1
|
Lu W, Wan G, Zhu H, Zhu T, Zhang X. MiR-497-5p regulates ox-LDL-induced dysfunction in vascular endothelial cells by targeting VEGFA/p38/MAPK pathway in atherosclerosis. Heliyon 2024; 10:e28887. [PMID: 38601630 PMCID: PMC11004747 DOI: 10.1016/j.heliyon.2024.e28887] [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: 11/17/2023] [Revised: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 04/12/2024] Open
Abstract
Background The impairment of endothelial cells triggered by oxidized low-density lipoprotein (ox-LDL) stands as a critical event in the advancement of atherosclerosis (AS). MiR-497-5p has been recognized as a potential predictor for AS, but its precise involvement in ox-LDL-induced endothelial cell dysfunction remains to be elucidated. Methods An in vitro AS cell model was established by exposing human umbilical vein endothelial cells (HUVECs) to 100 μg/mL ox-LDL for 24 h. The assessment of endothelial cell function included evaluating cell viability, caspase-3 activity, inflammatory factors, and oxidative markers. Molecular mechanisms were elucidated through quantitative real-time PCR, Western blot analysis, and luciferase reporter assays. Results Our investigation revealed that exposure to ox-LDL led to an upregulation in miR-497-5p and p-p38 levels, while downregulating the expression of vascular endothelial growth factor A (VEGFA) and phosphorylated (p)-endothelial nitric oxide synthase (p-eNOS) in HUVECs. Ox-LDL exposure resulted in decreased cell viability and angiogenic capacity, coupled with increased apoptosis, inflammation, and oxidative stress in HUVECs, partially mediated by the upregulation of miR-497-5p. We confirmed VEGFA as a direct target of miR-497-5p. Interfering with VEGFA expression significantly reversed the effects mediated by miR-497-5p silencing in HUVECs exposed to ox-LDL. Conclusions In summary, our findings demonstrate that miR-497-5p exacerbates ox-LDL-induced dysfunction in HUVECs through the activation of the p38/MAPK pathway, mediated by the targeting of VEGFA.
Collapse
Affiliation(s)
- Wei Lu
- Department of Cardiovascular Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
| | - Guoqing Wan
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - He Zhu
- Zhejiang Chinese Medical University, Zhejiang, China
| | - Tao Zhu
- Zhejiang Chinese Medical University, Zhejiang, China
| | - Xinmei Zhang
- Department of Cardiovascular Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, China
| |
Collapse
|
2
|
Wu MY, Lee AS, Lin YN, Chung WH, Chen KW, Lu CR, Chen YF, Chang CM, Tsai WC, Shiao YT, Chen CH, Chang KC. Role of low-density lipoprotein electronegativity and sexual dimorphism in contributing early ventricular tachyarrhythmias following ST-elevation myocardial infarction. Front Cardiovasc Med 2024; 11:1285068. [PMID: 38500756 PMCID: PMC10944913 DOI: 10.3389/fcvm.2024.1285068] [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: 08/29/2023] [Accepted: 02/09/2024] [Indexed: 03/20/2024] Open
Abstract
Background Early ventricular tachycardia/fibrillation (VT/VF) in patients with ST-elevation myocardial infarction (STEMI) has higher morbidity and mortality. This study examines gender-differentiated risk factors and underlying mechanisms for early onset VT/VF in STEMI. Methods We analyzed data from 2,964 consecutive STEMI patients between January 1, 2008 and December 31, 2021. Early VT/VF was defined as occurrence of spontaneous VT/VF of ≥30 s or requirement of immediate cardioversion/defibrillation within the first 48 h after symptoms. An ex vivo ischemic-reperfusion experiments were conducted in 8-week-old ApoE-/- mice fed a high-fat diet to explore the underlying mechanisms of early VT/VF. Results In 255 of out 2,964 STEMI patients who experienced early VT/VF, the age was younger (58.6 ± 13.8 vs. 61.0 ± 13.0 years old, P = 0.008) with a male predominance. The plasma levels of L5, the most electronegative subclass of low-density lipoprotein, was higher in early VT/VF patients compared to those without early VT/VF (n = 21, L5: 14.1 ± 22.6% vs. n = 46, L5: 4.3 ± 9.9%, P = 0.016). In the experimental setup, all male mice (n = 4) developed VT/VF post sham operation, whereas no such incidence was observed in the female mice (n = 3). Significantly, male mice exhibited considerably slower cardiac conduction velocity as compared to their female counterparts in whole heart preparations (25.01 ± 0.93 cm/s vs.42.32 ± 5.70 cm/s, P < 0.001), despite analogous action potential durations. Furthermore, isolated ventricular myocytes from male mice showed a distinctly lower sodium current density (-29.20 ± 3.04 pA/pF, n = 6) in comparison to female mice (-114.05 ± 6.41 pA/pF, n = 6, P < 0.001). This decreased sodium current density was paralleled by a reduced membrane expression of Nav1.5 protein (0.38 ± 0.06 vs. 0.89 ± 0.09 A.U., P < 0.001) and increased cytosolic Nav1.5 levels (0.59 ± 0.06 vs. 0.29 ± 0.04 A.U., P = 0.001) in male mice. Furthermore, it was observed that the overall expressions of sorting nexin 27 (SNX27) and vacuolar protein sorting 26 (VPS26) were significantly diminished in male mice as compared to female littermates (0.91 ± 0.15 vs. 1.70 ± 0.28, P = 0.02 and 0.74 ± 0.09 vs. 1.57 ± 0.13, P < 0.01, respectively). Conclusions Our findings reveal that male STEMI patients with early VT/VF are associated with elevated L5 levels. The gender-based discrepancy in early VT/VF predisposition might be due to compromised sodium channel trafficking, possibly linked with increased LDL electronegativity.
Collapse
Affiliation(s)
- Mei-Yao Wu
- School of Post-Baccalaureate Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
| | - An-Sheng Lee
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Yen-Nien Lin
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Wei-Hsin Chung
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Ke-Wei Chen
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Chiung-Ray Lu
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Yun-Fang Chen
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Chia-Ming Chang
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Wei-Chung Tsai
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Tzone Shiao
- Center of Institutional Research and Development, Asia University, Taichung, Taiwan
| | - Chu-Huang Chen
- Vascular and Medicinal Research, Texas Heart Institute, Houston, TX, United States
- Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Kuan-Cheng Chang
- Division of Cardiovascular Medicine, Department of Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
3
|
Li JZ, Zhou XX, Wu WY, Qiang HF, Xiao GS, Wang Y, Li G. Concanavalin A promotes angiogenesis and proliferation in endothelial cells through the Akt/ERK/Cyclin D1 axis. PHARMACEUTICAL BIOLOGY 2022; 60:65-74. [PMID: 34913414 PMCID: PMC8725916 DOI: 10.1080/13880209.2021.2013259] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
CONTEXT Concanavalin A (Con A) exhibited multiple roles in cancer cells. However, the role of Con A in endothelial cells was not reported. OBJECTIVE Our present study investigated the potential angiogenic role of Con A in endothelial cells and ischaemic hind-limb mice. MATERIALS AND METHODS Human umbilical vein endothelial cells and Ea.hy926 cells were employed to determine the effect of Con A (0.3, 1, and 3 μg/mL) or vehicle on angiogenesis and cell proliferation with tube formation, ELISA, flow cytometry, EdU, and western blot. Hind-limb ischaemic mice were conducted to determine the pro-angiogenic effect of Con A (10 mg/kg) for 7 days. RESULTS Con A promoted tube formation to about three-fold higher than the control group and increased the secretion of VEGFa, PDGFaa, and bFGF in the medium. The cell viability was promoted to 1.3-fold by Con A 3 μg/mL, and cell cycle progression of G0G1 phase was decreased from 77% in the vehicle group to 70% in Con A 3 μg/mL, G2M was promoted from 15 to 19%, and S-phase was from 7 to 10%. Con A significantly stimulated phosphorylation of Akt and ERK1/2 and expression of cyclin D1 and decreased the expression of p27. These effects of Con A were antagonised by the PI3K inhibitor LY294002 (10 μM) and MEK pathway antagonist PD98059 (10 μM). Moreover, Con A (10 mg/kg) exhibited a repair effect in ischaemic hind-limb mice. DISCUSSION AND CONCLUSIONS This study will provide a new option for treating ischaemic disease by local injection with Con A.
Collapse
Affiliation(s)
- Jing-Zhou Li
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China
| | - Xiao-Xia Zhou
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China
| | - Wei-Yin Wu
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China
| | - Hai-Feng Qiang
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China
| | - Guo-Sheng Xiao
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China
| | - Yan Wang
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China
| | - Gang Li
- Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China
- CONTACT Gang Li ; Xiamen Cardiovascular Hospital, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
4
|
Li H, Song D, Liu Q, Li L, Sun X, Guo J, Li D, Li P. miR-351 promotes atherosclerosis in diabetes by inhibiting the ITGB3/PIK3R1/Akt pathway and induces endothelial cell injury and lipid accumulation. Mol Med 2022; 28:120. [PMID: 36180828 PMCID: PMC9523959 DOI: 10.1186/s10020-022-00547-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 09/14/2022] [Indexed: 12/01/2022] Open
Abstract
Background The miR-351 gene is significantly upregulated in diabetic mice with atherosclerosis. However, the mechanism by which its presence is important for the overall disease has not been elucidated. Therefore, this study will investigate the mechanism of miR-351 in the process of diabetes mellitus with atherosclerosis through miR-351 gene knockout mice. Methods In this study, miR-351−/− C57BL/6 mice were first induced to form a type 2 diabetes mellitus model with atherosclerosis by STZ injection and a high-fat diet. Pathological tests (oil red O, HE, and Masson staining) combined with biochemical indices (TC, TG, LDL-C, HDL-C, TNF-α, hs-CRP, NO, SOD, MDA, CAT, and GSH-Px) were performed to evaluate the pathological degree of atherosclerosis in each group. Mouse aortic endothelial cells were treated with oxidized low-density lipoprotein (ox-LDL) and 30 mM glucose to establish a diabetic atherosclerosis cell model. Combined with cell oil red O staining and flow cytometry, the effects of silencing miR-351 on lipid accumulation and cell apoptosis in the diabetic atherosclerosis cell model were determined. Fluorescence in situ hybridization was used to detect the localization and transcription levels of miR-351 in cells. The target genes of miR-351 were predicted by bioinformatics and verified by dual-luciferase activity reporting. Western blotting was used to detect the expression levels of phosphorylated inosine 3-kinase regulatory subunit 1 (PIK3R1)/serine/threonine kinase 1 (Akt) and apoptosis-related proteins after transfection with integrin subunit β3 (ITGB3) small interfering ribonucleic acid (siRNA). Results The expression of the miR-351 gene was significantly increased in the high-fat wild-type (HWT) group, and its expression was significantly decreased in the knockout mice. Silencing miR-351 effectively alleviated atherosclerosis in mice. The levels of miR-351 expression, apoptosis, lipid accumulation, and oxidative stress in ox-LDL + high glucose-induced endothelial cells were significantly increased. These phenomena were effectively inhibited in lentivirus-infected miR-351-silenced cell lines. Bioinformatics predicted that miR-351-5p could directly target the ITGB3 gene. Transfection of ITGB3 siRNA reversed the downregulation of apoptosis, decreased oil accumulation, and decreased oxidative stress levels induced by miR-351 silencing. In addition, it inhibited the activation of the PIK3R1/Akt pathway. Conclusion Silencing miR-351 upregulates ITGB3 and activates the PIK3R1/Akt pathway, thereby exerting anti-apoptosis and protective effects on endothelial cells. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00547-9.
Collapse
Affiliation(s)
- Hong Li
- Department of Cardiovascular, Shengjing Hospital of China Medical University, Shenyang, China
| | - Dan Song
- School of Pharmacy, China Medical University, Shenyang, China
| | - Qihui Liu
- School of Pharmacy, China Medical University, Shenyang, China
| | - Linlin Li
- Shenyang Open University, Shenyang, China
| | - Xiaoshi Sun
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Jiamei Guo
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Dianlian Li
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Ping Li
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
5
|
Understanding Accelerated Atherosclerosis in Systemic Lupus Erythematosus: Toward Better Treatment and Prevention. Inflammation 2021; 44:1663-1682. [PMID: 33821395 DOI: 10.1007/s10753-021-01455-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/01/2021] [Accepted: 03/18/2021] [Indexed: 12/13/2022]
Abstract
Systemic lupus erythematosus (SLE) carries a significant risk of cardiovascular disease (CVD). The prevalence of premature CVD is especially noteworthy because it occurs in premenopausal women with SLE who would otherwise have very low rates of CVD. While traditional risk factors likely play a role in development of CVD in the setting of SLE, they do not fully explain the excess risk. The pathogenesis of CVD in SLE is not fully understood, but the inflammatory nature of SLE is believed to be a key factor in accelerating atherosclerosis. Systemic inflammation may lead to an abnormal lipid profile with elevated triglycerides, total cholesterol, and low-density lipoprotein cholesterol and dysfunctional high-density lipoprotein cholesterol. Additionally, the inflammatory milieu of SLE plasma promotes endothelial dysfunction and vascular injury, early steps in the progression of atherosclerotic CVD. Despite the overall headway that has been achieved in treating lupus, innovative therapeutics specifically targeting the progression of atherosclerosis within the lupus population are currently lacking. However, there have been advancements in the development of promising modalities for diagnosis of subclinical atherosclerosis and detection of high CVD risk patients. Due to the significant impact of CVD on morbidity and mortality, research addressing prevention and treatment of CVD in SLE needs to be prioritized. This review explores the intricate interplay of SLE-specific properties that contribute to atherosclerosis and CVD within this population, as well as screening methods and possible therapies.
Collapse
|
6
|
DEMİREL S, ŞAHİNTÜRK S, ÖZYENER F. İrisin ve Vasküler Kontraktilite Üzerine Etkileri. SDÜ TIP FAKÜLTESI DERGISI 2021; 28:163-170. [DOI: 10.17343/sdutfd.718412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Abstract
Bu derleme, irisin hakkındaki mevcut bilgileri ve irisinin vasküler tonusa aracılık etmedeki etkin rolünü özetleme çabasıdır. Egzersiz kronik, bulaşıcı olmayan hastalıkların, tip 2 diyabetin ve özellikle kardiyovasküler hastalıkların önlenmesinde bir dayanak noktasıdır. Egzersiz/fiziksel aktiviteye yanıt olarak üretilen yeni bir miyokin olan irisin, beyaz yağ dokusunun ‘esmerleşmesini’ teşvik ederek enerji harcamalarında artışa neden olur. Bu hormonun ilk tanımında, öncül fibronektin tip III alan içeren protein 5’ten ayrılan irisinin dolaşımdaki yüksek seviyeleri, insülin direncinin azalmasıyla düzelmiş glikoz homeostazı ile ilişkilendirilmiştir. İrisinin insanda farklı hedef doku veya organlar üzerindeki etkileri, sağlığın desteklenmesi veya çeşitli metabolik hastalıkların düzenlenmesinde fizyolojik işlevlerini ortaya çıkarmıştır. İrisinin işlevinin anlaşılmasının birçok hastalık ve gelişiminin anlaşılmasında anahtar olabileceğine inanılmaktadır. İrisinin metabolik düzenleme, enerji harcaması ve glikoz homeostazında kilit bir rol oynadığı gösterilmiştir. Hem hayvanlarda hem de insanlarda yapılan çeşitli çalışmalardan elde edilen yeni bulgular, irisinin vasküler aktiviteyi modüle etmek gibi başka olumlu etkilere de sahip olabileceğini ve böylece egzersiz kaynaklı birçok sağlık yararına aracılık ettiğini göstermektedir. Deneysel bulgular, irisinin hipertansiyon gibi anormal vazokonstriksiyona bağlı hastalıkların tedavisinde yararlı bir ajan olabileceğini düşündürmektedir. Bununla birlikte, irisinin rolü ve işlevi hakkındaki veriler tartışmaya yol açmıştır. Hipertansiyon tedavisinde irisinin etkinliğini belirlemek için daha ayrıntılı mekanizma çalışmaları ve in vivo çalışmalar gereklidir. İrisinin etki mekanizmalarının anlaşılmasındaki bir başka güçlü sınırlama ise, insanlarda ve hayvanlarda bugüne kadar halen tanımlanamayan irisin reseptörü hakkındaki bilgi eksikliğidir.
Collapse
|
7
|
Roldán Gallardo FF, Quintar AA. The pathological growth of the prostate gland in atherogenic contexts. Exp Gerontol 2021; 148:111304. [PMID: 33676974 DOI: 10.1016/j.exger.2021.111304] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/26/2021] [Accepted: 02/27/2021] [Indexed: 02/06/2023]
Abstract
The human prostate is an androgen-dependent gland where an imbalance in cell proliferation can lead to benign prostatic hyperplasia (BPH), which results in voiding lower urinary tract symptoms in the elderly. In the last decades, novel evidence has suggested that BPH might represent an element into the wide spectrum of disorders conforming the Metabolic Syndrome (MS). The dyslipidemic state and the other atherogenic factors of the MS have been shown to induce, maintain and/or aggravate the pathological growth of different organs, with data regarding the prostate being still limited. We here review the available epidemiological and experimental studies about the association of BPH with dyslipidemias. In particular, we have focused on Oxidized Low-Density Lipoproteins (OxLDL) as a potential trigger for vascular disease and cellular proliferation in atherogenic contexts, analyzing their putative molecular mechanisms, including the induction of specific extracellular vesicles (EVs)-derived miRNAs. In addition to the epidemiological evidence, OxLDL is proposed to play a fundamental role in the upregulation of prostatic cell proliferation by activating the Rho/Akt/p27Kip1 pathway in atherogenic contexts. miR-21, miR-141, miR-143, miR-145, miR-155, and miR-221 would be involved in the transcription of genes related to the proliferative process. Although much remains to be investigated regarding the impact of OxLDL, its receptors, and molecular mechanisms on the prostate, it is clear that EVs and miRNAs represent a promising target for proliferative pathologies of the prostate gland.
Collapse
Affiliation(s)
- Franco F Roldán Gallardo
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Centro de Microscopía Electrónica, Córdoba, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud (INICSA), Córdoba, Argentina
| | - Amado A Quintar
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Centro de Microscopía Electrónica, Córdoba, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Investigaciones en Ciencias de la Salud (INICSA), Córdoba, Argentina.
| |
Collapse
|
8
|
Human cytomegalovirus promoting endothelial cell proliferation by targeting regulator of G-protein signaling 5 hypermethylation and downregulation. Sci Rep 2020; 10:2252. [PMID: 32041970 PMCID: PMC7010708 DOI: 10.1038/s41598-020-58680-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 01/15/2020] [Indexed: 01/21/2023] Open
Abstract
Interactions between human cytomegalovirus (HCMV) infection and environmental factors can increase susceptibility to essential hypertension (EH). Although endothelial dysfunction is the initial factor of EH, the epigenetic mechanisms through which HCMV infection induces endothelial cell dysfunction are poorly understood. Here, we evaluated whether HCMV regulated endothelial cell function and assessed the underlying mechanisms. Microarray analysis in human umbilical vein endothelial cells (HUVECs) treated with HCMV AD169 strain in the presence of hyperglycemia and hyperlipidemia revealed differential expression of genes involved in hypertension. Further analyses validated that the regulator of G-protein signaling 5 (RGS5) gene was downregulated in infected HUVECs and showed that HCMV infection promoted HUVEC proliferation, whereas hyperglycemia and hyperlipidemia inhibited HUVEC proliferation. Additionally, treatment with decitabine (DAC) and RGS5 reversed the effects of HCMV infection on HUVEC proliferation, but not triggered by hyperglycemia and hyperlipidemia. In summary, upregulation of RGS5 may be a promising treatment for preventing HCMV-induced hypertension.
Collapse
|
9
|
Lee AS, Wang YC, Chang SS, Lo PH, Chang CM, Lu J, Burns AR, Chen CH, Kakino A, Sawamura T, Chang KC. Detection of a High Ratio of Soluble to Membrane-Bound LOX-1 in Aspirated Coronary Thrombi From Patients With ST-Segment-Elevation Myocardial Infarction. J Am Heart Assoc 2020; 9:e014008. [PMID: 31928155 PMCID: PMC7033847 DOI: 10.1161/jaha.119.014008] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Background The circulating level of soluble lectin‐like oxidized low‐density lipoprotein receptor‐1 (sLOX‐1) is a valuable biomarker of acute myocardial infarction (AMI). The most electronegative low‐density lipoprotein, L5, signals through LOX‐1 to trigger atherogenesis. We examined the characteristics of LOX‐1 and the role of L5 in aspirated coronary thrombi of AMI patients. Methods and Results Intracoronary thrombi were aspirated by performing interventional thrombosuction in patients with ST‐segment–elevation myocardial infarction (STEMI; n=32) or non–ST‐segment–elevation myocardial infarction (n=12). LOX‐1 level and the ratio of sLOX‐1 to membrane‐bound LOX‐1 were higher in thrombi of STEMI patients than in those of non–ST‐segment–elevation myocardial infarction patients. In all aspirated thrombi, LOX‐1 colocalized with apoB100. When we explored the role of L5 in AMI, deconvolution microscopy showed that particles of L5 but not L1 (the least electronegative low‐density lipoprotein) quickly formed aggregates prone to retention in thrombi. Treating human monocytic THP‐1 cells with L5 or L1 showed that L5 induced cellular adhesion and promoted the differentiation of monocytes into macrophages in a dose‐dependent manner. In a second cohort of AMI patients, the L5 percentage and plasma concentration of sLOX‐1 were higher in STEMI patients (n=33) than in non–ST‐segment–elevation myocardial infarction patients (n=25), and sLOX‐1 level positively correlated with L5 level in AMI patients. Conclusions The level of LOX‐1 and the ratio of sLOX‐1 to membrane‐bound LOX‐1 in aspirated thrombi, as well as the circulating level of sLOX‐1 were higher in STEMI patients than in non–ST‐segment–elevation myocardial infarction patients. L5 may play a role in releasing a high level of sLOX‐1 into the circulation of STEMI patients.
Collapse
Affiliation(s)
- An-Sheng Lee
- Department of Medicine Mackay Medical College New Taipei City Taiwan.,Cardiovascular Research Laboratory China Medical University Hospital Taichung Taiwan
| | - Yu-Chen Wang
- Cardiovascular Research Laboratory China Medical University Hospital Taichung Taiwan.,Division of Cardiovascular Medicine Asia University Hospital Taichung Taiwan.,Department of Biotechnology Asia University Taichung Taiwan.,Division of Cardiovascular Medicine China Medical University Hospital Taichung Taiwan
| | - Shih-Sheng Chang
- Division of Cardiovascular Medicine China Medical University Hospital Taichung Taiwan
| | - Ping-Hang Lo
- Division of Cardiovascular Medicine China Medical University Hospital Taichung Taiwan
| | - Chia-Ming Chang
- Cardiovascular Research Laboratory China Medical University Hospital Taichung Taiwan
| | - Jonathan Lu
- Vascular and Medicinal Research Texas Heart Institute Houston TX.,InVitro Cell Research LLC Englewood NJ
| | - Alan R Burns
- College of Optometry University of Houston Houston TX
| | - Chu-Huang Chen
- Vascular and Medicinal Research Texas Heart Institute Houston TX.,New York Heart Research Foundation Mineola NY
| | - Akemi Kakino
- Department of Life Innovation Institute for Biomedical Sciences Shinshu University Matsumoto Japan.,Department of Molecular Pathophysiology Shinshu University School of Medicine Matsumoto Japan
| | - Tatsuya Sawamura
- Department of Life Innovation Institute for Biomedical Sciences Shinshu University Matsumoto Japan.,Department of Molecular Pathophysiology Shinshu University School of Medicine Matsumoto Japan
| | - Kuan-Cheng Chang
- Cardiovascular Research Laboratory China Medical University Hospital Taichung Taiwan.,Division of Cardiovascular Medicine China Medical University Hospital Taichung Taiwan.,Graduate Institute of Biomedical Sciences China Medical University Taichung Taiwan
| |
Collapse
|
10
|
Salsalate ameliorates the atherosclerotic response through HO-1- and SIRT1-mediated suppression of ER stress and inflammation. Inflamm Res 2019; 68:655-663. [DOI: 10.1007/s00011-019-01248-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 05/07/2019] [Accepted: 05/15/2019] [Indexed: 12/12/2022] Open
|
11
|
Zhou Z, Chen Y, Ni W, Liu T. Upregulation of Nuclear Factor IA Suppresses Oxidized Low-Density Lipoprotein-Induced Endoplasmic Reticulum Stress and Apoptosis in Human Umbilical Vein Endothelial Cells. Med Sci Monit 2019; 25:1009-1016. [PMID: 30721172 PMCID: PMC6373224 DOI: 10.12659/msm.912132] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Endoplasmic reticulum stress (ERS) is part of the cardiovascular pathological processes, including atherosclerosis. Nuclear factor IA (NFIA) influences atherosclerosis development; however, its effects on ERS remain unknown. This study investigated the effect of NFIA on oxidized low-density lipoprotein (ox-LDL)-induced ERS and apoptosis in endothelial cells. Material/Methods Ox-LDL was used to induce lipotoxicity in human umbilical vein endothelial cells (HUVECs) to establish an in vitro oxidative injury model transfected with pcDNA3.0-NFIA. The cytotoxic response was detected using an assay to determine the release of lactate dehydrogenase (LDH). Morphological changes in cell apoptosis were detected using Hoechst 33258 staining. The proportion of apoptotic cells, releases of reactive oxygen species (ROS), and mitochondrial membrane potential (ΔΨm) were determined using flow cytometry. The expression levels of apoptosis- and ERS-related molecules were detected through Western blotting. Results NFIA expression was downregulated in the in vitro oxidative cell-injury model. Exposure of HUVECs to ox-LDL resulted in a significant increase in apoptosis, decrease in ROS levels, and loss of ΔΨm. Overexpression of NFIA remarkably inhibited ERS and mitochondrial-mediated apoptosis induced by ox-LDL in HUVECs by reversing the effect of ox-LDL on the expression of JNK1, p-JNK1, CHOP, Cyt C, and Bax. Conclusions These results demonstrated that NFIA might have beneficial effects in the prevention of ox-LDL-induced ERS and apoptosis in vascular endothelial cells. This study provided new insights into the mechanism of atherosclerosis.
Collapse
Affiliation(s)
- Zhenyu Zhou
- Department of Cardiology, Central Hospital of Nanchong, The Second Clinical School of North Sichuan Medical College, Nanchong, Sichuan, China (mainland)
| | - Yu Chen
- Comprehensive Ward, Central Hospital of Nanchong, The Second Clinical School of North Sichuan Medical College, Nanchong, Sichuan, China (mainland)
| | - Wei Ni
- Department of Cardiology, Central Hospital of Nanchong, The Second Clinical School of North Sichuan Medical College, Nanchong, Sichuan, China (mainland)
| | - Tao Liu
- Department of Cardiology, Central Hospital of Nanchong, The Second Clinical School of North Sichuan Medical College, Nanchong, Sichuan, China (mainland)
| |
Collapse
|
12
|
Donaldson CJ, Lao KH, Zeng L. The salient role of microRNAs in atherogenesis. J Mol Cell Cardiol 2018; 122:98-113. [DOI: 10.1016/j.yjmcc.2018.08.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Revised: 08/05/2018] [Accepted: 08/06/2018] [Indexed: 12/17/2022]
|
13
|
Lin F, Pei L, Zhang Q, Han W, Jiang S, Lin Y, Dong B, Cui L, Li M. Ox-LDL induces endothelial cell apoptosis and macrophage migration by regulating caveolin-1 phosphorylation. J Cell Physiol 2018; 233:6683-6692. [PMID: 29323707 DOI: 10.1002/jcp.26468] [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: 09/30/2017] [Accepted: 01/05/2018] [Indexed: 12/16/2022]
Abstract
Oxidative low-density lipoprotein (ox-LDL) is a risk factor for atherosclerosis. Ox-LDL leads to endothelial injury in the initial stage of atherosclerosis. In this study, we investigated the role of ox-LDL in endothelial injury and macrophage recruitment. We demonstrated that ox-LDL promoted a dose-dependent phosphorylation of caveolin-1 in human umbilical vein endothelial cells. Phosphorylated caveolin-1 increased ox-LDL uptake. Intracellular accumulation of ox-LDL induced NF-κB p65 phosphorylation, promoted HMGB1 translocation from nucleus to cytoplasm and cytochrome C release from mitochondria to cytoplasm, and activated caspase 3, resulting in cell apoptosis. NF-κB activation also facilitated cavolin-1 phosphorylation and HMGB1 expression. In addition, caveolin-1 phosphorylation favored HMGB1 release and nuclear translocation of EGR1. Nuclear translocation of EGR1 contributed to cytoplasmic translocation of HMGB1. The extracellular HMGB1 induced the migration of PMBC-derived macrophages toward HUVECs in a TLR4-dependent manner. Our results suggested that ox-LDL promoted HUVECs apoptosis and macrophage recruitment by regulating caveolin-1 phosphorylation.
Collapse
Affiliation(s)
- Fei Lin
- Department of Cardiology, Shandong Energy Zibo Mining Group Co., Ltd Central Hospital, Zibo, China
| | - Likai Pei
- Department of Cardiology, Shandong Energy Zibo Mining Group Co., Ltd Central Hospital, Zibo, China
| | - Qingbin Zhang
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Weizhong Han
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Shiliang Jiang
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yanliang Lin
- Department of Center Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Bo Dong
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Lianqun Cui
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Min Li
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
14
|
Teixeira V, Tam LS. Novel Insights in Systemic Lupus Erythematosus and Atherosclerosis. Front Med (Lausanne) 2018; 4:262. [PMID: 29435447 PMCID: PMC5796914 DOI: 10.3389/fmed.2017.00262] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/28/2017] [Indexed: 01/22/2023] Open
Abstract
Introduction The systemic inflammatory nature of systemic lupus erythematosus (SLE) is well patent not only in the diverse clinical manifestations of the disease but also in the increased risk of premature atherosclerosis and cardiovascular events (CVE), making SLE one of the most complex diseases to study and manage in clinical practice. Aim To travel from old aspects to modern insights on the physiopathology, new molecular biomarkers, imaging methods of atherosclerosis assessment, and the potential treatments of atherosclerosis in SLE. Methods We conducted a literature search using PubMed database and performed a critical review. Conclusion/discussion Several developments have taken place in the understanding of the relationship between SLE and premature atherosclerosis. Nevertheless, cardiovascular diseases are still the major cause of reduced life expectancy in SLE and the main cause of death. The lack of standardization methods for the imaging assessment of atherosclerosis in SLE and the multifactorial nature of the disease are well patriated in the difficulty of achieving consistent and reproducible results among studies that focus in cardiovascular risk assessment and prediction. A raising number of molecular biomarkers of atherosclerosis have been proposed, but the combination of several biomarkers and risk factors may better estimate cardiovascular disease risk. Moreover, the development of effective therapies to prevent progression of atherosclerosis and CVE shall address systemic inflammation.
Collapse
Affiliation(s)
- Vítor Teixeira
- Rheumatology Department, Centro Hospitalar de Lisboa Norte, EPE, Hospital de Santa Maria, Lisbon, Portugal
| | - Lai-Shan Tam
- Department of Medicine and Therapeutics, The Prince of Wales Hospital, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| |
Collapse
|
15
|
Han Y, Jiang Q, Wang Y, Li W, Geng M, Han Z, Chen X. The anti-proliferative effects of oleanolic acid on A7r5 cells-Role of UCP2 and downstream FGF-2/p53/TSP-1. Cell Biol Int 2017; 41:1296-1306. [PMID: 28792088 DOI: 10.1002/cbin.10838] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/07/2017] [Indexed: 01/11/2023]
Abstract
Vascular smooth muscle cell (VSMC) proliferation is a major contributor to atherosclerosis. This study investigated the inhibitory effects of oleanolic acid (OA) against oxidized low-density lipoprotein (ox-LDL)-induced VSMC proliferation in A7r5 cells and explored underlying molecular mechanism. The cell proliferation was quantified with cell counting kit-8 (CCK-8), in which ox-LDL significantly increased A7r5 cells proliferation, while OA pretreatment effectively alleviated such changes without inducing overt cytotoxicity, as indicated by lactate dehydrogenase (LDH) assay. Quantitative real-time RT-PCR (qRT-PCR) and Western blotting revealed increased UCP2 and FGF-2 expression levels as well as decreased p53 and TSP-1 expression levels in A7r5 cells following ox-LDL exposure, while OA pretreatment reversed such changes. Furthermore, inhibiting UCP2 with genipin remarkably reversed the changes in the expression levels of FGF-2, p53, and TSP-1 induced by ox-LDL exposure; silencing FGF-2 with siRNA did not significantly change the expression levels of UCP2 but effectively reversed the changes in the expression levels of p53 and TSP-1, and activation of p53 with PRIMA-1 only significantly affected the changes in the expression levels of TSP-1, but not in UCP2 or FGF-2, suggesting a UCP-2/FGF-2/p53/TSP-1 signaling in A7r5 cells response to ox-LDL exposure. Additionally, co-treatment of OA and genipin exhibited similar effects to the expression levels of UCP2, FGF-2, p53, and TSP-1 as OA or genipin solo treatment in ox-LDL-exposed A7r5 cells, suggesting the involvement of UCP-2/FGF-2/p53/TSP-1 in the mechanism of OA. In conclusion, OA inhibits ox-LDL-induced VSMC proliferation in A7r5 cells, the mechanism involves the changes in UCP-2/FGF-2/p53/TSP-1.
Collapse
Affiliation(s)
- Yantao Han
- Qingdao University Medical College, 308 Ningxia Road, Qingdao 266071, Shandong, China
| | - Qixiao Jiang
- Qingdao University Medical College, 308 Ningxia Road, Qingdao 266071, Shandong, China
| | - Yu Wang
- Qingdao University Medical College, 308 Ningxia Road, Qingdao 266071, Shandong, China
| | - Wenqian Li
- Qingdao University Medical College, 308 Ningxia Road, Qingdao 266071, Shandong, China
| | - Min Geng
- Qingdao University Medical College, 308 Ningxia Road, Qingdao 266071, Shandong, China
| | - Zhiwu Han
- The Affiliated Hospital of Qingdao University, 16 Jiansu Road, Qingdao 266021, Shandong, China
| | - Xuehong Chen
- Qingdao University Medical College, 308 Ningxia Road, Qingdao 266071, Shandong, China
| |
Collapse
|
16
|
Zhang C, Adamos C, Oh MJ, Baruah J, Ayee MAA, Mehta D, Wary KK, Levitan I. oxLDL induces endothelial cell proliferation via Rho/ROCK/Akt/p27 kip1 signaling: opposite effects of oxLDL and cholesterol loading. Am J Physiol Cell Physiol 2017; 313:C340-C351. [PMID: 28701359 DOI: 10.1152/ajpcell.00249.2016] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 07/05/2017] [Accepted: 07/05/2017] [Indexed: 12/19/2022]
Abstract
Oxidized modifications of LDL (oxLDL) play a key role in the development of endothelial dysfunction and atherosclerosis. However, the underlying mechanisms of oxLDL-mediated cellular behavior are not completely understood. Here, we compared the effects of two major types of oxLDL, copper-oxidized LDL (Cu2+-oxLDL) and lipoxygenase-oxidized LDL (LPO-oxLDL), on proliferation of human aortic endothelial cells (HAECs). Cu2+-oxLDL enhanced HAECs' proliferation in a dose- and degree of oxidation-dependent manner. Similarly, LPO-oxLDL also enhanced HAEC proliferation. Mechanistically, both Cu2+-oxLDL and LPO-oxLDL enhance HAEC proliferation via activation of Rho, Akt phosphorylation, and a decrease in the expression of cyclin-dependent kinase inhibitor 1B (p27kip1). Both Cu2+-oxLDL or LPO-oxLDL significantly increased Akt phosphorylation, whereas an Akt inhibitor, MK2206, blocked oxLDL-induced increase in HAEC proliferation. Blocking Rho with C3 or its downstream target ROCK with Y27632 significantly inhibited oxLDL-induced Akt phosphorylation and proliferation mediated by both Cu2+- and LPO-oxLDL. Activation of RhoA was blocked by Rho-GDI-1, which also abrogated oxLDL-induced Akt phosphorylation and HAEC proliferation. In contrast, blocking Rac1 in these cells had no effect on oxLDL-induced Akt phosphorylation or cell proliferation. Moreover, oxLDL-induced Rho/Akt signaling downregulated cell cycle inhibitor p27kip1 Preloading these cells with cholesterol, however, prevented oxLDL-induced Akt phosphorylation and HAEC proliferation. These findings provide a new understanding of the effects of oxLDL on endothelial proliferation, which is essential for developing new treatments against neovascularization and progression of atherosclerosis.
Collapse
Affiliation(s)
- Chongxu Zhang
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Crystal Adamos
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Myung-Jin Oh
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Jugajyoti Baruah
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Manuela A A Ayee
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Dolly Mehta
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Kishore K Wary
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Irena Levitan
- Division of Pulmonary and Critical Care, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| |
Collapse
|
17
|
Chen Z, Wang M, He Q, Li Z, Zhao Y, Wang W, Ma J, Li Y, Chang G. MicroRNA-98 rescues proliferation and alleviates ox-LDL-induced apoptosis in HUVECs by targeting LOX-1. Exp Ther Med 2017; 13:1702-1710. [PMID: 28565756 PMCID: PMC5443247 DOI: 10.3892/etm.2017.4171] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 05/16/2016] [Indexed: 12/26/2022] Open
Abstract
Oxidized low-density lipoprotein (ox-LDL) is a major and critical mediator of atherosclerosis, and the underlying mechanism is thought to involve the ox-LDL-induced dysfunction of endothelial cells (ECs). MicroRNAs (miRNAs), which are a group of small non-coding RNA molecules that post-transcriptionally regulate the expression of target genes, have been associated with diverse cellular functions and the pathogenesis of various diseases, including atherosclerosis. miRNA-98 (miR-98) has been demonstrated to be involved in the regulation of cellular apoptosis; however, the role of miR-98 in ox-LDL-induced dysfunction of ECs and atherosclerosis has yet to be elucidated. Therefore, the present study aimed to investigate the role of miR-98 in ox-LDL-induced dysfunction of ECs and the underlying mechanism. It was demonstrated that miR-98 expression was markedly downregulated in ox-LDL-treated human umbilical vein ECs (HUVECs) and that miR-98 promoted the proliferation and alleviated apoptosis of HUVECs exposed to ox-LDL. In addition, the results demonstrated that lectin-like oxidized low-density lipoprotein receptor 1 (LOX-1) was a direct target of miR-98 in HUVECs, as indicated by a luciferase assay. The results of the present study suggested that miR-98 may inhibit the uptake of toxic ox-LDL, maintain HUVEC proliferation and protect HUVECs against apoptosis via the suppression of LOX-1.
Collapse
Affiliation(s)
- Zhibo Chen
- Division of Vascular Surgery, Guangdong Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Mian Wang
- Division of Vascular Surgery, Guangdong Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Qiong He
- Division of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Zilun Li
- Division of Vascular Surgery, Guangdong Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yang Zhao
- Division of Vascular Surgery, Guangdong Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Wenjian Wang
- Laboratory of General Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Jieyi Ma
- Laboratory of General Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yongxin Li
- Department of Vascular Surgery, The First Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Guangqi Chang
- Division of Vascular Surgery, Guangdong Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
18
|
Camaré C, Pucelle M, Nègre-Salvayre A, Salvayre R. Angiogenesis in the atherosclerotic plaque. Redox Biol 2017; 12:18-34. [PMID: 28212521 PMCID: PMC5312547 DOI: 10.1016/j.redox.2017.01.007] [Citation(s) in RCA: 272] [Impact Index Per Article: 38.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/04/2017] [Accepted: 01/05/2017] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is a multifocal alteration of the vascular wall of medium and large arteries characterized by a local accumulation of cholesterol and non-resolving inflammation. Atherothrombotic complications are the leading cause of disability and mortality in western countries. Neovascularization in atherosclerotic lesions plays a major role in plaque growth and instability. The angiogenic process is mediated by classical angiogenic factors and by additional factors specific to atherosclerotic angiogenesis. In addition to its role in plaque progression, neovascularization may take part in plaque destabilization and thromboembolic events. Anti-angiogenic agents are effective to reduce atherosclerosis progression in various animal models. However, clinical trials with anti-angiogenic drugs, mainly anti-VEGF/VEGFR, used in anti-cancer therapy show cardiovascular adverse effects, and require additional investigations.
Collapse
Affiliation(s)
- Caroline Camaré
- INSERM - I2MC, U-1048, 1 avenue Jean Poulhès, BP 84225, 31432 Toulouse cedex 4, France; Université Paul Sabatier Toulouse III, Faculty of Medicine, Biochemistry Departement, Toulouse, France; CHU Toulouse, Rangueil, 1 avenue Jean Poulhès, TSA 50032, 31059 Toulouse Cedex 9, France
| | - Mélanie Pucelle
- INSERM - I2MC, U-1048, 1 avenue Jean Poulhès, BP 84225, 31432 Toulouse cedex 4, France
| | - Anne Nègre-Salvayre
- INSERM - I2MC, U-1048, 1 avenue Jean Poulhès, BP 84225, 31432 Toulouse cedex 4, France.
| | - Robert Salvayre
- INSERM - I2MC, U-1048, 1 avenue Jean Poulhès, BP 84225, 31432 Toulouse cedex 4, France; Université Paul Sabatier Toulouse III, Faculty of Medicine, Biochemistry Departement, Toulouse, France; CHU Toulouse, Rangueil, 1 avenue Jean Poulhès, TSA 50032, 31059 Toulouse Cedex 9, France.
| |
Collapse
|
19
|
Overexpression of LOXIN Protects Endothelial Progenitor Cells From Apoptosis Induced by Oxidized Low Density Lipoprotein. J Cardiovasc Pharmacol 2017; 67:326-35. [PMID: 26771151 DOI: 10.1097/fjc.0000000000000358] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Human endothelial progenitor cells (hEPC) are adult stem cells located in the bone marrow and peripheral blood. Studies have indicated that hEPC play an important role in the recovery and repair of injured endothelium, however, their quantity and functional capacity is reduced in several diseases including hypercholesterolemia. Recently, it has been demonstrated that hEPC express lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) and its activation by oxidized low-density lipoprotein (ox-LDL) induces cellular dysfunction and apoptosis. This study aimed to investigate whether overexpression of LOXIN, a truncated isoform of LOX-1 that acts as a dominant negative, plays a protective role against ox-LDL-induced apoptosis in hEPC. Human endothelial progenitor cells exposed to ox-LDL showed a significant increase in LOX-1 expression, and apoptosis began at ox-LDL concentrations above 50 μg/mL. All hEPC apoptosed at 200 μg/mL ox-LDL. High LOXIN expression was generated using adenoviral systems in hEPC and SiHa cells transduced with 100 colony-forming units per cell. Transduced LOXIN localized to the plasma membrane and blocked ox-LDL uptake mediated by LOX-1. Overexpression of LOXIN protected hEPC from ox-LDL-induced apoptosis, and therefore maybe a novel way of improving hEPC function and quantity. These results suggest that adenoviral vectors of LOXIN may provide a possible treatment for diseases related to ox-LDL and vascular endothelium dysfunction, including atherosclerosis.
Collapse
|
20
|
Laffont B, Rayner KJ. MicroRNAs in the Pathobiology and Therapy of Atherosclerosis. Can J Cardiol 2017; 33:313-324. [PMID: 28232017 DOI: 10.1016/j.cjca.2017.01.001] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 01/02/2017] [Accepted: 01/02/2017] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs are short noncoding RNAs, expressed in humans and involved in sequence-specific post-transcriptional regulation of gene expression. They have emerged as key players in a wide array of biological processes, and changes in their expression and/or function have been associated with plethora of human diseases. Atherosclerosis and its related clinical complications, such as myocardial infarction or stroke, represent the leading cause of death in the Western world. Accumulating experimental evidence has revealed a key role for microRNAs in regulating cellular and molecular processes related to atherosclerosis development, ranging from risk factors, to plaque initiation and progression, up to atherosclerotic plaque rupture. In this review, we focus on how microRNAs can influence atherosclerosis biology, as well as the potential clinical applications of microRNAs, which are being developed as targets as well as therapeutic agents for a growing industry hoping to harness the power of RNA-guided gene regulation to fight disease and infection.
Collapse
Affiliation(s)
- Benoit Laffont
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Katey J Rayner
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
21
|
Yang TC, Chang PY, Lu SC. L5-LDL from ST-elevation myocardial infarction patients induces IL-1β production via LOX-1 and NLRP3 inflammasome activation in macrophages. Am J Physiol Heart Circ Physiol 2016; 312:H265-H274. [PMID: 27864235 DOI: 10.1152/ajpheart.00509.2016] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 10/28/2016] [Accepted: 11/11/2016] [Indexed: 12/24/2022]
Abstract
L5-LDL, the most electronegative LDL associated with major cardiovascular risks, significantly rises in patients with ST-segment elevation myocardial infarction (STEMI). The inflammatory nature of atherosclerotic vascular diseases has prompted us to investigate whether L5-LDL induces the production of inflammatory cytokines, especially vascular ischemia-related interleukin (IL)-1β, in the pathogenesis of STEMI. Clinical data showed that plasma levels of L5-LDL and IL-1β were higher in the STEMI patients than in the controls (P < 0.05). In THP-1-derived human macrophages, L5-LDL significantly increased the levels of both IL-1β and cleaved caspase-1, indicating the activation of NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasomes by L5-LDL. Knockdown of NLRP3 and its adaptor protein apoptosis-associated speck-like protein containing a CARD (ASC) resulted in decreased L5-LDL-induced IL-1β. Furthermore, knock down of the lectin-type oxidized LDL receptor (LOX-1) in THP-1 cells attenuated L5-LDL-induced activation of NF-κB and caspase-1, leading to subsequent inhibition of IL-1β in macrophages. Furthermore, blockade LOX-1 with neutralizing antibody also inhibited L5-LDL-induced IL-1β in human peripheral blood mononuclear cell-derived macrophages. In conclusion, L5-LDL induces IL-1β production in macrophages by activation of NF-κB and caspase-1 through the LOX-1-dependent pathway. This study represents the evidence linking L5-LDL and the inflammatory cytokine IL-1β in STEMI, and identifies L5-LDL as a novel therapeutic target in acute myocardial infarction. NEW & NOTEWORTHY This study represents the evidence linking L5-LDL and the inflammatory cytokine IL-1β in ST-segment elevation myocardial infarction (STEMI). We elucidate the molecular mechanism underlying L5-LDL-induced production of IL-1β in macrophages. The results showed that L5-LDL induced activation of caspase-1 and NF-κB through the lectin-type oxidized LDL receptor (LOX-1)-dependent pathway, leading to the production of IL-1β.
Collapse
Affiliation(s)
- Tzu-Ching Yang
- Department of Biochemistry and Molecular Biology, National Taiwan University College of Medicine, Taipei, Taiwan; and
| | - Po-Yuan Chang
- Cardiovasccular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Shao-Chun Lu
- Department of Biochemistry and Molecular Biology, National Taiwan University College of Medicine, Taipei, Taiwan; and
| |
Collapse
|
22
|
Zhang M, Jiang L. Oxidized low-density lipoprotein decreases VEGFR2 expression in HUVECs and impairs angiogenesis. Exp Ther Med 2016; 12:3742-3748. [PMID: 28105106 DOI: 10.3892/etm.2016.3823] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 09/06/2016] [Indexed: 12/22/2022] Open
Abstract
Atherosclerosis (AS), which is triggered by endothelial cell injury, evolves into a chronic inflammatory disease. Oxidized low-density lipoprotein (ox-LDL) is an important risk factor for the development of atherosclerosis; ox-LDL induces atherosclerotic plaque formation via scavenging receptors. The present study used ox-LDL-treated human umbilical vein endothelial cells (HUVECs) to investigate the effect of ox-LDL on angiogenesis. ox-LDL decreased HUVEC proliferation by MTT, induced apoptosis by Annexin V-fluorescein isothiocyanate (FITC) staining and markedly suppressed HUVEC tube formation by the Matrigel assay in a dose-dependent manner. Angiogenesis has been correlated with monocyte invasion, plaque instability and atherosclerotic lesion formation. In addition, ox-LDL induced the overproduction of reactive oxygen species using DCFH-DA staining and increased caspase-3 activity. Vascular endothelial growth factor receptor 2 (VEGFR2) were detected by quantitative polymerase chain reaction and western blot analysis and has previously been observed to have a key role in angiogenesis. Furthermore, the present study demonstrated that the abundance of VEGFR2 was decreased in ox-LDL-treated HUVECs. These results suggested that ox-LDL impairs angiogenesis via VEGFR2 degradation, thus suggesting that VEGFR2 may be involved in adaptation to oxidative stress and AS.
Collapse
Affiliation(s)
- Min Zhang
- Division of Cardiology, Shanghai Tongren Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200336, P.R. China
| | - Li Jiang
- Division of Cardiology, Shanghai Tongren Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200336, P.R. China
| |
Collapse
|
23
|
Yao G, Zhang Q, Doeppner TR, Niu F, Li Q, Yang Y, Kuckelkorn U, Hagemann N, Li W, Hermann DM, Dai Y, Zhou W, Jin F. LDL suppresses angiogenesis through disruption of the HIF pathway via NF-κB inhibition which is reversed by the proteasome inhibitor BSc2118. Oncotarget 2016; 6:30251-62. [PMID: 26388611 PMCID: PMC4745795 DOI: 10.18632/oncotarget.4943] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 09/04/2015] [Indexed: 02/05/2023] Open
Abstract
Since disturbance of angiogenesis predisposes to ischemic injuries, attempts to promote angiogenesis have been made to improve clinical outcomes of patients with many ischemic disorders. While hypoxia inducible factors (HIFs) stimulate vascular remodeling and angiogenesis, hyperlipidemia impairs angiogenesis in response to various pro-angiogenic factors. However, it remains uncertain how HIFs regulate angiogenesis under hyperlipidemia. Here, we report that exposure to low-density lipoprotein (LDL) suppressed in vitro angiogenesis of human brain microvascular endothelial cells. Whereas LDL exposure diminished expression of HIF-1α and HIF-2α induced by hypoxia, it inhibited DMOG- and TNFα-induced HIF-1α and HIF-2α expression in normoxia. Notably, in both hypoxia and normoxia, LDL markedly reduced expression of HIF-1β, a constitutively stable HIF subunit, an event associated with NF-κB inactivation. Moreover, knockdown of HIF-1β down-regulated HIF-1α and HIF-2α expression, in association with increased HIF-1α hydroxylation and 20S proteasome activity after LDL exposure. Significantly, the proteasome inhibitor BSc2118 prevented angiogenesis attenuation by LDL through restoring expression of HIFs. Together, these findings argue that HIF-1β might act as a novel cross-link between the HIF and NF-κB pathways in suppression of angiogenesis by LDL, while proteasome inhibitors might promote angiogenesis by reactivating this signaling cascade under hyperlipidemia.
Collapse
Affiliation(s)
- Gang Yao
- Cancer Center, The First Affiliated Hospital, Jilin University, Changchun, Jilin, China.,Department of Neurology, The Second Affiliated Hospital, Jilin University, Changchun, Jilin, China
| | - Qi Zhang
- Cancer Center, The First Affiliated Hospital, Jilin University, Changchun, Jilin, China
| | | | - Feng Niu
- Cancer Center, The First Affiliated Hospital, Jilin University, Changchun, Jilin, China
| | - Qiaochuan Li
- Department of Hematology, The First Affiliated Hospital, Guangxi Medical University, Nanning, Guangxi, China
| | - Yanping Yang
- Cancer Center, The First Affiliated Hospital, Jilin University, Changchun, Jilin, China
| | - Ulrike Kuckelkorn
- Department of Biochemistry, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Nina Hagemann
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Wei Li
- Cancer Center, The First Affiliated Hospital, Jilin University, Changchun, Jilin, China
| | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Yun Dai
- Department of Medicine, Virginia Commonwealth University, Massey Cancer Center, Richmond, Virginia, USA
| | - Wen Zhou
- Cancer Research Institute, Central South University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Key Laboratory of Carcinogenesis, National Health and Family Planning Commission, Changsha, Hunan, China
| | - Fengyan Jin
- Cancer Center, The First Affiliated Hospital, Jilin University, Changchun, Jilin, China
| |
Collapse
|
24
|
Zhang Y, Mu Q, Zhou Z, Song H, Zhang Y, Wu F, Jiang M, Wang F, Zhang W, Li L, Shao L, Wang X, Li S, Yang L, Wu Q, Zhang M, Tang D. Protective Effect of Irisin on Atherosclerosis via Suppressing Oxidized Low Density Lipoprotein Induced Vascular Inflammation and Endothelial Dysfunction. PLoS One 2016; 11:e0158038. [PMID: 27355581 PMCID: PMC4927070 DOI: 10.1371/journal.pone.0158038] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 06/09/2016] [Indexed: 12/27/2022] Open
Abstract
Irisin, a newly discovered myokine, is considered as a promising candidate for the treatment of metabolic disturbances and cardiovascular diseases. In the present study, we used two animal models, apolipoprotein E-deficient mice fed on a high-cholesterol diet and a mouse carotid partial ligation model to test the anti-atherosclerotic effect of irisin. Irisin treatment (0.5 μg/g body weight/day) significantly reduced the severity of aortic atherosclerosis in apolipoprotein E-deficient mice fed on a high-cholesterol diet and suppressed carotid neointima formation in a carotid partial ligation model. It was associated with decreased inflammation and cell apoptosis in aortic tissues. In addition, in a cell culture model, irisin restored ox-LDL-induced human umbilical vein endothelial cell dysfunction by reducing the levels of inflammatory genes via inhibiting the reactive oxygen species (ROS)/ p38 MAPK/ NF-κB signaling pathway activation and inhibiting cell apoptosis via up-regulating Bcl-2 and down-regulating Bax and caspase-3 expression. Our study demonstrated that irisin significantly reduced atherosclerosis in apolipoprotein E-deficient mice via suppressing ox-LDL-induced cell inflammation and apoptosis, which might have a direct therapeutic effect on atherosclerotic diseases.
Collapse
Affiliation(s)
- Yuzhu Zhang
- Center for Stem Cell and Regenerative Medicine, The Second Hospital of Shandong University, Jinan, People’s Republic of China
| | - Qian Mu
- Center for Stem Cell and Regenerative Medicine, The Second Hospital of Shandong University, Jinan, People’s Republic of China
| | - Zheng Zhou
- Chaohu Road Community Health Center of Qingdao, Qingdao, People’s Republic of China
| | - Haibo Song
- Center for Stem Cell and Regenerative Medicine, The Second Hospital of Shandong University, Jinan, People’s Republic of China
| | - Yuan Zhang
- Center for Stem Cell and Regenerative Medicine, The Second Hospital of Shandong University, Jinan, People’s Republic of China
| | - Fei Wu
- Center for Stem Cell and Regenerative Medicine, The Second Hospital of Shandong University, Jinan, People’s Republic of China
| | - Miao Jiang
- Center for Stem Cell and Regenerative Medicine, The Second Hospital of Shandong University, Jinan, People’s Republic of China
| | - Fang Wang
- Center for Stem Cell and Regenerative Medicine, The Second Hospital of Shandong University, Jinan, People’s Republic of China
| | - Wen Zhang
- Center for Stem Cell and Regenerative Medicine, The Second Hospital of Shandong University, Jinan, People’s Republic of China
| | - Liang Li
- Center for Stem Cell and Regenerative Medicine, The Second Hospital of Shandong University, Jinan, People’s Republic of China
| | - Lei Shao
- Center for Stem Cell and Regenerative Medicine, The Second Hospital of Shandong University, Jinan, People’s Republic of China
| | - Xingli Wang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Shiwu Li
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Lijun Yang
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Qi Wu
- Department of Anatomy, School of Medicine Shandong University, Jinan, People’s Republic of China
| | - Mingxiang Zhang
- Key Laboratory of Cardiovascular Remodeling and Function Research, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Dongqi Tang
- Center for Stem Cell and Regenerative Medicine, The Second Hospital of Shandong University, Jinan, People’s Republic of China
- * E-mail:
| |
Collapse
|
25
|
Giacomini A, Chiodelli P, Matarazzo S, Rusnati M, Presta M, Ronca R. Blocking the FGF/FGFR system as a two-compartment antiangiogenic/antitumor approach in cancer therapy. Pharmacol Res 2016; 107:172-185. [DOI: 10.1016/j.phrs.2016.03.024] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 03/15/2016] [Accepted: 03/16/2016] [Indexed: 12/22/2022]
|
26
|
Santovito D, Egea V, Weber C. Small but smart: MicroRNAs orchestrate atherosclerosis development and progression. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1861:2075-2086. [PMID: 26738655 DOI: 10.1016/j.bbalip.2015.12.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 12/21/2015] [Indexed: 02/07/2023]
Abstract
MicroRNAs (miRNAs) are short non-coding RNA able to bind specific sequences on target messenger RNAs (mRNAs) and thereby to post-transcriptionally modulate gene expression. Being expressed in all vertebrate cell types, miRNAs have emerged as key players in a wide array of biological processes, including cell proliferation, differentiation and apoptosis. Over the past decade, knowledge concerning the contribution of miRNAs to human pathology has grown with an astonishing pace. In particular, a major involvement of miRNAs in atherosclerosis as a leading cause of global mortality has been supported by ample evidence from in vitro, in vivo and clinical studies. This review aims to summarize and highlight current concepts of miRNA function in the continuum of atherogenesis ranging from risk factors (i.e. dyslipidemia, diabetes, hypertension), to endothelial dysfunction up to the events leading to plaque rupture. Areas in need for further research and potential perspectives for translational applications will be scrutinized. This article is part of a Special Issue entitled: MicroRNAs and lipid/energy metabolism and related diseases edited by Carlos Fernández-Hernando and Yajaira Suárez.
Collapse
Affiliation(s)
- Donato Santovito
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Virginia Egea
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany; German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
27
|
Phosphocreatine protects endothelial cells from oxidized low-density lipoprotein-induced apoptosis by modulating the PI3K/Akt/eNOS pathway. Apoptosis 2015; 20:1563-76. [DOI: 10.1007/s10495-015-1175-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
28
|
Lopez-Pastrana J, Ferrer LM, Li YF, Xiong X, Xi H, Cueto R, Nelson J, Sha X, Li X, Cannella AL, Imoukhuede PI, Qin X, Choi ET, Wang H, Yang XF. Inhibition of Caspase-1 Activation in Endothelial Cells Improves Angiogenesis: A NOVEL THERAPEUTIC POTENTIAL FOR ISCHEMIA. J Biol Chem 2015; 290:17485-94. [PMID: 26037927 DOI: 10.1074/jbc.m115.641191] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Indexed: 12/12/2022] Open
Abstract
Deficient angiogenesis may contribute to worsen the prognosis of myocardial ischemia, peripheral arterial disease, ischemic stroke, etc. Dyslipidemic and inflammatory environments attenuate endothelial cell (EC) proliferation and angiogenesis, worsening the prognosis of ischemia. Under these dyslipidemic and inflammatory environments, EC-caspase-1 becomes activated and induces inflammatory cell death that is defined as pyroptosis. However, the underlying mechanism that correlates caspase-1 activation with angiogenic impairment and the prognosis of ischemia remains poorly defined. By using flow cytometric analysis, enzyme and receptor inhibitors, and hind limb ischemia model in caspase-1 knock-out (KO) mice, we examined our novel hypothesis, i.e. inhibition of caspase-1 in ECs under dyslipidemic and inflammatory environments attenuates EC pyroptosis, improves EC survival mediated by vascular endothelial growth factor receptor 2 (VEGFR-2), angiogenesis, and the prognosis of ischemia. We have made the following findings. Proatherogenic lipids induce higher caspase-1 activation in larger sizes of human aortic endothelial cells (HAECs) than in smaller sizes of HAECs. Proatherogenic lipids increase pyroptosis significantly more in smaller sizes of HAECs than in larger sizes of the cells. VEGFR-2 inhibition increases caspase-1 activation in HAECs induced by lysophosphatidylcholine treatment. Caspase-1 activation inhibits VEGFR-2 expression. Caspase-1 inhibition improves the tube formation of lysophosphatidylcholine-treated HAECs. Finally, caspase-1 depletion improves angiogenesis and blood flow in mouse hind limb ischemic tissues. Our results have demonstrated for the first time that inhibition of proatherogenic caspase-1 activation in ECs improves angiogenesis and the prognosis of ischemia.
Collapse
Affiliation(s)
- Jahaira Lopez-Pastrana
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research
| | - Lucas M Ferrer
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, the Department of Bioengineering, University of Illinois-Urbana Champaign, Urbana, Illinois 61801
| | - Ya-Feng Li
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research
| | - Xinyu Xiong
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Departments of Pharmacology
| | - Hang Xi
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Departments of Pharmacology
| | - Ramon Cueto
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Departments of Pharmacology
| | - Jun Nelson
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research
| | - Xiaojin Sha
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research
| | - Xinyuan Li
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Departments of Pharmacology
| | - Ann L Cannella
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Departments of Pharmacology
| | - Princess I Imoukhuede
- the Department of Bioengineering, University of Illinois-Urbana Champaign, Urbana, Illinois 61801
| | | | - Eric T Choi
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Surgery, Temple University School of Medicine, Philadelphia, Pennsylvania 19140 and
| | - Hong Wang
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Departments of Pharmacology
| | - Xiao-Feng Yang
- From the Centers for Metabolic Disease Research, Cardiovascular Research and Thrombosis Research, Departments of Pharmacology,
| |
Collapse
|
29
|
Abstract
Atherosclerosis is characterised by the accumulation of lipid-laden macrophages in atherosclerotic lesions and occurs preferentially at arterial branching points, which are prone to inflammation during hyperlipidaemic stress. The increased susceptibility at branching sites of arteries is attributable to poor adaptation of arterial endothelial cells to disturbed blood flow. In the past 5 years, several studies have provided mechanistic insights into the regulatory roles of microRNAs (miRNAs) in inflammatory activation, proliferation, and regeneration of endothelial cells during this maladaptive process. The intercellular transfer of vesicle-bound miRNAs contributes to arterial homeostasis, and the combinatorial effect of multiple miRNAs controls the unresolved inflammation orchestrated by macrophages in atherosclerotic lesions. In this Review, we highlight the miRNA-dependent regulation of the endothelial phenotype and the proliferative reserve that occurs in response to altered haemodynamic conditions as a prerequisite for atherogenic inflammation. In particular, we discuss the regulation of transcriptional modules by miRNAs and the protective role of complementary strand pairs, which encompasses remote miRNA signalling. In addition, we review the roles of miRNA tandems and describe the relevance of RNA target selection and competition to the behaviour of lesional macrophages. Elucidating miRNA-mediated regulatory mechanisms can aid the development of novel diagnostic and therapeutic strategies for atherosclerosis.
Collapse
|
30
|
CHENG YUNPENG, LIU XIAOYUN, ZHANG LIJIAO, ZHANG YING, LIU YING, LU YAN, JIANG YINONG. Effect of oxidized low-density lipoprotein on the expression of the prorenin receptor in human aortic smooth muscle cells. Mol Med Rep 2015; 11:4341-4. [DOI: 10.3892/mmr.2015.3254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 12/17/2014] [Indexed: 11/06/2022] Open
|
31
|
Camaré C, Trayssac M, Garmy-Susini B, Mucher E, Sabbadini R, Salvayre R, Negre-Salvayre A. Oxidized LDL-induced angiogenesis involves sphingosine 1-phosphate: prevention by anti-S1P antibody. Br J Pharmacol 2014; 172:106-18. [PMID: 25176316 DOI: 10.1111/bph.12897] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 08/13/2014] [Accepted: 08/24/2014] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE Neovascularization occurring in atherosclerotic lesions may promote plaque expansion, intraplaque haemorrhage and rupture. Oxidized LDL (oxLDL) are atherogenic, but their angiogenic effect is controversial; both angiogenic and anti-angiogenic effects have been reported. The angiogenic mechanism of oxLDL is partly understood, but the role of the angiogenic sphingolipid, sphingosine 1-phosphate (S1P), in this process is not known. Thus, we investigated whether S1P is involved in the oxLDL-induced angiogenesis and whether an anti-S1P monoclonal antibody can prevent this effect. EXPERIMENTAL APPROACH Angiogenesis was assessed by capillary tube formation by human microvascular endothelial cells (HMEC-1) cultured on Matrigel and in vivo by the Matrigel plug assay in C57BL/6 mice. KEY RESULTS Human oxLDL exhibited a biphasic angiogenic effect on HMEC-1; low concentrations were angiogenic, higher concentrations were cytotoxic. The angiogenic response to oxLDL was blocked by the sphingosine kinase (SPHK) inhibitor, dimethylsphingosine, by SPHK1-siRNA and by an anti-S1P monoclonal antibody. Moreover, inhibition of oxLDL uptake and subsequent redox signalling by anti-CD36 and anti-LOX-1 receptor antibodies and by N-acetylcysteine, respectively, blocked SPHK1 activation and tube formation. In vivo, in the Matrigel plug assay, low concentrations of human oxLDL or murine oxVLDL also triggered angiogenesis, which was prevented by i.p. injection of the anti-S1P antibody. CONCLUSION AND IMPLICATIONS These data highlight the role of S1P in angiogenesis induced by oxLDL both in HMEC-1 cultured on Matrigel and in vivo in the Matrigel plug model in mice, and demonstrate that the anti-S1P antibody effectively blocks the angiogenic effect of oxLDL.
Collapse
Affiliation(s)
- Caroline Camaré
- Inserm UMR-1048, Toulouse, France; Department of Biochemistry, University of Toulouse, France
| | | | | | | | | | | | | |
Collapse
|
32
|
Nazari-Jahantigh M, Egea V, Schober A, Weber C. MicroRNA-specific regulatory mechanisms in atherosclerosis. J Mol Cell Cardiol 2014; 89:35-41. [PMID: 25450610 DOI: 10.1016/j.yjmcc.2014.10.021] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 10/30/2014] [Accepted: 10/31/2014] [Indexed: 10/24/2022]
Abstract
During the past decade, the crucial role of microRNAs (miRs) controlling tissue homeostasis and disease in the cardiovascular system has become widely recognized. By controlling the expression levels of their targets, several miRs have been shown to modulate the function of endothelial cells, vascular smooth muscle cells, and macrophages, thereby regulating the development and progression of atherosclerosis. For instance, miR-155 can exacerbate early stages of atherosclerosis by increasing the inflammatory activation and disturbing efficient lipid handling in macrophages. Conversely, miRs can exert atheroprotective roles, as has been established for the complementary miR-126 strand pair, which forms a dual system sustaining the endothelial proliferative reserve and promoting endothelial regeneration to counteract atherogenic effects of disturbed flow and hyperlipidemia. Under some conditions, miRs are released from cells and are transported by microvesicles, ribonucleoprotein complexes, and lipoproteins, being remarkably stable in circulation. Conferred by such delivery modules, miRs can regulate target mRNAs in recipient cells, representing a new tool for cell-cell communication in the context of atherosclerotic disease. Here, we will discuss novel aspects of miR-mediated regulatory mechanisms, namely the regulation by competing RNA targets, miRNA tandems, or complementary miR strand pairs, as well as their potential diagnostic and therapeutic value in atherosclerosis. This article is part of a Special Issue entitled 'Non-coding RNAs'.
Collapse
Affiliation(s)
| | - Virginia Egea
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Andreas Schober
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany; German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany; German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
33
|
Abstract
The prevalence of atherosclerosis (ATH) is higher in patients with systemic lupus erythematosus (SLE) and occurs at an earlier age. The lupus-related factors that account for this increased risk are likely numerous and related to the factors described in this article. Identifying of at-risk subjects and increasing the understanding of pathogenesis of ATH in SLE is critical for improving the quality of care and improving mortality in this vulnerable population.
Collapse
Affiliation(s)
- Maureen McMahon
- Division of Rheumatology, David Geffen School of Medicine, University of California, Los Angeles, 1000 Veteran Avenue, Room 32-59, Los Angeles, CA 90095, USA.
| | - Brian Skaggs
- Division of Rheumatology, David Geffen School of Medicine, University of California, Los Angeles, 1000 Veteran Avenue, Room 32-59, Los Angeles, CA 90095, USA
| |
Collapse
|
34
|
Tiwari RL, Singh V, Singh A, Rana M, Verma A, Kothari N, Kohli M, Bogra J, Dikshit M, Barthwal MK. PKCδ-IRAK1 axis regulates oxidized LDL-induced IL-1β production in monocytes. J Lipid Res 2014; 55:1226-44. [PMID: 24792928 DOI: 10.1194/jlr.m045658] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Indexed: 12/19/2022] Open
Abstract
This study examined the role of interleukin (IL)-1 receptor-associated kinase (IRAK) and protein kinase C (PKC) in oxidized LDL (Ox-LDL)-induced monocyte IL-1β production. In THP1 cells, Ox-LDL induced time-dependent secretory IL-1β and IRAK1 activity; IRAK4, IRAK3, and CD36 protein expression; PKCδ-JNK1 phosphorylation; and AP-1 activation. IRAK1/4 siRNA and inhibitor (INH)-attenuated Ox-LDL induced secreted IL-1β and pro-IL-1β mRNA and pro-IL-1β and mature IL-1β protein expression, respectively. Diphenyleneiodonium chloride (NADPH oxidase INH) and N-acetylcysteine (free radical scavenger) attenuated Ox-LDL-induced reactive oxygen species generation, caspase-1 activity, and pro-IL-1β and mature IL-1β expression. Ox-LDL-induced secretory IL-1β production was abrogated in the presence of JNK INH II, Tanshinone IIa, Ro-31-8220, Go6976, Rottlerin, and PKCδ siRNA. PKCδ siRNA attenuated the Ox-LDL-induced increase in IRAK1 kinase activity, JNK1 phosphorylation, and AP-1 activation. In THP1 macrophages, CD36, toll-like receptor (TLR)2, TLR4, TLR6, and PKCδ siRNA prevented Ox-LDL-induced PKCδ and IRAK1 activation and IL-1β production. Enhanced Ox-LDL and IL-1β in systemic inflammatory response syndrome (SIRS) patient plasma demonstrated positive correlation with each other and with disease severity scores. Ox-LDL-containing plasma induced PKCδ and IRAK1 phosphorylation and IL-1β production in a CD36-, TLR2-, TLR4-, and TLR6-dependent manner in primary human monocytes. Results suggest involvement of CD36, TLR2, TLR4, TLR6, and the PKCδ-IRAK1-JNK1-AP-1 axis in Ox-LDL-induced IL-1β production.
Collapse
Affiliation(s)
- Rajiv Lochan Tiwari
- Pharmacology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India
| | - Vishal Singh
- Pharmacology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India
| | - Ankita Singh
- Pharmacology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India
| | - Minakshi Rana
- Pharmacology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India
| | - Anupam Verma
- Department of Transfusion Medicine, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Nikhil Kothari
- Department of Anaesthesia, King George's Medical University, Lucknow, India
| | - Monica Kohli
- Department of Anaesthesia, King George's Medical University, Lucknow, India
| | - Jaishri Bogra
- Department of Anaesthesia, King George's Medical University, Lucknow, India
| | - Madhu Dikshit
- Pharmacology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India
| | - Manoj Kumar Barthwal
- Pharmacology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India
| |
Collapse
|
35
|
MicroRNA-126-5p promotes endothelial proliferation and limits atherosclerosis by suppressing Dlk1. Nat Med 2014; 20:368-76. [PMID: 24584117 DOI: 10.1038/nm.3487] [Citation(s) in RCA: 489] [Impact Index Per Article: 48.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 01/29/2014] [Indexed: 12/14/2022]
Abstract
Atherosclerosis, a hyperlipidemia-induced chronic inflammatory process of the arterial wall, develops preferentially at sites where disturbed laminar flow compromises endothelial cell (EC) function. Here we show that endothelial miR-126-5p maintains a proliferative reserve in ECs through suppression of the Notch1 inhibitor delta-like 1 homolog (Dlk1) and thereby prevents atherosclerotic lesion formation. Endothelial recovery after denudation was impaired in Mir126(-/-) mice because lack of miR-126-5p, but not miR-126-3p, reduced EC proliferation by derepressing Dlk1. At nonpredilection sites, high miR-126-5p levels in endothelial cells confer a proliferative reserve that compensates for the antiproliferative effects of hyperlipidemia, such that atherosclerosis was exacerbated in Mir126(-/-) mice. In contrast, downregulation of miR-126-5p by disturbed flow abrogated EC proliferation at predilection sites in response to hyperlipidemic stress through upregulation of Dlk1 expression. Administration of miR-126-5p rescued EC proliferation at predilection sites and limited atherosclerosis, introducing a potential therapeutic approach.
Collapse
|
36
|
Yang TC, Chen YJ, Chang SF, Chen CH, Chang PY, Lu SC. Malondialdehyde mediates oxidized LDL-induced coronary toxicity through the Akt-FGF2 pathway via DNA methylation. J Biomed Sci 2014; 21:11. [PMID: 24490960 PMCID: PMC3931320 DOI: 10.1186/1423-0127-21-11] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Accepted: 01/29/2014] [Indexed: 12/22/2022] Open
Abstract
Background Oxidized LDL (oxLDL) is involved in the development of atherosclerotic heart disease through a mechanism that is not fully understood. In this study, we examined the role of malondialdehyde (MDA), an important oxidative stress epitope of oxLDL, in mediating coronary endothelial cytotoxicity. Results Human coronary artery endothelial cells (HCAECs) were treated with oxLDL in the presence or absence of antibody against MDA (anti-MDA) or apoB100 (anti-apoB100). In HCAECs treated with oxLDL (100 μg/ml) alone, DNA synthesis, cell viability, and expression of prosurvival fibroblast growth factor 2 (FGF2) were significantly reduced (P < 0.01 vs phosphate buffered saline–treated cells). These inhibitory effects of oxLDL were significantly attenuated in HCAECs cotreated with anti-MDA (0.15 μg/ml; P < 0.05 vs oxLDL-treated cells), but not in those cotreated with anti-apoB100. When we tested the effects of a panel of signal transduction modifiers on the signal transduction pathways of MDA in oxLDL-treated HCAECs, we found that MDA-induced cytotoxicity was mediated partly through the Akt pathway. Using a reporter gene assay, we identified an oxLDL-response element in the FGF2 promoter that was responsible for the transcriptional repression of FGF2 by oxLDL. The results of bisulfite genomic DNA sequencing showed that in HCAECs treated with oxLDL, the GC-rich promoter of FGF2 was heavily methylated at cytosine residues, whereas cotreatment with anti-MDA markedly reduced oxLDL-induced FGF2 promoter methylation. Conclusion OxLDL disrupts the growth and survival of HCAECs through an MDA-dependent pathway involving methylation of the FGF2 promoter and repression of FGF2 transcription. This novel epigenetic mechanism of oxLDL may underlie its atherogenicity in patients with atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
| | | | | | | | - Po-Yuan Chang
- Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, No, 7, Chung-Shan South Road, Taipei 100, Taiwan.
| | | |
Collapse
|
37
|
Jin F, Hagemann N, Schäfer ST, Brockmeier U, Zechariah A, Hermann DM. SDF-1 restores angiogenesis synergistically with VEGF upon LDL exposure despite CXCR4 internalization and degradation. Cardiovasc Res 2013; 100:481-91. [DOI: 10.1093/cvr/cvt209] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
38
|
Chu CS, Wang YC, Lu LS, Walton B, Yilmaz HR, Huang RY, Sawamura T, Dixon RAF, Lai WT, Chen CH, Lu J. Electronegative low-density lipoprotein increases C-reactive protein expression in vascular endothelial cells through the LOX-1 receptor. PLoS One 2013; 8:e70533. [PMID: 23950953 PMCID: PMC3738565 DOI: 10.1371/journal.pone.0070533] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 06/19/2013] [Indexed: 12/14/2022] Open
Abstract
Objectives Increased plasma C-reactive protein (CRP) levels are associated with the occurrence and severity of acute coronary syndrome. We investigated whether CRP can be generated in vascular endothelial cells (ECs) after exposure to the most electronegative subfraction of low-density lipoprotein (LDL), L5, which is atherogenic to ECs. Because L5 and CRP are both ligands for the lectin-like oxidized LDL receptor-1 (LOX-1), we also examined the role of LOX-1. Methods and Results Plasma LDL samples isolated from asymptomatic hypercholesterolemic (LDL cholesterol [LDL-C] levels, 154.6±20 mg/dL; n = 7) patients and normocholesterolemic (LDL-C levels, 86.1±21 mg/dL; P<0.001; n = 7) control individuals were chromatographically resolved into 5 subfractions, L1-L5. The L5 percentage (L5%) and the plasma L5 concentration ([L5] = L5% × LDL-C) in the patient and control groups were 8.1±2% vs. 2.3±1% (P<0.001) and 12.6±4 mg/dL vs. 1.9±1 mg/dL (P<0.001), respectively. In hypercholesterolemic patients treated with atorvastatin for 6 months (10 mg/day), [L5] decreased from 12.6±4 mg/dL to 4.5±1.1 mg/dL (P = 0.011; n = 5), whereas both [L5] and L5% returned to baseline levels in 2 noncompliant patients 3 months after discontinuation. In cultured human aortic ECs (HAECs), L5 upregulated CRP expression in a dose- and time-dependent manner up to 2.5-fold (P<0.01), whereas the least electronegative subfraction, L1, had no effect. DiI-labeled L1, internalized through the LDL receptor, became visible inside HAECs within 30 seconds. In contrast, DiI-labeled L5, internalized through LOX-1, became apparent after 5 minutes. L5-induced CRP expression manifested at 30 minutes and was attenuated by neutralizing LOX-1. After 30 minutes, L5 but not L1 induced reactive oxygen species (ROS) production. Both L5-induced ROS and CRP production were attenuated by ROS inhibitor N-acetyl cysteine. Conclusions Our results suggest that CRP, L5, and LOX-1 form a cyclic mechanism in atherogenesis and that reducing plasma L5 levels with atorvastatin disrupts the vascular toxicity of L5.
Collapse
Affiliation(s)
- Chih-Sheng Chu
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Zechariah A, ElAli A, Hagemann N, Jin F, Doeppner TR, Helfrich I, Mies G, Hermann DM. Hyperlipidemia Attenuates Vascular Endothelial Growth Factor–Induced Angiogenesis, Impairs Cerebral Blood Flow, and Disturbs Stroke Recovery via Decreased Pericyte Coverage of Brain Endothelial Cells. Arterioscler Thromb Vasc Biol 2013; 33:1561-7. [DOI: 10.1161/atvbaha.112.300749] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Objective—
Therapeutic angiogenesis aims at the promotion of vascular growth, usually under conditions of atherosclerosis. It was unknown how hyperlipidemia, a risk factor that is closely associated with atherosclerosis of brain vessels in humans, influences vascular endothelial growth factor–induced angiogenesis and stroke recovery.
Approach and Results—
Wild-type and apolipoprotein-E (ApoE)
−/−
mice were kept on regular or cholesterol-rich diet for mimicking different severities of hyperlipidemia. Mice were treated intracerebroventricularly with recombinant human vascular endothelial growth factor for 21 days (0.02 µg/d) and subsequently subjected to 90-minute middle cerebral artery occlusion followed by 1 or 24 hours of reperfusion. Histochemical, autoradiographic, and regional bioluminescence techniques were used to evaluate effects of blood lipids on postischemic angiogenesis, histopathologic brain injury, cerebral blood flow, protein synthesis and energy state, and pericyte coverage of brain endothelial cells. Hyperlipidemia dose-dependently attenuated vascular endothelial growth factor–induced capillary formation and pericyte coverage of brain endothelial cells, abolishing the improvement of cerebral blood flow during subsequent stroke, resulting in the loss of the metabolic penumbra and increased brain infarction. The enhanced angiogenesis after vascular endothelial growth factor treatment was accompanied by increased expression of the adhesion protein N-cadherin, which mediates endothelial-pericytic interactions, in ischemic brain microvessels of wild-type mice on regular diet that was blunted in wild-type mice on Western diet and ApoE
−/−
mice on either diet.
Conclusions—
The compromised vessel formation and hemodynamics question the concept of therapeutic angiogenesis in ischemic stroke where hyperlipidemia is highly prevalent.
Collapse
Affiliation(s)
- Anil Zechariah
- From the Departments of Neurology (A.Z., F.J., N.H., T.R.D., A.E., D.M.H.), and Dermatology (I.H.), University Hospital Essen, Germany; and Max-Planck-Institute for Neurological Research, Cologne, Germany (G.M.)
| | - Ayman ElAli
- From the Departments of Neurology (A.Z., F.J., N.H., T.R.D., A.E., D.M.H.), and Dermatology (I.H.), University Hospital Essen, Germany; and Max-Planck-Institute for Neurological Research, Cologne, Germany (G.M.)
| | - Nina Hagemann
- From the Departments of Neurology (A.Z., F.J., N.H., T.R.D., A.E., D.M.H.), and Dermatology (I.H.), University Hospital Essen, Germany; and Max-Planck-Institute for Neurological Research, Cologne, Germany (G.M.)
| | - Fengyan Jin
- From the Departments of Neurology (A.Z., F.J., N.H., T.R.D., A.E., D.M.H.), and Dermatology (I.H.), University Hospital Essen, Germany; and Max-Planck-Institute for Neurological Research, Cologne, Germany (G.M.)
| | - Thorsten Roland Doeppner
- From the Departments of Neurology (A.Z., F.J., N.H., T.R.D., A.E., D.M.H.), and Dermatology (I.H.), University Hospital Essen, Germany; and Max-Planck-Institute for Neurological Research, Cologne, Germany (G.M.)
| | - Iris Helfrich
- From the Departments of Neurology (A.Z., F.J., N.H., T.R.D., A.E., D.M.H.), and Dermatology (I.H.), University Hospital Essen, Germany; and Max-Planck-Institute for Neurological Research, Cologne, Germany (G.M.)
| | - Günter Mies
- From the Departments of Neurology (A.Z., F.J., N.H., T.R.D., A.E., D.M.H.), and Dermatology (I.H.), University Hospital Essen, Germany; and Max-Planck-Institute for Neurological Research, Cologne, Germany (G.M.)
| | - Dirk Matthias Hermann
- From the Departments of Neurology (A.Z., F.J., N.H., T.R.D., A.E., D.M.H.), and Dermatology (I.H.), University Hospital Essen, Germany; and Max-Planck-Institute for Neurological Research, Cologne, Germany (G.M.)
| |
Collapse
|
40
|
Chang PY, Chen YJ, Chang FH, Lu J, Huang WH, Yang TC, Lee YT, Chang SF, Lu SC, Chen CH. Aspirin protects human coronary artery endothelial cells against atherogenic electronegative LDL via an epigenetic mechanism: a novel cytoprotective role of aspirin in acute myocardial infarction. Cardiovasc Res 2013; 99:137-45. [PMID: 23519265 DOI: 10.1093/cvr/cvt062] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
AIMS L5 is the most negatively charged subfraction of human low-density lipoprotein (LDL) and is the only subfraction of LDL capable of inducing apoptosis in cultured vascular endothelial cells (ECs) by inhibiting fibroblast growth factor-2 (FGF2) transcription. We examined whether plasma L5 levels are elevated in patients with ST-segment elevation myocardial infarction (STEMI) and whether aspirin provides epigenetic protection of human coronary artery ECs (HCAECs) exposed to L5. METHODS AND RESULTS Plasma L5 levels were compared between patients with STEMI (n = 10) and control subjects with chest pain syndrome but a normal coronary arteriogram (n = 5). L5 was isolated from the plasma of STEMI patients and control subjects, and apoptosis, FGF2 expression, and FGF2 promoter methylation were examined in HCAECs treated with L5 and aspirin. Plasma L5 levels were significantly higher in STEMI patients than in control subjects (P < 0.001). Treatment of HCAECs with L5 resulted in reduced survival and FGF2 expression and increased CpG methylation of the FGF2 promoter. Co-treatment of HCAECs with L5 and a physiologically relevant, low concentration of aspirin (0.2 mM) attenuated the adverse effects of L5 on HCAEC survival, FGF2 expression, and FGF2 promoter methylation. In contrast, high concentrations of aspirin (≥1.0 mM) accentuated the effects of L5. CONCLUSIONS Our results show that L5 levels are significantly increased in STEMI patients. Furthermore, L5 impairs HCAEC function through CpG methylation of the FGF2 promoter, which is suppressed in the presence of low-concentration aspirin. Our results provide evidence of a novel mechanism of aspirin in the prevention of MI.
Collapse
Affiliation(s)
- Po-Yuan Chang
- Department of Internal Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
LDL attenuates VEGF-induced angiogenesis via mechanisms involving VEGFR2 internalization and degradation following endosome-trans-Golgi network trafficking. Angiogenesis 2013; 16:625-37. [PMID: 23429999 DOI: 10.1007/s10456-013-9340-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2012] [Accepted: 02/11/2013] [Indexed: 01/15/2023]
Abstract
Considerable efforts have been made to amplify angiogenesis under conditions of hypoxia and ischemia by vascular endothelial growth factor (VEGF) delivery, so far with limited success. Ischemic vascular diseases are often associated with hypercholesterolemia. To elucidate whether the exposure to blood lipids influences VEGF responses of microvessels, we characterized effects of low density lipoprotein (LDL) exposure on the proliferation, migration and tube formation of human umbilical vein endothelial cells. By examining the expression, phosphorylation and downstream signals of VEGF's receptor VEGFR2, we characterized mechanisms controlling angiogenic responses following LDL exposure. LDL attenuated endothelial proliferation, migration and tube formation in a dose-dependent way. Reduced abundance of VEGFR2 and VEGFR1 were noticed in LDL-exposed endothelial cells. In subcellular localization studies that we combined with pharmacological experiments, we showed that the loss of VEGFR2 resulted from its internalization and degradation, the latter of which required syntaxin-16-dependent endosome-trans-Golgi network trafficking. As a consequence, VEGFR2 phosphorylation and downstream signals -specifically Akt and ERK1/2 phosphorylation- were attenuated in response to VEGF treatment. VEGF only partly reversed the effects of LDL on angiogenesis under conditions of normoxia and hypoxia. Our results suggest that angiogenic responses to VEGF are compromised in hypercholesterolemia as a consequence of endosomal VEGFR2 degradation.
Collapse
|
42
|
Reiss AB, Cronstein BN. Regulation of foam cells by adenosine. Arterioscler Thromb Vasc Biol 2012; 32:879-86. [PMID: 22423040 PMCID: PMC3306592 DOI: 10.1161/atvbaha.111.226878] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 12/02/2011] [Indexed: 12/20/2022]
Abstract
Macrophages rely on reverse cholesterol transport mechanisms to rid themselves of excess cholesterol. By reducing accumulation of cholesterol in the artery wall, reverse cholesterol transport slows or prevents development of atherosclerosis. In stable macrophages, efflux mechanisms balance influx mechanisms, and accumulating lipids do not overwhelm the cell. Under atherogenic conditions, inflow of cholesterol exceeds outflow, and the cell is ultimately transformed into a foam cell, the prototypical cell in the atherosclerotic plaque. Adenosine is an endogenous purine nucleoside released from metabolically active cells by facilitated diffusion and generated extracellularly from adenine nucleotides. Under stress conditions, such as hypoxia, a depressed cellular energy state leads to an acute increase in the extracellular concentration of adenosine. Extracellular adenosine interacts with 1 or more of a family of G protein-coupled receptors (A(1), A(2A), A(2B), and A(3)) to modulate the function of nearly all cells and tissues. Modulation of adenosine signaling participates in regulation of reverse cholesterol transport. Of particular note for the development of atherosclerosis, activation of A(2A) receptors dramatically inhibits inflammation and protects against tissue injury. Potent antiatherosclerotic effects of A(2A) receptor stimulation include inhibition of macrophage foam cell transformation and upregulation of the reverse cholesterol transport proteins cholesterol 27-hydroxylase and ATP binding cassette transporter A1. Thus, A(2A) receptor agonists may correct or prevent the adverse effects of inflammatory processes on cellular cholesterol homeostasis. This review focuses on the importance of extracellular adenosine acting at specific receptors as a regulatory mechanism to control the formation of foam cells under conditions of lipid loading.
Collapse
Affiliation(s)
- Allison B. Reiss
- Department of Medicine and Winthrop Research Institute, Winthrop-University Hospital, 222 Station Plaza North, Suite 502A, Mineola, NY 11501, USA
| | - Bruce N. Cronstein
- Division of Translational Medicine, Department of Medicine, New York University School of Medicine, NBV16N-1, 550 First Avenue, New York, NY, 10016, USA
| |
Collapse
|
43
|
Lu J, Mitra S, Wang X, Khaidakov M, Mehta JL. Oxidative stress and lectin-like ox-LDL-receptor LOX-1 in atherogenesis and tumorigenesis. Antioxid Redox Signal 2011; 15:2301-33. [PMID: 21338316 DOI: 10.1089/ars.2010.3792] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) has been identified as a major receptor for oxidized low-density lipoprotein (ox-LDL) in endothelial cells, monocytes, platelets, cardiomyocytes, and vascular smooth muscle cells. Its expression is minimal under physiological conditions but can be induced under pathological conditions. The upregulation of LOX-1 by ox-LDL appears to be important for physiologic processes, such as endothelial cell proliferation, apoptosis, and endothelium remodeling. Pathophysiologic effects of ox-LDL in atherogenesis have also been firmly established, including endothelial cell dysfunction, smooth muscle cell growth and migration, monocyte transformation into macrophages, and finally platelet aggregation-seen in atherogenesis. Recent studies show a positive correlation between increased serum ox-LDL levels and an increased risk of colon, breast, and ovarian cancer. As in atherosclerosis, ox-LDL and its receptor LOX-1 activate the inflammatory pathway through nuclear factor-kappa B, leading to cell transformation. LOX-1 is important for maintaining the transformed state in developmentally diverse cancer cell lines and for tumor growth, suggesting a molecular connection between atherogenesis and tumorigenesis.
Collapse
Affiliation(s)
- Jingjun Lu
- Cardiovascular Division, VA Medical Center, University of Arkansas for Medical Sciences, Little Rock, AR 72212, USA
| | | | | | | | | |
Collapse
|
44
|
|
45
|
Qiu J, Wang G, Zheng Y, Hu J, Peng Q, Yin T. Coordination of Id1 and p53 Activation by Oxidized LDL Regulates Endothelial Cell Proliferation and Migration. Ann Biomed Eng 2011; 39:2869-78. [DOI: 10.1007/s10439-011-0382-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2011] [Accepted: 08/08/2011] [Indexed: 10/17/2022]
|
46
|
McMahon M, Hahn BH, Skaggs BJ. Systemic lupus erythematosus and cardiovascular disease: prediction and potential for therapeutic intervention. Expert Rev Clin Immunol 2011; 7:227-41. [PMID: 21426260 DOI: 10.1586/eci.10.98] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Patients with systemic lupus erythematosus have a significantly increased risk of cardiovascular events due to atherosclerosis. Traditional cardiac risk factors cannot fully explain this increased risk. Recent evidence strongly suggests that atherosclerotic plaque is largely driven by inflammation and an active immunological response, in contrast to the long-held belief that plaque is a passive accumulation of lipids in the arterial wall. Current approaches to the prevention of atherosclerosis in systemic lupus erythematosus involve targeting modifiable cardiac risk factors. Future preventive strategies may include therapies that counteract the immunologic responses that lead to plaque formation.
Collapse
Affiliation(s)
- Maureen McMahon
- Division of Rheumatology, Department of Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA.
| | | | | |
Collapse
|
47
|
Togliatto G, Trombetta A, Dentelli P, Rosso A, Brizzi MF. MIR221/MIR222-driven post-transcriptional regulation of P27KIP1 and P57KIP2 is crucial for high-glucose- and AGE-mediated vascular cell damage. Diabetologia 2011; 54:1930. [PMID: 21461636 DOI: 10.1007/s00125-011-2125-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 02/23/2011] [Indexed: 11/30/2022]
Abstract
AIMS/HYPOTHESIS MicroRNAs (miRNAs) are a novel group of small non-coding RNAs that regulate gene expression at the post-transcriptional level and act on their target mRNAs in a tissue- and cell-type-specific manner. Herein, the relevance of MIR221/MIR222 in high-glucose- and AGE-mediated vascular damage was investigated. METHODS Functional studies were performed using human mature endothelial cells and endothelial progenitor cells subjected to high glucose or AGE. Quantitative real-time amplification was performed to analyse MIR221/MIR222 expression in these experimental conditions. Luciferase assay was used to identify MIR221/MIR222 targets. Functional studies were performed in vitro and in vivo in mice using gain- and loss-of-function approaches. RESULTS Using an in vivo mouse model we demonstrated that exposure to AGE and high glucose impaired vessel formation. Moreover, in vitro functional studies revealed that both high glucose and AGE inhibit cell-cycle progression by modulating the expression of P27KIP1 (also known as CDKN1B) and P57KIP2 (also known as CDKN1C), which encode cyclin-dependent kinase inhibitor 1B (p27, Kip1) (P27KIP1) and cyclin-dependent kinase inhibitor 1C (p57, Kip2) (P57KIP2), respectively. Crucial to AGE- and high-glucose-mediated cell-cycle arrest was the downregulation of MIR221/MIR222 expression. Luciferase assay showed that MIR221 and MIR222 specifically bind to the P27KIP1 and P57KIP2 mRNA 3'-untranslated regions, implicating P27KIP1 and P57KIP2 as MIR221/MIR222 targets. These results were confirmed by gain-of-function experiments in vitro, and by injecting mice with endothelial cells overexpressing MIR221 and MIR222. CONCLUSIONS/INTERPRETATION We provide evidence that high-glucose- and AGE-induced inhibition of vascular cell proliferation is controlled by MIR221/MIR222-driven post-transcriptional regulation of P27KIP1 and P57KIP2. These data add further insight to the possible contribution of miRNAs in vascular damage mediated by a high-glucose environment.
Collapse
Affiliation(s)
- G Togliatto
- Department of Internal Medicine, University of Torino, Corso Dogliotti 14, 10126, Torino, Italy
| | - A Trombetta
- Department of Internal Medicine, University of Torino, Corso Dogliotti 14, 10126, Torino, Italy
| | - P Dentelli
- Department of Internal Medicine, University of Torino, Corso Dogliotti 14, 10126, Torino, Italy
| | - A Rosso
- Department of Internal Medicine, University of Torino, Corso Dogliotti 14, 10126, Torino, Italy
| | - M F Brizzi
- Department of Internal Medicine, University of Torino, Corso Dogliotti 14, 10126, Torino, Italy.
| |
Collapse
|
48
|
Togliatto G, Trombetta A, Dentelli P, Rosso A, Brizzi MF. MIR221/MIR222-driven post-transcriptional regulation of P27KIP1 and P57KIP2 is crucial for high-glucose- and AGE-mediated vascular cell damage. Diabetologia 2011. [PMID: 21461636 DOI: 10.1007/s00125-011-2125 -5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
AIMS/HYPOTHESIS MicroRNAs (miRNAs) are a novel group of small non-coding RNAs that regulate gene expression at the post-transcriptional level and act on their target mRNAs in a tissue- and cell-type-specific manner. Herein, the relevance of MIR221/MIR222 in high-glucose- and AGE-mediated vascular damage was investigated. METHODS Functional studies were performed using human mature endothelial cells and endothelial progenitor cells subjected to high glucose or AGE. Quantitative real-time amplification was performed to analyse MIR221/MIR222 expression in these experimental conditions. Luciferase assay was used to identify MIR221/MIR222 targets. Functional studies were performed in vitro and in vivo in mice using gain- and loss-of-function approaches. RESULTS Using an in vivo mouse model we demonstrated that exposure to AGE and high glucose impaired vessel formation. Moreover, in vitro functional studies revealed that both high glucose and AGE inhibit cell-cycle progression by modulating the expression of P27KIP1 (also known as CDKN1B) and P57KIP2 (also known as CDKN1C), which encode cyclin-dependent kinase inhibitor 1B (p27, Kip1) (P27KIP1) and cyclin-dependent kinase inhibitor 1C (p57, Kip2) (P57KIP2), respectively. Crucial to AGE- and high-glucose-mediated cell-cycle arrest was the downregulation of MIR221/MIR222 expression. Luciferase assay showed that MIR221 and MIR222 specifically bind to the P27KIP1 and P57KIP2 mRNA 3'-untranslated regions, implicating P27KIP1 and P57KIP2 as MIR221/MIR222 targets. These results were confirmed by gain-of-function experiments in vitro, and by injecting mice with endothelial cells overexpressing MIR221 and MIR222. CONCLUSIONS/INTERPRETATION We provide evidence that high-glucose- and AGE-induced inhibition of vascular cell proliferation is controlled by MIR221/MIR222-driven post-transcriptional regulation of P27KIP1 and P57KIP2. These data add further insight to the possible contribution of miRNAs in vascular damage mediated by a high-glucose environment.
Collapse
Affiliation(s)
- G Togliatto
- Department of Internal Medicine, University of Torino, Corso Dogliotti 14, 10126, Torino, Italy
| | - A Trombetta
- Department of Internal Medicine, University of Torino, Corso Dogliotti 14, 10126, Torino, Italy
| | - P Dentelli
- Department of Internal Medicine, University of Torino, Corso Dogliotti 14, 10126, Torino, Italy
| | - A Rosso
- Department of Internal Medicine, University of Torino, Corso Dogliotti 14, 10126, Torino, Italy
| | - M F Brizzi
- Department of Internal Medicine, University of Torino, Corso Dogliotti 14, 10126, Torino, Italy.
| |
Collapse
|
49
|
Inhibition of melanoma angiogenesis by telomere homolog oligonucleotides. JOURNAL OF ONCOLOGY 2010; 2010:928628. [PMID: 20652008 PMCID: PMC2906154 DOI: 10.1155/2010/928628] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Revised: 05/06/2010] [Accepted: 05/07/2010] [Indexed: 01/13/2023]
Abstract
Telomere homolog oligonucleotides (T-oligos) activate an innate telomere-based program that leads to multiple anticancer effects. T-oligos act at telomeres to initiate signaling through the Werner protein and ATM kinase. We wanted to determine if T-oligos have antiangiogenic effects. We found that T-oligo-treated human melanoma (MM-AN) cells had decreased expression of vascular endothelial growth factor (VEGF), VEGF receptor 2, angiopoeitin-1 and -2 and decreased VEGF secretion. T-oligos activated the transcription factor E2F1 and inhibited the activity of the angiogenic transcription factor, HIF-1α. T-oligos inhibited EC tubulogenesis and total tumor microvascular density matrix invasion by MM-AN cells and ECs in vitro. In melanoma SCID xenografts, two systemic T-oligo injections decreased by 60% (P < .004) total tumor microvascular density and the functional vessels density by 80% (P < .002). These findings suggest that restriction of tumor angiogenesis is among the host's innate telomere-based anticancer responses and provide further evidence that T-oligos may offer a powerful new approach for melanoma treatment.
Collapse
|
50
|
Stasi MA, Scioli MG, Arcuri G, Mattera GG, Lombardo K, Marcellini M, Riccioni T, De Falco S, Pisano C, Spagnoli LG, Borsini F, Orlandi A. Propionyl-
l
-Carnitine Improves Postischemic Blood Flow Recovery and Arteriogenetic Revascularization and Reduces Endothelial NADPH-Oxidase 4–Mediated Superoxide Production. Arterioscler Thromb Vasc Biol 2010; 30:426-35. [DOI: 10.1161/atvbaha.109.201533] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
The beneficial effect of the natural compound propionyl-
l
-carnitine (PLC) on intermittent claudication in patients with peripheral arterial disease is attributed to its anaplerotic function in ischemic tissues, but inadequate information is available concerning action on the vasculature.
Methods and Results—
We investigated the effects of PLC in rabbit hind limb collateral vessels after femoral artery excision, mouse dorsal air pouch, chicken chorioallantoic membrane, and vascular cells by angiographic, Doppler flow, and histomorphometrical and biomolecular analyses. PLC injection accelerated hind limb blood flow recovery after 4 days (
P
<0.05) and increased angiographic quadriceps collateral vascularization after 7 days (
P
<0.001) Histomorphometry confirmed the increased vascular area (
P
<0.05), with unchanged intramuscular capillary density. PLC-induced dilatative adaptation, and growth was found associated with increased inducible nitric oxide synthase and reduced arterial vascular endothelial growth factor and intracellular adhesion molecule-1 expression. PLC also increased vascularization in air pouch and chorioallantoic membrane (
P
<0.05), particularly in large vessels. PLC increased endothelial and human umbilical vascular endothelial cell proliferation and rapidly reduced inducible nitric oxide synthase and NADPH-oxidase 4–mediated reactive oxygen species production in human umbilical vascular endothelial cells; NADPH-oxidase 4 also regulated NF-κB–independent intracellular adhesion molecule-1 expression.
Conclusion—
Our results provided strong evidence that PLC improves postischemic flow recovery and revascularization and reduces endothelial NADPH-oxidase–related superoxide production. We recommend that PLC should be included among therapeutic interventions that target endothelial function.
Collapse
Affiliation(s)
- Maria Antonietta Stasi
- From General Pharmacology and Oncology Department (M.A.S., G.G.M., K.L., M.M., T.R., C.P., F.B.) Sigma-Tau, Pomezia, Rome, Italy; Anatomic Pathology (M.G.S., L.G.S., A.O.), Tor Vergata University, Rome, Italy; Experimental Medicine and Biochemical Sciences (G.A.), Tor Vergata University of Rome, Italy; Institute of Genetics and Biophysics (S.D.F.), CNR, Napoli, Italy
| | - Maria Giovanna Scioli
- From General Pharmacology and Oncology Department (M.A.S., G.G.M., K.L., M.M., T.R., C.P., F.B.) Sigma-Tau, Pomezia, Rome, Italy; Anatomic Pathology (M.G.S., L.G.S., A.O.), Tor Vergata University, Rome, Italy; Experimental Medicine and Biochemical Sciences (G.A.), Tor Vergata University of Rome, Italy; Institute of Genetics and Biophysics (S.D.F.), CNR, Napoli, Italy
| | - Gaetano Arcuri
- From General Pharmacology and Oncology Department (M.A.S., G.G.M., K.L., M.M., T.R., C.P., F.B.) Sigma-Tau, Pomezia, Rome, Italy; Anatomic Pathology (M.G.S., L.G.S., A.O.), Tor Vergata University, Rome, Italy; Experimental Medicine and Biochemical Sciences (G.A.), Tor Vergata University of Rome, Italy; Institute of Genetics and Biophysics (S.D.F.), CNR, Napoli, Italy
| | - Giovan Giuseppe Mattera
- From General Pharmacology and Oncology Department (M.A.S., G.G.M., K.L., M.M., T.R., C.P., F.B.) Sigma-Tau, Pomezia, Rome, Italy; Anatomic Pathology (M.G.S., L.G.S., A.O.), Tor Vergata University, Rome, Italy; Experimental Medicine and Biochemical Sciences (G.A.), Tor Vergata University of Rome, Italy; Institute of Genetics and Biophysics (S.D.F.), CNR, Napoli, Italy
| | - Katia Lombardo
- From General Pharmacology and Oncology Department (M.A.S., G.G.M., K.L., M.M., T.R., C.P., F.B.) Sigma-Tau, Pomezia, Rome, Italy; Anatomic Pathology (M.G.S., L.G.S., A.O.), Tor Vergata University, Rome, Italy; Experimental Medicine and Biochemical Sciences (G.A.), Tor Vergata University of Rome, Italy; Institute of Genetics and Biophysics (S.D.F.), CNR, Napoli, Italy
| | - Marcella Marcellini
- From General Pharmacology and Oncology Department (M.A.S., G.G.M., K.L., M.M., T.R., C.P., F.B.) Sigma-Tau, Pomezia, Rome, Italy; Anatomic Pathology (M.G.S., L.G.S., A.O.), Tor Vergata University, Rome, Italy; Experimental Medicine and Biochemical Sciences (G.A.), Tor Vergata University of Rome, Italy; Institute of Genetics and Biophysics (S.D.F.), CNR, Napoli, Italy
| | - Teresa Riccioni
- From General Pharmacology and Oncology Department (M.A.S., G.G.M., K.L., M.M., T.R., C.P., F.B.) Sigma-Tau, Pomezia, Rome, Italy; Anatomic Pathology (M.G.S., L.G.S., A.O.), Tor Vergata University, Rome, Italy; Experimental Medicine and Biochemical Sciences (G.A.), Tor Vergata University of Rome, Italy; Institute of Genetics and Biophysics (S.D.F.), CNR, Napoli, Italy
| | - Sandro De Falco
- From General Pharmacology and Oncology Department (M.A.S., G.G.M., K.L., M.M., T.R., C.P., F.B.) Sigma-Tau, Pomezia, Rome, Italy; Anatomic Pathology (M.G.S., L.G.S., A.O.), Tor Vergata University, Rome, Italy; Experimental Medicine and Biochemical Sciences (G.A.), Tor Vergata University of Rome, Italy; Institute of Genetics and Biophysics (S.D.F.), CNR, Napoli, Italy
| | - Claudio Pisano
- From General Pharmacology and Oncology Department (M.A.S., G.G.M., K.L., M.M., T.R., C.P., F.B.) Sigma-Tau, Pomezia, Rome, Italy; Anatomic Pathology (M.G.S., L.G.S., A.O.), Tor Vergata University, Rome, Italy; Experimental Medicine and Biochemical Sciences (G.A.), Tor Vergata University of Rome, Italy; Institute of Genetics and Biophysics (S.D.F.), CNR, Napoli, Italy
| | - Luigi Giusto Spagnoli
- From General Pharmacology and Oncology Department (M.A.S., G.G.M., K.L., M.M., T.R., C.P., F.B.) Sigma-Tau, Pomezia, Rome, Italy; Anatomic Pathology (M.G.S., L.G.S., A.O.), Tor Vergata University, Rome, Italy; Experimental Medicine and Biochemical Sciences (G.A.), Tor Vergata University of Rome, Italy; Institute of Genetics and Biophysics (S.D.F.), CNR, Napoli, Italy
| | - Franco Borsini
- From General Pharmacology and Oncology Department (M.A.S., G.G.M., K.L., M.M., T.R., C.P., F.B.) Sigma-Tau, Pomezia, Rome, Italy; Anatomic Pathology (M.G.S., L.G.S., A.O.), Tor Vergata University, Rome, Italy; Experimental Medicine and Biochemical Sciences (G.A.), Tor Vergata University of Rome, Italy; Institute of Genetics and Biophysics (S.D.F.), CNR, Napoli, Italy
| | - Augusto Orlandi
- From General Pharmacology and Oncology Department (M.A.S., G.G.M., K.L., M.M., T.R., C.P., F.B.) Sigma-Tau, Pomezia, Rome, Italy; Anatomic Pathology (M.G.S., L.G.S., A.O.), Tor Vergata University, Rome, Italy; Experimental Medicine and Biochemical Sciences (G.A.), Tor Vergata University of Rome, Italy; Institute of Genetics and Biophysics (S.D.F.), CNR, Napoli, Italy
| |
Collapse
|