECs, while silencing of H19 rescued this phenomenon. In addition, the level of H19 was negatively correlated with miR-20a-5p, and miR-20a-5p inhibitor restored the effect of H19 silencing on HCAECs function. HDAC4 was the downstream mRNA of miR-20a-5p, and miR-20a-5p upregulation reversed ox-LDL-induced HCAECs injury through targeting HDAC4. Furthermore, H19 silencing significantly alleviated the coronary atherosclerotic plaques and inhibited the inflammatory responses in vivo. CONCLUSIONS We proved that knockdown of H19 alleviated ox-LDL-induced HCAECs injury via miR-20a-5p/HDAC4 axis, which might provide a new tactics against CAD.
Collapse
Affiliation(s)
- Yilin Yang
- Department of Geriatrics, Changsha Third Hospital, No. 176, Laodong West Road, Tianxin District, Changsha, 410004, Hunan Province, People's Republic of China
| | - Zhaofei Wang
- Department of Cardiology, Changsha First Hospital, Changsha, 410010, Hunan Province, People's Republic of China
| | - Ying Xu
- Department of Geriatrics, Changsha Third Hospital, No. 176, Laodong West Road, Tianxin District, Changsha, 410004, Hunan Province, People's Republic of China
| | - Xiaofang Liu
- Department of Geriatrics, Changsha Third Hospital, No. 176, Laodong West Road, Tianxin District, Changsha, 410004, Hunan Province, People's Republic of China
| | - Yehai Sun
- Department of Geriatrics, Changsha Third Hospital, No. 176, Laodong West Road, Tianxin District, Changsha, 410004, Hunan Province, People's Republic of China
| | - Wei Li
- Department of Geriatrics, Changsha Third Hospital, No. 176, Laodong West Road, Tianxin District, Changsha, 410004, Hunan Province, People's Republic of China.
| |
Collapse
|
5
|
Cheng X, Liu Z, Zhang H, Lian Y. Inhibition of LOXL1-AS1 alleviates oxidative low-density lipoprotein induced angiogenesis via downregulation of miR-590-5p mediated KLF6/VEGF signaling pathway. Cell Cycle 2021; 20:1663-1680. [PMID: 34334119 PMCID: PMC8489901 DOI: 10.1080/15384101.2021.1958484] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 07/11/2021] [Accepted: 07/18/2021] [Indexed: 10/20/2022] Open
Abstract
Increasing evidences have confirmed that long non-coding RNA LOXL1-AS1 functions in multiple human diseases. Here, we aim to explore the function and mechanism of LOXL1-AS1 in modulating oxidized low-density lipoprotein (ox-LDL)-induced angiogenesis of endothelial cells (ECs). Presently, we found that LOXL1-AS1 and KLF6 were upregulated in ECs treated by Ox-LDL in a dose- and time-dependent manner while miR-590-5p was downregulated. Overexpression of LOXL1-AS1 aggravated Ox-LDL mediated ECs proliferation and migration, and promoted angiogenesis both in vitro and in vivo. On the contrary, enhancing miR-590-5p or inhibiting LOXL1-AS1 level led to suppressive effects on the proliferation, migration and angiogenesis of ECs. Moreover, LOXL1-AS1 upregulation promoted the expression of vascular endothelial growth factor (VEGF), MMPs (including MMP2, MMP9 and MMP14) and also activated VEGF/VEGFR2/PI3K/Akt/eNOS pathway. Mechanistically, LOXL1-AS1 works as a competitive endogenous RNA (ceRNA) by sponging miR-590-5p, which targeted at the 3'-untranslated region (3'UTR) of KLF6. Additionally, the proliferation, migration and angiogenesis of ECs were elevated following KLF6 upregulation. By detecting the expression of LOXL1-AS1 and miR-590-5p in the serum of healthy donors and atherosclerosis patients, it was found that LOXL1-AS1 was upregulated in atherosclerosis patients (compared with healthy donors) and had a negative relationship with miR-590-5p. Taken together, LOXL1-AS1 promoted Ox-LDL induced angiogenesis via regulating miR-590-5p-modulated KLF6/VEGF signaling pathway. The LOXL1-AS1-miR-590-5p axis exerts a novel role in the progression of atherosclerosis.
Collapse
Affiliation(s)
- Xuan Cheng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou Henan, China
| | - Zhiwei Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou Henan, China
| | - Haifeng Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou Henan, China
| | - Yajun Lian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou Henan, China
| |
Collapse
|
6
|
Cheng X, Liu Z, Zhang H, Lian Y. Inhibition of LOXL1-AS1 alleviates oxidative low-density lipoprotein induced angiogenesis via downregulation of miR-590-5p mediated KLF6/VEGF signaling pathway. Cell Cycle 2021; 20:1-18. [PMID: 34382896 DOI: 10.1080/15384101.2021.1958501] [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: 04/11/2021] [Revised: 07/11/2021] [Accepted: 07/18/2021] [Indexed: 10/20/2022] Open
Abstract
Increasing evidences have confirmed that long non-coding RNA LOXL1-AS1 functions in multiple human diseases. Here, we aim to explore the function and mechanism of LOXL1-AS1 in modulating oxidized low-density lipoprotein (ox-LDL)-induced angiogenesis of endothelial cells (ECs). Presently, we found that LOXL1-AS1 and KLF6 were upregulated in ECs treated by Ox-LDL in a dose- and time-dependent manner while miR-590-5p was downregulated. Overexpression of LOXL1-AS1 aggravated Ox-LDL mediated ECs proliferation and migration, and promoted angiogenesis both in vitro and in vivo. On the contrary, enhancing miR-590-5p or inhibiting LOXL1-AS1 level led to suppressive effects on the proliferation, migration and angiogenesis of ECs. Moreover, LOXL1-AS1 upregulation promoted the expression of vascular endothelial growth factor (VEGF), MMPs (including MMP2, MMP9, and MMP14) and also activated VEGF/VEGFR2/PI3K/Akt/eNOS pathway. Mechanistically, LOXL1-AS1 works as a competitive endogenous RNA (ceRNA) by sponging miR-590-5p, which targeted at the 3'-untranslated region (3'UTR) of KLF6. Additionally, the proliferation, migration, and angiogenesis of ECs were elevated following KLF6 upregulation. By detecting the expression of LOXL1-AS1 and miR-590-5p in the serum of healthy donors and atherosclerosis patients, it was found that LOXL1-AS1 was upregulated in atherosclerosis patients (compared with healthy donors) and had a negative relationship with miR-590-5p. Taken together, LOXL1-AS1 promoted Ox-LDL induced angiogenesis via regulating miR-590-5p-modulated KLF6/VEGF signaling pathway. The LOXL1-AS1-miR-590-5p axis exerts a novel role in the progression of atherosclerosis.
Collapse
Affiliation(s)
- Xuan Cheng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou Henan, China
| | - Zhiwei Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou Henan, China
| | - Haifeng Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou Henan, China
| | - Yajun Lian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou Henan, China
| |
Collapse
|
7
|
Knockdown of circ_0004104 Alleviates Oxidized Low-Density Lipoprotein-Induced Vascular Endothelial Cell Injury by Regulating miR-100/TNFAIP8 Axis. J Cardiovasc Pharmacol 2021; 78:269-279. [PMID: 34554678 DOI: 10.1097/fjc.0000000000001063] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 05/08/2021] [Indexed: 01/12/2023]
Abstract
ABSTRACT Coronary artery disease (CAD) is a common cardiovascular disease, mainly due to vascular endothelial cell (VEC) injury caused by atherosclerosis. Circular RNA has been shown to be involved in the regulation of various diseases. However, the role and mechanism of circ_0004104 in CAD are still unclear. Oxidized low-density lipoprotein (ox-LDL) was used to construct the VEC injury model in vitro. The expression levels of circ_0004104 and miR-100 were measured by quantitative real-time polymerase chain reaction. The proliferation of VECs was determined using 3-(45)-dimethylthiahiazo (-z-y1)-35-di-phenytetrazoliumromide assay and 5-ethynyl-2'-deoxyuridine staining assay. VEC apoptosis rate was assessed using flow cytometry, and caspase-3 activity was measured using a Caspase-3 Assay Kit. The protein expression levels of Ki-67, cleaved-caspase3, and tumor necrosis factor-α-induced protein 8 (TNFAIP8) were detected by western blot analysis. Furthermore, enzyme-linked immunosorbent assay was performed to assess the concentrations of inflammatory cytokines. In addition, the relationship between miR-100 and circ_0004104 or TNFAIP8 was confirmed by dual-luciferase reporter assay and biotin-labeled RNA pull-down assay. Our results revealed that circ_0004104 was upregulated and miR-100 was downregulated in patients with CAD and ox-LDL-induced VECs. Ox-LDL could inhibit the proliferation and promote the apoptosis and inflammation of VECs to induce VEC injury. However, silenced circ_0004104 could alleviate VEC injury induced by ox-LDL. Moreover, we found that circ_0004104 could sponge miR-100 and a miR-100 inhibitor could reverse the inhibition effect of circ_0004104 knockdown on ox-LDL-induced VEC injury. In addition, TNFAIP8 was a target of miR-100, and miR-100 alleviated ox-LDL-induced VEC injury by targeting TNFAIP8. Our data suggested that circ_0004104 promoted ox-LDL-induced VEC injury by the miR-100/TNFAIP8 axis, indicating that circ_0004104 might be a potential biomarker for CAD treatment.
Collapse
|
8
|
Chiang HY, Chu PH, Chen SC, Lee TH. MFG-E8 Regulates Vascular Smooth Muscle Cell Migration Through Dose-Dependent Mediation of Actin Polymerization. J Am Heart Assoc 2021; 10:e020870. [PMID: 34041925 PMCID: PMC8483510 DOI: 10.1161/jaha.121.020870] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Migration of vascular smooth muscle cells (VSMCs) is the main contributor to neointimal formation. The Arp2/3 (actin-related proteins 2 and 3) complex activates actin polymerization and is involved in lamellipodia formation during VSMC migration. Milk fat globule-epidermal growth factor 8 (MFG-E8) is a glycoprotein expressed in VSMCs. We hypothesized that MFG-E8 regulates VSMC migration through modulation of Arp2/3-mediated actin polymerization. Methods and Results To determine whether MFG-E8 is essential for VSMC migration, a model of neointimal hyperplasia was induced in the common carotid artery of wild-type and MFG-E8 knockout mice, and the extent of neointimal formation was evaluated. Genetic deletion of MFG-E8 in mice attenuated injury-induced neointimal hyperplasia. Cultured VSMCs deficient in MFG-E8 exhibited decreased cell migration. Immunofluorescence and immunoblotting revealed decreased Arp2 but not Arp3 expression in the common carotid arteries and VSMCs deficient in MFG-E8. Exogenous administration of recombinant MFG-E8 biphasically and dose-dependently regulated the cultured VSMCs. At a low concentration, MFG-E8 upregulated Arp2 expression. By contrast, MFG-E8 at a high concentration reduced the Arp2 level and significantly attenuated actin assembly. Arp2 upregulation mediated by low-dose MFG-E8 was abolished by treating cultured VSMCs with β1 integrin function-blocking antibody and Rac1 inhibitors. Moreover, treatment of the artery with a high dose of recombinant MFG-E8 diminished injury-induced neointimal hyperplasia and reduced VSMC migration. Conclusions MFG-E8 plays a critical role in VSMC migration through dose-dependent regulation of Arp2-mediated actin polymerization. These findings suggest that high doses of MFG-E8 may have therapeutic potential for treating vascular occlusive diseases.
Collapse
Affiliation(s)
- Hou-Yu Chiang
- Department of Anatomy College of Medicine Chang Gung University Taoyuan Taiwan.,Graduate Institute of Biomedical Sciences College of Medicine Chang Gung University Taoyuan Taiwan.,Division of Cardiology Department of Internal Medicine Chang Gung Memorial Hospital Linkou Taiwan
| | - Pao-Hsien Chu
- Division of Cardiology Department of Internal Medicine Chang Gung Memorial Hospital Linkou Taiwan.,College of Medicine Chang Gung University Taoyuan Taiwan
| | - Shao-Chi Chen
- Department of Anatomy College of Medicine Chang Gung University Taoyuan Taiwan
| | - Ting-Hein Lee
- Department of Anatomy College of Medicine Chang Gung University Taoyuan Taiwan.,Graduate Institute of Biomedical Sciences College of Medicine Chang Gung University Taoyuan Taiwan.,Division of Cardiology Department of Internal Medicine Chang Gung Memorial Hospital Linkou Taiwan
| |
Collapse
|
9
|
Li Y, Huang J, Yan H, Li X, Ding C, Wang Q, Lu Z. Protective effect of microRNA‑381 against inflammatory damage of endothelial cells during coronary heart disease by targeting CXCR4. Mol Med Rep 2020; 21:1439-1448. [PMID: 32016478 PMCID: PMC7003055 DOI: 10.3892/mmr.2020.10957] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 06/19/2019] [Indexed: 12/12/2022] Open
Abstract
Coronary heart disease (CHD) is the leading cause of human morbidity and mortality worldwide. MicroRNA (miRNA) profiling is an innovative method of identifying biomarkers for many diseases and may be a powerful tool in the diagnosis and treatment of CHD. The present study aimed to analyze the effects of miRNA (miR)‑381 on the inflammatory damage of endothelial cells during CHD. A total of 21 patients with CHD and 21 healthy control patients were enrolled in this study. Reverse transcription‑quantitative PCR, western blotting and immunofluorescence assays were conducted to examine the expression levels of miR‑381, C‑X‑C chemokine receptor type 4 (CXCR4), Bcl‑2, Bax, Cleaved‑Caspases‑3 and ‑9, p38, ERK1/2 and JNK. Cell Counting Kit‑8, EdU and flow cytometry experiments were performed to evaluate cell proliferation and apoptosis. An ELISA was adopted to determine the expressions of inflammatory factors (interleukins‑8, ‑6 and ‑1β, and tumor necrosis factor‑α). In addition, a dual‑luciferase reporter assay was used to determine the relationship between miR‑381 and CXCR4. Decreased miR‑381 expression and increased CXCR4 expression in the plasma were observed in the CHD group compared with the normal group, which indicated a negative relationship between miR‑381 and CXCR4. Overexpression of miR‑381 significantly promoted the proliferation and inhibited the apoptosis of oxidized low‑density lipoprotein (OX‑LDL)‑induced human umbilical vein endothelial cells (HUVECs) through mitogen‑activated protein kinase pathway by targeting and inhibiting CXCR4. Furthermore, overexpression of miR‑381 reduced the release of inflammatory factors in OX‑LDL‑induced HUVECs. By contrast, reduced expression of miR‑381 exerted the opposite effects, which were subsequently reversed by silencing CXCR4 expression. Results from the present study indicated that miR‑381 was a CHD‑related factor that may serve as a potential molecular target for CHD treatment.
Collapse
Affiliation(s)
- Yimin Li
- Department of Cardiology, Nanjing Chest Hospital, Nanjing, Jiangsu 210029, P.R. China
| | - Jin Huang
- Department of Cardiology, Nanjing Chest Hospital, Nanjing, Jiangsu 210029, P.R. China
| | - Hong Yan
- Inspection Center, Nanjing Chest Hospital, Nanjing, Jiangsu 210029, P.R. China
| | - Xiangyu Li
- Department of Cardiology, Nanjing Chest Hospital, Nanjing, Jiangsu 210029, P.R. China
| | - Chang Ding
- Department of Cardiology, Nanjing Chest Hospital, Nanjing, Jiangsu 210029, P.R. China
| | - Qian Wang
- Department of Cardiology, Nanjing Chest Hospital, Nanjing, Jiangsu 210029, P.R. China
| | - Zhiping Lu
- Department of Cardiology, Nanjing Chest Hospital, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
10
|
Zhang Q, Liu C, Li Q, Li J, Wu Y, Liu J. MicroRNA-25-5p counteracts oxidized LDL-induced pathological changes by targeting neuronal growth regulator 1 (NEGR1) in human brain micro-vessel endothelial cells. Biochimie 2019; 165:141-149. [PMID: 31365884 DOI: 10.1016/j.biochi.2019.07.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 07/26/2019] [Indexed: 12/13/2022]
Abstract
MicroRNA-25-5p (miR-25-5p) may be involved in the pathogenesis and processes of vascular diseases. The aim of this study was to investigate the role of miR-25-5p in oxidized low-density lipoprotein (ox-LDL)-treated human brain microvessel endothelial cells (HBMECs) and the underlying mechanisms. RT-qPCR and/or Western blot were used to detect the expression levels of miR-25-5p and neuronal growth regulator 1 (NEGR1). The effect of miR-25-5p overexpression and NEGR1 silencing on cell proliferation, migration, apoptosis and reactive oxygen species (ROS) production of HBMECs were measured by using CCK-8 assay, transwell assay and flow cytometry, respectively. The expression levels of apoptosis-related protein (cleaved caspase-3 and pro-caspase-3) were detected using Western blot, and the nitric oxide (NO) production was measured by a nitric oxide assay kit. The expression level of miR-25-5p was decreased in HBMECs treated with ox-LDL. Compared with the control group, miR-25-5p overexpression significantly promoted the proliferation and migration of HBMECs treated with ox-LDL (p < 0.01). Overexpression of miR-25-5p significantly suppressed cell apoptosis, ROS production and NO reduction of ox-LDL-induced HBMECs (p < 0.01). In addition, the target gene of miR-25-5p was predicted to be NEGR1 through Targetscan online analysis. The effect of NEGR1 silencing on cell proliferation, migration, apoptosis, ROS and NO production of ox-LDL-induced HBMECs was similar to that of miR-25-5p overexpression. Furthermore, miR-25-5p overexpression and NEGR1 silencing significantly downregulated the protein expression levels of JAK2 and STAT3. Thus, miR-25-5p neutralizes the effects of ox-LDL on multiple functions of HBMECs through suppressing the expression of NEGR1 via regulating the JAK/STA signaling pathway.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Cerebrovascular Diseases, Blue Cross Brain Hospital Affiliated to Tongji University, Shanghai, 201101, China.
| | - Chun Liu
- Department of Cerebrovascular Diseases, Blue Cross Brain Hospital Affiliated to Tongji University, Shanghai, 201101, China
| | - Qiang Li
- Department of Neurosurgery, Changhai Hospital of Shanghai Affiliated to Naval Military Medical University, Shanghai, 200433, China
| | - Jianan Li
- Department of Neurosurgery, Changhai Hospital of Shanghai Affiliated to Naval Military Medical University, Shanghai, 200433, China
| | - Yina Wu
- Department of Neurosurgery, Changhai Hospital of Shanghai Affiliated to Naval Military Medical University, Shanghai, 200433, China
| | - Jianmin Liu
- Department of Neurosurgery, Changhai Hospital of Shanghai Affiliated to Naval Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
11
|
Ji G, Song X, Wang L, Li Z, Wu H, Dong H. Golgi apparatus fragmentation participates in oxidized low‐density lipoprotein‐induced endothelial cell injury. J Cell Biochem 2019; 120:18862-18870. [PMID: 31264250 DOI: 10.1002/jcb.29205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 11/11/2022]
Affiliation(s)
- Guang Ji
- Department of NeurologyThe Second Hospital of Hebei Medical University Shijiazhuang People's Republic of China
| | - Xueqin Song
- Department of NeurologyThe Second Hospital of Hebei Medical University Shijiazhuang People's Republic of China
| | - Liang Wang
- Department of NeurologyThe Second Hospital of Hebei Medical University Shijiazhuang People's Republic of China
| | - Zhenfei Li
- Department of NeurologyThe Second Hospital of Hebei Medical University Shijiazhuang People's Republic of China
| | - Hongran Wu
- Department of NeurologyThe Second Hospital of Hebei Medical University Shijiazhuang People's Republic of China
| | - Hui Dong
- Department of NeurologyThe Second Hospital of Hebei Medical University Shijiazhuang People's Republic of China
| |
Collapse
|
12
|
Schnitzler JG, Dallinga-Thie GM, Kroon J. The Role of (Modified) Lipoproteins in Vascular Function: A Duet Between Monocytes and the Endothelium. Curr Med Chem 2019; 26:1594-1609. [PMID: 29546830 DOI: 10.2174/0929867325666180316121015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/05/2017] [Accepted: 12/06/2017] [Indexed: 12/24/2022]
Abstract
Over the last century, many studies have demonstrated that low-density lipoprotein (LDL) is a key risk factor of cardiovascular diseases (CVD) related to atherosclerosis. Thus, for these CVD patients, LDL lowering agents are commonly used in the clinic to reduce the risk for CVD. LDL, upon modification, will develop distinct inflammatory and proatherogenic potential, leading to impaired endothelial integrity, influx of immune cells and subsequent increased foam cell formation. LDL can also directly affect peripheral monocyte composition, rendering them in a more favorable position to migrate and accumulate in the subendothelial space. It has become apparent that other lipoprotein particles, such as triglyceride- rich lipoproteins or remnants (TRL) and lipoprotein(a) [Lp(a)] may also impact on atherogenic pathways. Evidence is accumulating that Lp(a) can promote peripheral monocyte activation, eventually leading to increased transmigration through the endothelium. Similarly, remnant cholesterol has been identified to play a key role in endothelial dysfunction and monocyte behavior. In this review, we will discuss recent developments in understanding the role of different lipoproteins in the context of inflammation at both the level of the monocyte and the endothelium.
Collapse
Affiliation(s)
- Johan G Schnitzler
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Geesje M Dallinga-Thie
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jeffrey Kroon
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Takeda S, Mukasa K, Hui SP, Chiba H. Interaction between LDL-mimetic liposomes and acid-treated carbon nanotube electrode during Cu 2+-mediated oxidation. Biochem Biophys Res Commun 2019; 513:275-279. [PMID: 30954226 DOI: 10.1016/j.bbrc.2019.03.106] [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: 02/26/2019] [Accepted: 03/17/2019] [Indexed: 11/16/2022]
Abstract
Oxidation of low-density lipoproteins (LDL) causes atherosclerosis. Detection of oxidation of LDL-mimetic liposomes using an electrode might serve as a convenient tool in the search of antioxidants for the prevention of atherosclerosis. This report proposes a reaction mechanism between LDL-mimetic liposomes and an acid-treated carbon nanotube (CNT) electrode. Oxidation of the liposomes, mediated by Cu2+, was monitored by the change in electrode potential, and the fluorescence intensity generated by diphenyl-1-pyrenylphosphine (DPPP) as control. The electrode potential and fluorescence intensity increased concomitantly during oxidation, followed by a gradual decrease. Although the electrical potential peaked faster than the fluorescence intensity, addition of CNT to the DPPP reaction accelerated the latter, suggesting the role of CNT as an accelerator of liposome oxidation. Atomic force microscopy showed increased binding of liposomes to CNT along with liposomal deformation. Further, binding of Cu2+ to the liposome-bound CNT surface was observed by quartz crystal microbalance. In conclusion, the interaction of liposomes with Cu2+ and CNT surface explains the rapid response of the electrode in liposome oxidation.
Collapse
Affiliation(s)
- Seiji Takeda
- Faculty of Health Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | | | - Shu-Ping Hui
- Faculty of Health Sciences, Hokkaido University, Sapporo, 060-0812, Japan.
| | - Hitoshi Chiba
- Department of Nutrition, Sapporo University of Health Sciences, Sapporo, 007-0894, Japan
| |
Collapse
|
14
|
Actin related protein 3 (ARP3) promotes apoptosis of intestinal epithelial cells in ulcerative colitis. Pathol Res Pract 2019; 215:235-242. [DOI: 10.1016/j.prp.2018.10.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/15/2018] [Accepted: 10/17/2018] [Indexed: 01/26/2023]
|
15
|
Wang X, Mao R, Chen W. FSD-C10 Shows Therapeutic Effects in Suppressing oxidized low-density lipoprotein (ox-LDL)-Induced Human Brain Microvascular Endothelial Cells Apoptosis via Rho-Associated Coiled-Coil Kinase (ROCK)/Mitogen-Activated Protein Kinase (MAPK) Signaling. Med Sci Monit 2018; 24:5509-5516. [PMID: 30088495 PMCID: PMC6097139 DOI: 10.12659/msm.911481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND ox-LDL-induced injury of brain microvascular endothelial cells (BMECs) is strongly associated with cerebral vascular diseases such as cerebral arterial atherosclerosis. ROCK inhibitor was proved to be anti-apoptotic and has been used in treating cerebral vascular diseases. Research on the neuroprotective effects of a novel ROCK inhibitor, FSD-C10, is still limited. The present study investigated the anti-apoptotic effect and underlying molecular mechanism of FSD-C10 in ox-LDL-mediated apoptosis of BMECs. MATERIAL AND METHODS ox-LDL and/or FSD-C10 were used to incubate immortalized human BMECs. MTT assay was used to assess cell viability. Cell apoptosis was evaluated by TUNEL assay. A colorimetric method was used to assess ROCK activity. Western blot analysis was used to examine the expression and phosphorylation levels of proteins. RESULTS ox-LDL incubation reduced the viability of BMECs by inducing cell apoptosis in a concentration-dependent manner. ROCK activity was also elevated by ox-LDL incubation in BMECs in a concentration-dependent manner. Expression level of Bcl2 was reduced while expression levels of Bax and active caspase3 were increased by ox-LDL treatment in a concentration-dependent manner. ox-LDL also increased the phosphorylation levels of p38, JNK, and ERK1/2 in a concentration-dependent manner. FSD-C10 treatment increased the cell viability by reducing apoptosis of BMECs exposed to ox-LDL. Moreover, FSD-C10 was found to suppress the phosphorylation levels of p38, JNK, and ERK1/2 and the expression levels of Bax and active caspase3 in ox-LDL treated BMECs. CONCLUSIONS FSD-C10 increases cell viability in ox-LDL-treated BMECs by reducing cell apoptosis. ROCK/MAPKs-mediated apoptosis appears to be the underlying molecular mechanism.
Collapse
Affiliation(s)
- Xin Wang
- Department of Neurology, Yiwu Central Hospital, Yiwu, Zhejiang, China (mainland)
| | - Rongyan Mao
- Department of Neurology, Yiwu Central Hospital, Yiwu, Zhejiang, China (mainland)
| | - Weiwei Chen
- Department of Neurology, Yiwu Central Hospital, Yiwu, Zhejiang, China (mainland)
| |
Collapse
|
16
|
Effects of enzymes on elastic modulus of low-density lipoproteins were investigated using atomic force microscopy. Biochem Biophys Res Commun 2018; 501:607-611. [PMID: 29709480 DOI: 10.1016/j.bbrc.2018.04.211] [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: 04/16/2018] [Accepted: 04/26/2018] [Indexed: 11/23/2022]
Abstract
Oxidation of low-density lipoproteins (LDLs) induces development of cardiovascular disease. Recently, reports of studies using atomic force microscopy (AFM) have described that the elastic modulus of metal-induced oxidized LDLs is lower than the modulus before oxidation. However, the mechanisms of change of the elastic modulus have not been well investigated. We postulated that disorder of the LDL structure might decrease the elastic modulus. This study measured the elastic modulus of LDLs before and after enzyme treatment with V8 protease, α-chymotrypsin, and phospholipase A2. After LDLs were obtained from serum by ultracentrifugation, LDLs or enzyme-treated LDLs were physically absorbed. They were crowded on a mica surface. Although V8 protease and α-chymotrypsin did not induce the elastic modulus change, treatment with PLA2 decreased the elastic modulus. The LDL particle size did not change during the enzyme treatment. Results suggest that disordering of the lipid structure of the LDL might contribute to the elastic modulus change. Results show that AFM might be a useful tool to evaluate disorders of complex nanoscale particle structures from lipids and proteins such as lipoproteins.
Collapse
|
17
|
Puerarin inhibits expression of tissue factor induced by oxidative low-density lipoprotein through activating the PI3K/Akt/eNOS pathway and inhibiting activation of ERK1/2 and NF-κB. Life Sci 2017; 191:115-121. [PMID: 29037842 DOI: 10.1016/j.lfs.2017.10.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 10/09/2017] [Accepted: 10/13/2017] [Indexed: 12/21/2022]
Abstract
AIMS The present study aimed to investigate whether puerarin regulated tissue factor (TF) expression induced by oxidative low-density lipoprotein (ox-LDL), an independent risk factor for atherosclerosis, and its mechanisms. MAIN METHODS TF expression at the mRNA level was determined by reverse transcription-quantitative polymerase chain reaction, and its expression at the protein level, as well as other target proteins, was assessed by western blotting. Nitric oxide (NO) production was measured by a nitrate reduction method. KEY FINDINGS Results demonstrated that treatment with ox-LDL (50mg/l) for 24h significantly increased (P<0.01) TF expression at the mRNA and protein levels in human umbilical vein endothelial cells (HUVECs). Such an ox-LDL exposure also triggered the dephosphorylation of Akt, resulting in a reduction of NO production and activated the extracellular signal-regulated kinase (ERK)1/2 and nuclear factor (NF)-κB signaling pathways. Pre-treatment with puerarin (50-200μM) for 1h significantly attenuated the ox-LDL-induced TF expression, augmented the phosphorylation of Akt, with a resultant increase of the NO production, and inhibited the activation of ERK1/2 and NF-κB (P<0.01). However, this beneficial effect of puerarin (100μM) was abolished by LY294002 (10μM), an inhibitor of phosphoinositide 3-kinase (PI3K), or NG-nitro-L-arginine methyl ester (100μM), an inhibitor of NO synthase. SIGNIFICANCE These results suggested that puerarin suppressed TF expression in HUVECs through activating the PI3K/Akt/endothelial nitric oxide synthase signaling pathway and inhibiting the activation of ERK1/2 and NF-κB. These findings suggested that puerarin possessed certain anticoagulation and may be a potential novel therapeutic drug for thrombosis in coronary artery disease.
Collapse
|