147 results on '"cardiac microvascular endothelial cells"'
Search Results
2. Engineered Biomimetic Nanoparticles-Mediated Targeting Delivery of Allicin Against Myocardial Ischemia-Reperfusion Injury by Inhibiting Ferroptosis
- Author
-
Li M, Wu J, Yang T, Zhao Y, Ren P, Chang L, Shi P, Yang J, Liu Y, Li X, Wang P, and Cao Y
- Subjects
neutrophil membrane ,ischemia-reperfusion ,allicin ,cardiac microvascular endothelial cells ,ferroptosis ,Medicine (General) ,R5-920 - Abstract
Minghui Li,1,* Jiabi Wu,1,* Tao Yang,1 Yuhang Zhao,1 Ping Ren,1 Lingling Chang,2 Pilong Shi,1 Jing Yang,1 Yuhang Liu,2 Xiaolei Li,3 Peng Wang,2 Yonggang Cao1 1Department of Pharmaceutics, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China; 2Department of Physiology, Harbin Medical University, Heilongjiang, 163319, People’s Republic of China; 3Department of Pathology, Jiangsu College of Nursing, Jiangsu, 223003, People’s Republic of China*These authors contributed equally to this workCorrespondence: Peng Wang, Department of Physiology, Harbin Medical University, Xinyang Road 39, Gaoxin District, Daqing, Heilongjiang, People’s Republic of China, Email wangpeng810122@126.com Yonggang Cao, Department of Pharmaceutics, Harbin Medical University, Xinyang Road 39, Gaoxin District, Daqing, Heilongjiang, People’s Republic of China, Email caoyonggang@hmudq.edu.cnBackground: Cardiac microvascular damage is substantially related with the onset of myocardial ischaemia-reperfusion (IR) injury. Reportedly, allicin (AL) effectively protects the cardiac microvascular system from IR injury. However, the unsatisfactory therapeutic efficacy of current drugs and insufficient drug delivery to the damaged heart are major concerns. Here, inspired by the natural interaction between neutrophils and inflamed cardiac microvascular endothelial cells (CMECs), a neutrophil membrane-camouflaged nanoparticle for non-invasive active-targeting therapy for IR injury by improving drug delivery to the injured heart is constructed.Methods: In this study, we engineered mesoporous silica nanoparticles (MSNs) coated with a neutrophil membrane to act as a drug delivery system, encapsulating AL. The potential of the nanoparticles (named AL@MSNs@NM) for specific targeting of infarcted myocardium was assessed using small animal vivo imaging system. The cardiac function of AL@MSNs@NM after treatment was evaluated by Animal Ultrasound Imaging system, HE staining, and Laser Speckle Imaging System. The therapeutic mechanism was analyzed by ELISA kits, immunofluorescence, and PCR.Results: We discovered that AL@MSNs@NM significantly improves cardiac function index, reduced infarct size and fibrosis, increased vascular perfusion in ischemic areas, and also promoted the function of CMECs, including migration, tube formation, shear stress adaptation, and nitric oxide production. Further research revealed that AL@MSNs@NM have cardio-protective functions in IR rats by inhibiting CMEC ferroptosis and increasing platelet endothelial cell adhesion molecule-1 (PECAM-1) expression.Conclusion: Our results indicated that AL@MSNs@NM significantly reversed CMEC ferroptosis and increased PECAM-1 expression, enhanced cardiac function, and reduced myocardial infarction size. Therefore, this strategy demonstrates that engineered biomimetic nanotechnology effectively delivers AL for targeted therapy of myocardial infarction.Keywords: neutrophil membrane, ischemia-reperfusion, allicin, cardiac microvascular endothelial cells, ferroptosis
- Published
- 2024
3. Endothelial identity not found – Beyond passage 38, commercial cardiac microvascular endothelial cells do not express CD31 and VE-cadherin
- Author
-
Sarah Hilderink, Jolanda van der Velden, and Diederik W.D. Kuster
- Subjects
Cardiac microvascular endothelial cells ,Validation ,Commercial cell lines ,Reproducibility ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Few immortalized cardiac microvascular endothelial cell (CMEC) lines are available, particularly mouse lines. We purchased the CLU510 mCMEC line (Cedarlane), isolated by fluorescence-activated cell sorting for CD31 and VE-cadherin. The cell line has been used in previous studies, although none report CD31 or VE-cadherin expression. We analyzed endothelial profile of two vials of passage 38 cells. CD31 and VE-cadherin mRNA were hardly expressed in mCMECs compared to primary mouse lung ECs. CD31 and VE-cadherin protein levels were also negligible compared to multiple EC lines. Thus, CLU510 mCMECs beyond P38 do not harbor an endothelial phenotype. Caution should be warranted when using commercial cells and journals should carefully consider the validity of results when essential characterization of cell lines is omitted.
- Published
- 2024
- Full Text
- View/download PDF
4. FOXO6 transcription inhibition of CTRP3 promotes OGD/R-triggered cardiac microvascular endothelial barrier disruption via SIRT1/Nrf2 signalling.
- Author
-
Sanfu Zheng, Yu Wang, Weixi Guo, and Hongyu Tan
- Abstract
Background: C1q/TNF-related protein 3 (CTRP3) has been clarified to display its protective roles in cardiac function. The current study is concentrated on exploring the impacts of CTRP3 on myocardial ischaemia. Materials and methods: Oxygen and glucose hypoxia/reoxygenation (OGD/R) model was constructed in human cardiac microvascular endothelial cells (HCMECs). Reverse transcription-quantitative polymerase chain reaction and western blot analysis of CTRP3 expression were conducted. CCK-8 assay was to estimate cell activity and lactate dehydrogenase (LDH) assay kit was to test LDH release. TUNEL assay and western blot were to judge apoptosis. Endothelial barrier function was detected by in vitro vascular permeability assay kit. Zonula occludens-1 (ZO-1) expression was evaluated by immunofluorescence assay. The interaction between CTRP3 promoter and Forkhead Box O6 (FOXO6) was predicted by JASPAR database and verified by chromatin immunoprecipitation and luciferase reporter assays. After OGD/R-induced HCMECs were co-transfected with CTRP3 overexpression and FOXO6 overexpression plasmids, the above functional experiments above were conducted again. Lastly, the expression of sirtuin 1 (SIRT1)/nuclear factor erythroid 2-related factor 2 (Nrf2) signalling-related proteins was examined by western blot. Results: CTRP3 was down-regulated in OGD/R-induced HCMECs. CTRP3 enhanced the viability and barrier integrity while reduced the apoptosis and permeability of OGD/R-insulted HCMECs. This process may be regulated by FOXO6 transcription. Also, FOXO6 inhibition-mediated CTRP3 up-regulation activated the SIRT1/Nrf2 signalling. Conclusions: FOXO6 transcription inhibition of CTRP3 promotes OGD/R-triggered cardiac microvascular endothelial barrier disruption via SIRT1/Nrf2 signalling. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. TLN1 synergizes with ITGA5 to ameliorate cardiac microvascular endothelial cell dysfunction.
- Author
-
Xianfeng Wang, Wenkai Mao, and Xiaofeng Ma
- Abstract
Background: The complex process of atherosclerosis is thought to begin with endothelial cell dysfunction, and advanced atherosclerosis is the underlying cause of coronary artery disease (CAD). Uncovering the underlying mechanisms of CAD-related endothelial cell injury may contribute to the treatment. Materials and methods: Cardiac microvascular endothelial cells (CMVECs) were treated with oxidised low-density lipoprotein (ox-LDL) to mimic an injury model. The involvement of Talin-1 (TLN1) and integrin alpha 5 (ITGA5) in the proliferation, apoptosis, angiogenesis, inflammatory response, and oxidative stress in CMVECs were assessed. Results: TLN1 overexpression assisted CMVECs in resistance to ox-LDL stimulation, with alleviated cell proliferation and angiogenesis, reduced apoptosis, inflammatory response, and oxidative stress. TLN1 overexpression triggered increased ITGA5, and ITGA5 knockdown reversed the effects of TLN1 overexpression on the abovementioned aspects. Together, TLN1 synergized with ITGA5 to ameliorate the dysfunction in CMVECs. Conclusions: This finding suggests their probable involvement in CAD, and increasing their levels is beneficial to disease relief. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Fatty acid β‐oxidation and mitochondrial fusion are involved in cardiac microvascular endothelial cell protection induced by glucagon receptor antagonism in diabetic mice.
- Author
-
Wang, Peng, Wei, Rui, Cui, Xiaona, Jiang, Zongzhe, Yang, Jin, Zu, Lingyun, and Hong, Tianpei
- Subjects
- *
GLUCAGON receptors , *ENDOTHELIAL cells , *FATTY acids , *CARNITINE palmitoyltransferase , *DIABETIC cardiomyopathy - Abstract
Introduction: The role of cardiac microvascular endothelial cells (CMECs) in diabetic cardiomyopathy is not fully understood. We aimed to investigate whether a glucagon receptor (GCGR) monoclonal antibody (mAb) ameliorated diabetic cardiomyopathy and clarify whether and how CMECs participated in the process. Research Design and Methods: The db/db mice were treated with GCGR mAb or immunoglobulin G (as control) for 4 weeks. Echocardiography was performed to evaluate cardiac function. Immunofluorescent staining was used to determine microvascular density. The proteomic signature in isolated primary CMECs was analyzed by using tandem mass tag‐based quantitative proteomic analysis. Some target proteins were verified by using western blot. Results: Compared with db/m mice, cardiac microvascular density and left ventricular diastolic function were significantly reduced in db/db mice, and this reduction was attenuated by GCGR mAb treatment. A total of 199 differentially expressed proteins were upregulated in db/db mice versus db/m mice and downregulated in GCGR mAb‐treated db/db mice versus db/db mice. The enrichment analysis demonstrated that fatty acid β‐oxidation and mitochondrial fusion were the key pathways. The changes of the related proteins carnitine palmitoyltransferase 1B, optic atrophy type 1, and mitofusin‐1 were further verified by using western blot. The levels of these three proteins were upregulated in db/db mice, whereas this upregulation was attenuated by GCGR mAb treatment. Conclusion: GCGR antagonism has a protective effect on CMECs and cardiac diastolic function in diabetic mice, and this beneficial effect may be mediated via inhibiting fatty acid β‐oxidation and mitochondrial fusion in CMECs. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
7. Cardiac endothelial ischemia/reperfusion injury-derived protein damage-associated molecular patterns disrupt the integrity of the endothelial barrier
- Author
-
Sarawut Kumphune, Porrnthanate Seenak, Nitchawat Paiyabhrom, Worawat Songjang, Panyupa Pankhong, Noppadon Jumroon, Siriwan Thaisakun, Narumon Phaonakrop, Sittiruk Roytrakul, Wachirawadee Malakul, Arunya Jiraviriyakul, and Nitirut Nernpermpisooth
- Subjects
Cardiac microvascular endothelial cells ,Damage-associated molecular patterns ,Ischemia reperfusion injury ,Endothelial nitric oxide synthase ,Vascular endothelial-cadherin ,Science (General) ,Q1-390 ,Social sciences (General) ,H1-99 - Abstract
Human cardiac microvascular endothelial cells (HCMECs) are sensitive to ischemia and vulnerable to damage during reperfusion. The release of damage-associated molecular patterns (DAMPs) during reperfusion induces additional tissue damage. The current study aimed to identify early protein DAMPs in human cardiac microvascular endothelial cells subjected to ischemia-reperfusion injury (IRI) using a proteomic approach and their effect on endothelial cell injury. HCMECs were subjected to 60 min of simulated ischemia and 6 h of reperfusion, which can cause lethal damage. DAMPs in the culture media were subjected to liquid chromatography-tandem mass spectrometry proteomic analysis. The cells were treated with endothelial IRI-derived DAMP medium for 24 h. Endothelial injury was assessed by measuring lactate dehydrogenase activity, morphological features, and the expression of endothelial cadherin, nitric oxide synthase (eNOS), and caveolin-1. The top two upregulated proteins, DNAJ homolog subfamily B member 11 and pyrroline-5-carboxylate reductase 2, are promising and sensitive predictors of cardiac microvascular endothelial damage. HCMECs expose to endothelial IRI-derived DAMP, the lactate dehydrogenase activity was significantly increased compared with the control group (10.15 ± 1.03 vs 17.67 ± 1.19, respectively). Following treatment with endothelial IRI-derived DAMPs, actin-filament dysregulation, and downregulation of vascular endothelial cadherin, caveolin-1, and eNOS expressions were observed, along with cell death. In conclusion, the early protein DAMPs released during cardiac microvascular endothelial IRI could serve as novel candidate biomarkers for acute myocardial IRI. Distinct features of impaired plasma membrane integrity can help identify therapeutic targets to mitigate the detrimental consequences mediated of endothelial IRI-derived DAMPs.
- Published
- 2024
- Full Text
- View/download PDF
8. Diosmetin‐7‐O‐β‐D‐glucopyranoside suppresses endothelial–mesenchymal transformation through endoplasmic reticulum stress in cardiac fibrosis.
- Author
-
Wang, Huahua, Zhang, Xiaoyu, Liu, Yangyang, Zhang, Yunyun, Wang, Yingyu, Peng, Yunru, and Ding, Yongfang
- Subjects
- *
HEART fibrosis , *ENDOPLASMIC reticulum , *UNFOLDED protein response , *GLUCOSE-regulated proteins , *FLAVONOID glycosides , *TRANSMISSION electron microscopy , *PATHOLOGICAL physiology - Abstract
Diosmetin‐7‐O‐β‐D‐glucopyranoside (Diosmetin‐7‐O‐glucoside) is a natural flavonoid glycoside known to have a therapeutic application for cardiovascular diseases. Cardiac fibrosis is the main pathological change in the end stage of cardiovascular diseases. Endothelial‐mesenchymal transformation (EndMT) induced by endoplasmic reticulum stress (ER stress) via Src pathways is involved in the process of cardiac fibrosis. However, it is unclear whether and how diosmetin‐7‐O‐glucoside regulates EndMT and ER stress to treat cardiac fibrosis. In this study, molecular docking results showed that diosmetin‐7‐O‐glucoside bound well to ER stress and Src pathway markers. Diosmetin‐7‐O‐glucoside suppressed cardiac fibrosis induced by isoprenaline (ISO) and reduced the levels of EndMT, ER stress in mice heart. Primary cardiac microvascular endothelial cells (CMECs) were induced by transforming growth factor‐β1 (TGF‐β1) to perform EndMT. Diosmetin‐7‐O‐glucoside could effectively regulate EndMT and diminish the accumulation of collagen I and collagen III. We also showed that the tube formation in CMECs was restored, and the capacity of migration was partially inhibited. Diosmetin‐7‐O‐glucoside also ameliorated ER stress through the three unfolded protein response branches, as evidenced by organelle structure in transmission electron microscopy images and the expression of protein biomarkers like the glucose‐regulated protein 78 (GRP78) and C/EBP homologous protein (CHOP). Further analysis showed that diosmetin‐7‐O‐glucoside could suppress the expression level of Src phosphorylation, then block EndMT with the maintenance of endothelial appearance and endothelial marker expression. These results suggested that the diosmetin‐7‐O‐glucoside can regulate EndMT through ER stress, at least in part via Src‐dependent pathways. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
9. Association of circulating MtDNA with CVD in hemodialysis patients and in vitro effect of exogenous MtDNA on cardiac microvascular inflammation
- Author
-
Zhen Fan, Ya Feng, Li Zang, Yi Guo, and Xiao-yi Zhong
- Subjects
Maintenance hemodialysis ,Cardiovascular disease ,Cardiac microvascular endothelial cells ,Mitochondrial DNA ,Inflammation ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Abstract Background Chronic kidney disease (CKD) patients sustain a fairly high prevalence of cardiovascular disease (CVD). Microvascular inflammation is an early manifestation of CVD, and the released mitochondrial DNA (MtDNA) has been proposed to be a crucial integrator of inflammatory signals. Herein, the aim of this study was to determine the relationship between CVD, microvessel, and circulating MtDNA in the settings of uremia. Methods Forty-two maintenance hemodialysis (MHD) patients and 36 health controls were enrolled in this study. Plasma cell-free MtDNA was detected by TaqMan-based qPCR assay. CVD risk markers including high-sensitive C-reactive protein (Hs-CRP), monocyte chemoattractant protein-1 (MCP-1), fibrinogen, and erythrocyte sedimentation rate (ESR) were measured by standard assays. Ten-year CVD risk was calculated from the Framingham risk score (FRS) model. In vitro study, human cardiac microvascular endothelial cells (HCMECs) were incubated with normal or uremic serum, with or without exogenous MtDNA. Intracellular toll-like receptor 9 (TLR9), adhesion molecule 1 (ICAM-1), MCP-1 and tumor necrosis factor-α (TNF-α) and cytosolic MtDNA were detected by qPCR. Results Plasma MtDNA in MHD patients was significantly higher than healthy controls (4.74 vs. 2.41 × 105 copies/mL; p = 0.000). Subsequently, the MHD patients were classified into two groups based on the MtDNA median (4.34 × 105 copies/mL). In stratified analyses, the levels of Hs-CRP (5.02 vs. 3.73 mg/L; p = 0.042) and MCP-l (99.97 vs. 64.72 pg/mL; p = 0.008) and FRS (21.80 vs. 16.52; p = 0.016) in the high plasma MtDNA group were higher than those in the low plasma MtDNA group. In vitro study, we found that exogenous MtDNA aggravated uremic serum-induced microvascular inflammation (ICAM-1 and TNF-α) in HCMECs (all p
- Published
- 2023
- Full Text
- View/download PDF
10. 富马酸二甲酯对过氧化氢诱导心脏微血管 内皮细胞铁死亡的影响.
- Author
-
王赛 and 高静
- Abstract
Objective To explore the effects of dimethyl fumarate (DMF) on hydrogen peroxide (H2O2) - induced ferroptosis of human cardiac microvascular endothelial cells (HCMEC) and its potential mechanism. Methods HCMEC were cultured in vitro, incubated with H2O2, and then treated with different concentrations of DMF. Cell proliferation was measured by CCK - 8 method. The level of cell inflammation was detected by ELISA. Western blot was used to detect the changes of ferroptosis-related proteins GPX4 and ACSL4, as well as Nrf2/HO-1 and JAK2/STAT1 pathway proteins. Results The results of CCK8 assay showed that compared those in the control group, H2O2 significantly inhibited the proliferation of HCMEC cells, whereas, DMF treatment promoted H2O2 - induced cell proliferation in a concentration dependent manner (P < 0.05 for all). H2O2-induced increased levels of inflammatory cytokines TNF-α, IL-1β and IL-18 in HCMEC cells were reversed by DMF treatment (P < 0.05). Western blot revealed that H2O2 treatment caused the decrease in GPX4 protein and increase in ACSL4 protein; whereas DMF inhibited ferroptosis of HCMEC cells induced by H2O2 (P < 0.05). In addition, H2O2 induced a decrease in Nrf2 and HO-1 expression, while an increase in JAK2 and STAT1 phosphorylation levels in HCMEC (P < 0.05). Compared with those in the H2O2 group, DMF increased Nrf2 and HO-1 proteins and down-regulated the phosphorylation levels of JAK2 and STAT1 (P < 0.05). Finally, it was found that Nrf2 inhibitor ML385 significantly weakened the inhibition of ferroptosis and the protective effect of HCMEC damage. Conclusion DMF inhib- its H2O2 -induced inflammation of HCMEC and ferroptosis. Nrf2/HO-1 and JAK2/STAT1 pathways may play a key role in DMF-mediated protection of HCMEC. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
11. Association of circulating MtDNA with CVD in hemodialysis patients and in vitro effect of exogenous MtDNA on cardiac microvascular inflammation.
- Author
-
Fan, Zhen, Feng, Ya, Zang, Li, Guo, Yi, and Zhong, Xiao-yi
- Subjects
MITOCHONDRIAL DNA ,HEMODIALYSIS patients ,DISEASE risk factors ,BLOOD sedimentation ,CHRONIC kidney failure - Abstract
Background: Chronic kidney disease (CKD) patients sustain a fairly high prevalence of cardiovascular disease (CVD). Microvascular inflammation is an early manifestation of CVD, and the released mitochondrial DNA (MtDNA) has been proposed to be a crucial integrator of inflammatory signals. Herein, the aim of this study was to determine the relationship between CVD, microvessel, and circulating MtDNA in the settings of uremia. Methods: Forty-two maintenance hemodialysis (MHD) patients and 36 health controls were enrolled in this study. Plasma cell-free MtDNA was detected by TaqMan-based qPCR assay. CVD risk markers including high-sensitive C-reactive protein (Hs-CRP), monocyte chemoattractant protein-1 (MCP-1), fibrinogen, and erythrocyte sedimentation rate (ESR) were measured by standard assays. Ten-year CVD risk was calculated from the Framingham risk score (FRS) model. In vitro study, human cardiac microvascular endothelial cells (HCMECs) were incubated with normal or uremic serum, with or without exogenous MtDNA. Intracellular toll-like receptor 9 (TLR9), adhesion molecule 1 (ICAM-1), MCP-1 and tumor necrosis factor-α (TNF-α) and cytosolic MtDNA were detected by qPCR. Results: Plasma MtDNA in MHD patients was significantly higher than healthy controls (4.74 vs. 2.41 × 10
5 copies/mL; p = 0.000). Subsequently, the MHD patients were classified into two groups based on the MtDNA median (4.34 × 105 copies/mL). In stratified analyses, the levels of Hs-CRP (5.02 vs. 3.73 mg/L; p = 0.042) and MCP-l (99.97 vs. 64.72 pg/mL; p = 0.008) and FRS (21.80 vs. 16.52; p = 0.016) in the high plasma MtDNA group were higher than those in the low plasma MtDNA group. In vitro study, we found that exogenous MtDNA aggravated uremic serum-induced microvascular inflammation (ICAM-1 and TNF-α) in HCMECs (all p < 0.05). Besides, the addition of MtDNA to the medium resulted in a further increase in cytosolic MtDNA and TLR9 levels in uremic serum-treated cells (all p < 0.05). In patients with MHD, MtDNA levels in plasma were significantly reduced after a single routine hemodialysis (pre 4.47 vs. post 3.45 × 105 copies/mL; p = 0.001) or hemodiafiltration (pre 4.85 vs. post 4.09 × 105 copies/mL; p = 0.001). These two approaches seem similar in terms of MtDNA clearance rate (21.26% vs. 11.94%; p = 0.172). Conclusions: Overall, the present study suggests that MtDNA released into the circulation under the uremic toxin environment may adversely affect the cardiovascular system by exacerbating microvascular inflammation, and that reducing circulating MtDNA might be a future therapeutic strategy for the prevention of MHD-related CVD. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
12. Role of Mydgf in the regulation of hypoxia/reoxygenation-induced apoptosis in cardiac microvascular endothelial cells.
- Author
-
Wang, Yan, Zhang, Yu, Li, Jiao, Zhao, Ranzun, Long, Xianping, Li, Chaofu, Liu, Weiwei, Chen, Wenming, and Shi, Bei
- Abstract
We aimed to explore the effects of myeloid-derived growth factor (Mydgf) on the regulation of hypoxia/reoxygenation (HR)–induced apoptosis of cardiac microvascular endothelial cells (CMECs). CMECs were exposed to hypoxia for 24 h and reoxygenation for 6 h to establish an HR cell model. Subsequently, an adenovirus was used to overexpress Mydgf in CMECs. Flow cytometry and TUNEL staining were used to detect the extent of apoptosis, whereas qPCR was used to detect the relative expression of Mydgf mRNA. Western blotting was also performed to detect the expression of apoptosis-related proteins and endoplasmic reticulum stress (ERS)–related proteins, including C/EBP Homologous Protein (CHOP), glucose-regulated protein 78 (GRP 78), and cleaved Caspase-12. The endoplasmic reticulum stress agonist tunicamycin (TM) was used to stimulate CMECs for 24 h as a rescue experiment for Mydgf. Flow cytometry revealed that the HR model effectively induced endothelial cell apoptosis, whereas qPCR and western blotting showed that Mydgf mRNA and protein levels decreased significantly after HR treatment (P < 0.05). Overexpression of Mydgf in cells effectively reduced apoptosis after HR. Furthermore, western blotting showed that HR induced a significant upregulation of CHOP, GRP78, and cleaved-Caspase-12 expression in CMECs, whereas HR-treated cells downregulated the expression of CHOP, GRP78, and cleaved-Caspase-12 after Mydgf overexpression. Under HR conditions, TM significantly reversed the protective effect of Mydgf on CMECs. Mydgf may reduce CMEC apoptosis induced by HR by regulating oxidative stress in ERS. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
13. Elabela alleviates ferroptosis, myocardial remodeling, fibrosis and heart dysfunction in hypertensive mice by modulating the IL-6/STAT3/GPX4 signaling.
- Author
-
Zhang, Zhenzhou, Tang, Jianqiong, Song, Jiawei, Xie, Mengshi, Liu, Ying, Dong, Zhaojie, Liu, Xiaoyan, Li, Xueting, Zhang, Miwen, Chen, Yihang, Shi, Hongyu, and Zhong, Jiuchang
- Subjects
- *
HEART diseases , *HEART fibrosis , *HYPERTENSION , *SYSTOLIC blood pressure , *CARDIAC hypertrophy , *GLUTATHIONE peroxidase - Abstract
Hypertension-mediated pathological cardiac remodeling often progresses to heart failure. Elabela, mainly expressed in the cardiac microvascular endothelial cells (CMVECs), functions as a new endogenous ligand for apelin receptor. However, the exact roles of elabela in hypertension remain largely unclear. In this study, 10-week-old male C57BL/6 mice were randomly subjected to infusion of angiotensin (Ang) II (1.5 mg/kg/d) or saline for 2 weeks. Ang II infusion led to marked increases in systolic blood pressure levels and reduction of elabela levels in hypertensive mice with augmented myocardial hypertrophy and fibrosis. Furthermore, administration of elabela or ferroptosis inhibitor ferrostatin-1 significantly prevented Ang II-mediated pathological myocardial remodeling, dysfunction, and ultrastructural injury in hypertensive mice with downregulated expression of inflammation-, hypertrophy-, and fibrosis-related genes. Notably, elabela strikingly alleviated Ang II-induced upregulation of iron levels and lipid peroxidation in hypertensive mice by suppressing cardiac interleukin-6 (IL-6)/STAT3 signaling and activating the xCT/glutathione peroxidase (GPX4) signaling. In cultured CMVECs, exposure to Ang II resulted in a marked decrease in elabela levels and obvious increases in cellular ferroptosis, proliferation, inflammation, and superoxide production, which were rescued by elabela or ferrostatin-1 while were blocked by co-treatment with rhIL-6. Furthermore, knockdown of elabela by siRNA in CMVECs contributed to Ang II-mediated augmentations in cellular proliferation, migration, and oxidative stress in cultured cardiac fibroblasts and cardiomyocytes, respectively. In conclusion, elabela antagonizes Ang II-mediated promotion of CMVECs ferroptosis, adverse myocardial remodeling, fibrosis and heart dysfunction through modulating the IL-6/STAT3/GPX4 signaling pathway. Targeting elabela-APJ axis serves as a novel strategy for hypertensive heart diseases. [Display omitted] • Elabela and Fer-1 blunt Ang II-mediated promotion of lipid peroxidation and ROS generation in hypertensive mice and CMVECs. • Elabela alleviates cardiac ferroptosis and inflammation in hypertensive mice by modulating the IL-6/STAT3/GPX4 signaling. • Enhanced CMVEC ferroptosis contributes to Ang II-induced increases in proliferation and oxidative injury in CFs and CMs. • Targeting the Elabela-APJ axis is a promising therapy for hypertension and hypertensive heart diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
14. Overexpression of microRNA-136-3p Alleviates Myocardial Injury in Coronary Artery Disease via the Rho A/ROCK Signaling Pathway
- Author
-
Yongbo Lin, Hanliang Dan, and Jinguo Lu
- Subjects
coronary artery disease ,microrna-136-3p ,cardiac microvascular endothelial cells ,eif5a2 ,rho a/rock signaling pathway ,Dermatology ,RL1-803 ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 ,Diseases of the genitourinary system. Urology ,RC870-923 - Abstract
Objective: Coronary artery disease (CAD) is a cardiovascular disease that poses a fatal threat to human health, and the identification of potential biomarkers may help to delineate its pathophysiological mechanisms. Accumulating evidence has implicated microRNAs (miRNAs) in the pathogenesis and development of cardiovascular diseases. The present study aims to identify the expression of miRNA-136-3p (miR-136-3p) in CAD and further investigate its functional relevance in myocardial injury both in vitro and in vivo. Methods: Initially, CAD models were induced in rats by high-fat diet and intraperitoneal injection of pituitrin. Next, the effect of overexpressed miR-136-3p on cardiac function and pathological damage in myocardial tissue, cardiomyocyte apoptosis, oxidative stress and inflammatory response were assessed in CAD rats. Rat cardiac microvascular endothelial cells (CMECs) were isolated and cultured by the tissue explant method, and the CMEC injury model was induced by homocysteine (HCY). The function of miR-136-3p in vitro was further evaluated. Results: miR-136-3p was poorly expressed in the myocardial tissue of CAD rats and CMEC injury models. In vivo assays indicated that overexpressed miR-136-3p could improve cardiac function and alleviate pathological damage in myocardial tissue, accompanied by reduced oxidative stress and inflammatory response. Moreover,in vitro assays suggested that overexpression of miR-136-3p enhanced proliferation and migration while inhibiting apoptosis of HCY-stressed CMECs. Notably, we revealed that EIF5A2 was a target gene of miR-136-3p, and miR-136-3p inhibited EIF5A2 expression and activation of the Rho A/ROCK signaling pathway. Conclusion: In conclusion, the overexpression of miR-136-3p could potentially impede myocardial injury in vitro and in vivo in CAD through the blockade of the Rho A/ROCK signaling pathway, highlighting a potential miR-136-3p functional relevance in the treatment of CAD.
- Published
- 2020
- Full Text
- View/download PDF
15. Advanced glycation end products facilitate the proliferation and reduce early apoptosis of cardiac microvascular endothelial cells via PKCβ signaling pathway: Insight from diabetic cardiomyopathy
- Author
-
Yi Luan, Jiefang Zhang, Min Wang, Guosheng Fu, and Wenbin Zhang
- Subjects
advanced glycation end products ,cardiac microvascular endothelial cells ,diabetic cardiomyopathy ,protein kinase c ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Objective: To investigate the effects of advanced glycation end products (AGEs) on the proliferation and apoptosis of cardiac microvascular endothelial cells (CMECs) in rats and their underlying signaling pathway. Methods: CMECs were isolated from Sprague–Dawley rats. We first examined the effects of AGEs on the proliferation and apoptosis of CMECs and then tested whether protein kinase C (PKC) β blockers could counteract the effects of AGEs. The PKC agonists phorbol 12-myristate 13-acetate (PMA) and PKCβ blockers were also used to verify whether PKC could act independently on CMECs. The receptor for AGEs (RAGE)–small interfering RNA (siRNA) transfection was used to verify the effect of AGEs on PKC. Following the above steps, we explained whether AGEs regulated the CMEC proliferation and early apoptosis through the PKCβ signaling pathway. Proliferation of CMECs was detected using the Cell Counting Kit-8 (CCK-8) assay, and early apoptosis was determined using the Annexin V- Fluorescein Isothiocyanate (FITC)/propidium iodide (PI) double staining. Expression of proliferation and apoptosis-related proteins and PKC phosphorylation were determined by western blotting analysis. Cell cycle distributions were assayed using a BD FACSCalibur cell-sorting system. Results: AGEs facilitated the proliferation of CMECs, upregulated phosphorylated extracellular signal regulated kinase (p-ERK), and accelerated the entry of cells from G1 phase to the S+G2/M phase, which was consistent with the upregulated cyclin D1 by AGEs. AGEs inhibited early apoptosis of CMECs by increasing the expression of survivin and decreasing the expression of cleaved-caspase3. All these effects can be reversed by PKCβ1/2inhibitors. In addition, AGE upregulated the RAGE expression and phosphorylation of PKCβ1/2 in CMECs, while the inhibition of RAGE reversed the phosphorylation, as well as the effects of AGEs on proliferation and apoptosis in CMECs. Conclusion: The study indicated that AGEs facilitated the proliferation and reduced early apoptosis of CMECs via the PKCβ signaling pathway.
- Published
- 2020
- Full Text
- View/download PDF
16. Cardiac microvascular functions improved by MSC-derived exosomes attenuate cardiac fibrosis after ischemia–reperfusion via PDGFR-β modulation.
- Author
-
Wang, Xueqing, Bai, Long, Liu, Xinxin, Shen, Wenqian, Tian, Hai, Liu, Wei, and Yu, Bo
- Subjects
- *
MYOCARDIAL reperfusion , *HEART fibrosis , *EXOSOMES , *PLATELET-derived growth factor , *FIBROSIS , *MICROCIRCULATION disorders , *HEART failure - Abstract
Microvascular dysfunction caused by cardiac ischemia–reperfusion (I/R) leads to multiple severe cardiac adverse events, such as heart failure and ventricular modeling, which plays a critical role in outcomes. Though marrow mesenchymal stem cell (MSC) therapy has been proven effective for attenuating I/R injury, the limitations of clinical feasibility cannot be ignored. Since exosomes are recognized as the main vehicles for MSCs paracrine effects, we assumed that MSC-derived exosomes could prevent microvascular dysfunction and further protect cardiac function. By establishing a rat cardiac I/R model in vivo and a cardiac microvascular endothelial cells (CMECs) hypoxia–reperfusion (H/R) model in vitro, we demonstrated that MSC-derived exosomes enhanced microvascular regeneration under stress, inhibited fibrosis development, and eventually improved cardiac function through platelet-derived growth factor receptor-β (PDGFR-β) modulation. Furthermore, we found that MSC-derived exosomes possessed better therapeutic effects than MSCs themselves. • MSC-derived exosomes improve viability and regeneration of injured cardiac microvascular endothelial cells in vitro • MSC-derived exosomes exert better therapeutic effects than MSCs themselves for cardiac ischemia–reperfusion • MSC-derived exosomes maintain microvascular and long-term cardiac functions after I/R injury via PDGFR-β modulation [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
17. Mechanism of exosomes released from bone marrow stem cells regulating proliferation of cardiac microvascular endothelial cells
- Author
-
LI Chaofu, WANG Yan, ZHAO Ranzun, LONG Xianping, ZHANG Wei, CHEN Panke, and SHI Bei
- Subjects
bone marrow mesenchymal stem cells ,exosomes ,cardiac microvascular endothelial cells ,cell proliferation ,cell differentiation ,Medicine (General) ,R5-920 - Abstract
Objective To investigate the mechanism of exosomes released from bone marrow stem cells (BMSCs) on the proliferation in cardiac microvascular endothelial cells (CMVECs) by miR-214 regulation. Methods Rat CMVECs were cultured by enzyme digestion combined with differential adherence. Ultra-high speed centrifugation was used to extract exosomes from normal-oxygen or hypoxic-pretreated BMSCs. Immunofluorescence assay was employed to track the internalization of exosomes in CMVECs. Then the BMSCs were divided into 4 groups: ①control group, ②free exosomes (Free-Exos), ③normal exosomes (Nor-Exos), and ④hypoxia exosomes (Hypoxia-Exos, 400 μg/μL exosome co-cultured with CMVECs for 24 h). The cell proliferation was measured by Edu assay and flow cytometry. The miR-214 levels in exosomes or CMVECs were assessed using qRT-PCR, and the levels were measured again after miR-214 in exosomes were inhibited. And the cell proliferation was observed again by the above methods. Finally, after liposome 2000 was employed to transfect miR-214 mimics into CMVECs, the cell proliferation ability was measured by the Edu assay. Results Transmission electron microscopy and immunoblotting indicated that the extracted vesicles were exosomes. Immunofluorescence assay revealed that DiI-labeled exosomes could be internalized by CMVECs. Edu assay and cell cycle assay showed that Hypoxic-Exos had stronger potential to promote cell proliferation than Nor-Exos in CMVECs. qRT-PCR results showed that miR-214 was highly expressed in Hypoxic-Exos compared with Nor-Exos (P < 0.05). In CMVECs, the expression of miR-214 in Hypoxia-Exos group was significantly higher than that in Control group and Nor-Exos group (P < 0.05). In addition, Hypoxia-Exos's ability to promote cell proliferation was partially blocked by down-regulation of miR-214 in Hypoxia-Exos. However, after overexpression of mir-214 in CMVECs, cell proliferation ability was enhanced. Conclusion Exosomes released from hypoxic-pretreated BMSCs can promote CMVECs proliferation by medicating miR-214.
- Published
- 2019
- Full Text
- View/download PDF
18. miR-543 in human mesenchymal stem cell-derived exosomes promotes cardiac microvascular endothelial cell angiogenesis after myocardial infarction through COL4A1.
- Author
-
Mei Yang, Xueting Liu, Manli Jiang, Jian Li, Yaping Tang, and Lin Zhou
- Subjects
- *
ENDOTHELIAL cells , *CELL migration , *EXOSOMES , *NEOVASCULARIZATION , *HUMAN stem cells , *MESENCHYMAL stem cells , *ANIMAL disease models - Abstract
To explore the impact and mechanism of human mesenchymal stem cells (hMSCs) on the angiogenesis of cardiac microvascular endothelial cells (CMECs) after ischemia insult. Exosomes derived from hMSCs (hMSCs-Exo) were identified by Western blotting and labeled by PHK-67. CMECs were isolated from rat myocardial tissues. After hypoxic treatment, CMECs were cultured with hMSCs and exosome inhibitor (GW4869) or transfected with si-COL4A1 + miR-543 inhibitor. CMEC proliferation, migration, invasion, and angiogenesis were examined. Target genes of miR-543 were predicted and then were identified by dual luciferase assay. Myocardial infarction (MI) rat model established by suture occlusion was intravenously injected with hMSCs-Exo. Fluorescence microscope was applied to visualize exosomes in myocardial tissues. Infarction volume and pathologies of myocardial tissues were observed. Ki-67 and miR-543 expressions were detected. The isolated hMSC-Exo expressed TSG101, HSP70, and CD63. Hypoxia-treated CMECs cultured with hMSCs exhibited high proliferation, migration, invasion, and angiogenesis ability, while incubation with exosome inhibitor GW4969 offset the promoting effects of hMSCs on the proliferation, migration, invasion, and angiogenesis of CMECs. hMSCs transfected with miR-543 inhibitor brought CMECs weak viability and angiogenesis ability. CMECs transfected with si-COL4A1 and miR-543 inhibitor showed low proliferation, migration, invasion, and angiogenesis compared to those transfected with si-COL4A1 alone. hMSCs-Exo entered the myocardial tissues of MI rats. Injection of hMSCs-Exo in MI rats diminished infarction size, attenuated MI-induced injuries, and increased Ki-67 expression. hMSCs-Exo facilitates the proliferation, migration, invasion, and angiogenesis of CMECs through transferring miR-543 and downregulating COL4A1 expression. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
19. Overexpression of microRNA-136-3p Alleviates Myocardial Injury in Coronary Artery Disease via the Rho A/ROCK Signaling Pathway.
- Author
-
Lin, Yongbo, Dan, Hanliang, and Lu, Jinguo
- Subjects
- *
CORONARY disease , *TISSUE culture , *CARDIOVASCULAR diseases , *WOUNDS & injuries , *HIGH-fat diet , *ENDOTHELIAL cells - Abstract
Objective: Coronary artery disease (CAD) is a cardiovascular disease that poses a fatal threat to human health, and the identification of potential biomarkers may help to delineate its pathophysiological mechanisms. Accumulating evidence has implicated microRNAs (miRNAs) in the pathogenesis and development of cardiovascular diseases. The present study aims to identify the expression of miRNA-136-3p (miR-136-3p) in CAD and further investigate its functional relevance in myocardial injury both in vitro and in vivo. Methods: Initially, CAD models were induced in rats by high-fat diet and intraperitoneal injection of pituitrin. Next, the effect of overexpressed miR-136-3p on cardiac function and pathological damage in myocardial tissue, cardiomyocyte apoptosis, oxidative stress and inflammatory response were assessed in CAD rats. Rat cardiac microvascular endothelial cells (CMECs) were isolated and cultured by the tissue explant method, and the CMEC injury model was induced by homocysteine (HCY). The function of miR-136-3p in vitro was further evaluated. Results: miR-136-3p was poorly expressed in the myocardial tissue of CAD rats and CMEC injury models. In vivo assays indicated that overexpressed miR-136-3p could improve cardiac function and alleviate pathological damage in myocardial tissue, accompanied by reduced oxidative stress and inflammatory response. Moreover,in vitro assays suggested that overexpression of miR-136-3p enhanced proliferation and migration while inhibiting apoptosis of HCY-stressed CMECs. Notably, we revealed that EIF5A2 was a target gene of miR-136-3p, and miR-136-3p inhibited EIF5A2 expression and activation of the Rho A/ROCK signaling pathway. Conclusion: In conclusion, the overexpression of miR-136-3p could potentially impede myocardial injury in vitro and in vivo in CAD through the blockade of the Rho A/ROCK signaling pathway, highlighting a potential miR-136-3p functional relevance in the treatment of CAD. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
20. Upregulation of microRNA‐218 reduces cardiac microvascular endothelial cells injury induced by coronary artery disease through the inhibition of HMGB1.
- Author
-
Gao, Wenhui, Cui, Hanbin, Li, Qianjun, Zhong, Hai, Yu, Jingjing, Li, Ping, and He, Xijie
- Subjects
- *
CORONARY disease , *ENDOTHELIAL cells , *VASCULAR endothelial growth factors , *WESTERN immunoblotting , *TROPONIN I - Abstract
This study is performed to examine the impacts of microRNA‐218 (miR‐218) on cardiac microvascular endothelial cells (CMECs) injury induced by coronary artery disease (CAD). Reverse‐transcription quantitative polymerase chain reaction (RT‐qPCR) was applied for detecting miR‐218 expression in serum of patients with CAD and healthy controls, and the correlation between miR‐218 expression and the clinical indexes such as creatine kinase, creatine kinase‐myocardial band, cardiac troponin I, and coronary Gensini score was analyzed. CMECs were coincubated with homocysteine for 24 hr for CMECs injury, and the cells were transfected with miR‐218 mimics or miR‐218 inhibitors. Besides, we used oxidized low density lipoprotein as an inducer to incubate with CMECs for 24 hr, and the model of CMECs injury was established to be transfected with miR‐218 mimics. RT‐qPCR and western blot analysis were used to detect miR‐218 and HMGB1 expression in CMECs. A series of experiments were used to determine cell proliferation, apoptosis, migration, and angiogenesis ability of CMECs. Vascular endothelial growth factor expression and inflammatory factor contents were measured. The obtained results suggested that miR‐218 expression in peripheral blood of patients with CAD descended substantially versus that of healthy controls. Low miR‐218 expression was found in CAD‐induced CMECs injury. Overexpressed miR‐218 promoted the proliferation, migration, angiogenesis ability, induced apoptosis, and alleviated the inflammatory injury of CAD‐induced CMECs. miR‐218 may negatively regulate the expression of HMGB1 in CAD. This study demonstrates that upregulation of miR‐218 reduces CMECs injury induced by CAD through the inhibition of HMGB1. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
21. Superoxide dismutase mimetic nanozymes attenuate cardiac microvascular ischemia–reperfusion injury associated with hyperhomocysteinemia.
- Author
-
Ding, Lu, Zhang, Shifei, Li, Yao, Wu, Yuhao, Liu, Xiaoming, Xu, Dunwu, Zhao, Kaiyang, Xu, Chuan, Yu, Bentong, Huang, Xiaolin, Tang, Ben Zhong, and Zhang, Wan
- Subjects
- *
REPERFUSION injury , *SUPEROXIDE dismutase , *REPERFUSION , *SYNTHETIC enzymes , *HYPERHOMOCYSTEINEMIA , *MYOCARDIAL infarction - Abstract
[Display omitted] • Cerium vanadate nanorods (CVNRs) with outstanding superoxide dismutase-like activity were designed and constructed. • CVNRs were used as an efficient reactive oxygen species scavenger for treating myocardial ischemia–reperfusion injury (IRI) in hyperhomocysteinemic rat. • CVNRs could effectively protect the microvascular, reduce the myocardial infarction and improve the heart function after IRI. • CVNRs exhibited excellent biocompatibility and biosafety. The combination of homocysteine (Hcy) and copper ion (Cu2+) induces cardiac microvascular ischemia–reperfusion injury (IRI) by causing oxidative stress in the reperfusion treatment of ischemic cardiopathy. Therefore, antioxidant elimination of reactive oxygen species (ROS) shows great potential in the treatment of IRI. Herein, cerium vanadate nanorods (CVNRs) were designed and developed as an efficient ROS scavenger for the treatment of myocardial IRI in hyperhomocysteinemic rat (HHcyR) model. Thanks to its outstanding superoxide dismutase (SOD)-like activity, the CVNRs showed a significant antioxidant effect in human cardiac microvascular endothelial cells treated with Hcy and Cu2+ under hypoxia/reperfusion condition and greatly preserved the morphology and function of mitochondria. Further intravenous administration of CVNRs significantly maintained the integrity and perfusion of microvasculature, minimized the microcirculation malfunction, and finally reduced the myocardial infarction and maintained the heart function after IRI. Moreover, the administration of CVNRs in HHcyR exhibited negligible organ toxicity and inflammation response, indicating their excellent biocompatibility and biosafety. Collectively, the SOD-mimicking CVNRs can serve as a promising antioxidant nanozyme to treat cardiac microvascular IRI complicated with hyperhomocysteinemia, providing a new prospect for the development of antioxidant nanozyme in IRI treatment even with other risk factors. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
22. Quantitative Proteomics Analysis of Ischemia/Reperfusion Injury-Modulated Proteins in Cardiac Microvascular Endothelial Cells and the Protective Role of Tongxinluo
- Author
-
Qing Li, He-He Cui, Yue-Jin Yang, Xiang-Dong Li, Gui-Hao Chen, Xia-Qiu Tian, Chen Jin, Qiu-Ting Dong, Pei-Sen Huang, and Jun Xu
- Subjects
Tongxinluo ,Proteomics ,Cardiac microvascular endothelial cells ,Ischemic/reperfusion injury ,Physiology ,QP1-981 ,Biochemistry ,QD415-436 - Abstract
Background: The protection of endothelial cells (ECs) against reperfusion injury has received little attention. In this study, we used Tandem Mass Tag (TMT) labeling proteomics to investigate the modulated proteins in an in vitro model of cardiac microvascular endothelial cells (CMECs) subjected to ischemia/reperfusion (I/R) injury and their alteration by traditional Chinese medicine Tongxinluo (TXL). Methods: Human CMECs were subjected to 2 h of hypoxia followed by 2 h of reoxygenation with different concentrations of TXL Protein expression profiles of CMECs were determined using tandem mass spectrometry. We evaluated several proteins with altered expression in I/R injury and summarized some reported proteins related to I/R injury. Results: TXL dose-dependently decreased CMEC apoptosis, and the optimal concentration was 800 µg/mL. I/R significantly altered proteins in CMECs, and 30 different proteins were detected between a normal group and a hypoxia and serum deprivation group. In I/R injury, TXL treatment up-regulated 6 types of proteins including acyl-coenzyme A synthetase ACSM2B mitochondrial (ACSM2B), cyclin-dependent kinase inhibitor 1B (CDKN1B), heme oxygenase 1 (HMOX1), transcription factor SOX-17 (SOX17), sequestosome-1 isoform 1 (SQSTM1), and TBC1 domain family member 10B (TBC1D10B). Also, TXL down-regulated 5 proteins including angiopoietin-2 isoform c precursor (ANGPT2), cytochrome c oxidase assembly factor 5 (COA5), connective tissue growth factor precursor (CTGF), cathepsin L1 isoform 2 (CTSL), and eukaryotic elongation factor 2 kinase (LOC101930123). These types of proteins mainly had vital functions, including cell proliferation, stress response, and regulation of metabolic process. Conclusions: The study presented differential proteins upon I/R injury through a proteomic analysis. TXL modulated the expression of proteins in CMECs and has a protective role in response to I/R.
- Published
- 2017
- Full Text
- View/download PDF
23. Substance-P Inhibits Cardiac Microvascular Endothelial Dysfunction Caused by High Glucose-Induced Oxidative Stress
- Author
-
Do Young Kim, Jiyuan Piao, and Hyun Sook Hong
- Subjects
hyperglycemia ,oxidative stress ,substance-P ,cardiac microvascular endothelial cells ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Diabetes is characterized by high glucose (HG) levels in the blood circulation, leading to exposure of the vascular endothelium to HG conditions. Hyperglycemia causes oxidative stress via excessive reactive oxygen species (ROS) production in the endothelium, which leads to cellular dysfunction and the development of diabetic vascular diseases. Substance-P (SP) is an endogenous peptide involved in cell proliferation and migration by activating survival-related signaling pathways. In this study, we evaluated the role of SP in cardiac microvascular endothelial cells (CMECs) in HG-induced oxidative stress. CMECs were treated with diverse concentrations of glucose, and then the optimal dose was determined. Treatment of CMECs with HG reduced their viability and induced excessive ROS secretion, inactivation of PI3/Akt signaling, and loss of vasculature-forming ability in vitro. Notably, HG treatment altered the cytokine profile of CMECs. However, SP treatment inhibited the HG-mediated aggravation of CMECs by restoring viability, free radical balance, and paracrine potential. SP-treated CMECs retained the capacity to form compact and long stretching-tube structures. Collectively, our data provide evidence that SP treatment can block endothelial dysfunction in hyperglycemia and suggest the possibility of using SP for treating diabetic complications as an antioxidant.
- Published
- 2021
- Full Text
- View/download PDF
24. Aspirin Enhances the Protection of Hsp90 from Heat-Stressed Injury in Cardiac Microvascular Endothelial Cells Through PI3K-Akt and PKM2 Pathways.
- Author
-
Xiaohui Zhang, Bixia Chen, Jiaxin Wu, Junzhou Sha, Bo Yang, Jie Zhu, Jiarui Sun, Jörg Hartung, and Endong Bao
- Subjects
- *
ENDOTHELIAL cells , *PHYSIOLOGICAL effects of heat , *ASPIRIN , *CARDIAC contraction , *MOLECULAR chaperones , *SUDDEN death - Abstract
Heat stress (HS) often causes sudden death of humans and animals due to heart failure, mainly resulting from the contraction of cardiac microvasculature followed by myocardial ischemia. Cardiac microvascular endothelial cells (CMVECs) play an important role in maintaining vasodilatation. Aspirin (ASA) is well known for its protective abilities of febrile animals. However, there is little knowledge about molecular resistance mechanisms of CMVECs and which role ASA may play in this context. Therefore, we used a heat stress model of rat cardiac microvascular endothelial cell cultures in vitro and investigated the cell injuries and molecular resistance mechanism of CMVECs caused by heat stress, and the effect of aspirin (ASA) on it. HS induced severe pathological damage of CMVECs and cellular oxidative stress and dysfunction of NO release. Hsp90 was proven to be indispensable for resisting HS-injury of CMVECs through PI3K-Akt and PKM2 signaling pathways. Meanwhile, PKM2 functioned in reducing Akt phosphorylation. ASA treatment of CMVECs induced a significant expression of Hsp90, which promoted both Akt and PKM2 signals, which are beneficial for relieving HS damage and maintaining the function of CMVECs. Akt activation also promoted HSF-1 that regulates the expression of Hsp70, which is known to assist Hsp90′s molecular chaperone function and when released to the extracellular liquid to protect myocardial cells from HS damage. To the best of our knowledge, this is the first study to show that HS damages CMVECs and the protection mechanism of Hsp90 on it, and that ASA provides a new potential strategy for regulating cardiac microcirculation preventing HS-induced heart failure. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
25. Angiotensin II induces apoptosis of cardiac microvascular endothelial cells via regulating PTP1B/PI3K/Akt pathway.
- Author
-
Wang, Yanyan, Fan, Yuyuan, Song, Yu, Han, Xueting, Fu, Mingqiang, Wang, Jingfeng, Cui, Xiaotong, Cao, Juan, Chen, Li, Hu, Kai, Sun, Aijun, Zhou, Jingmin, and Ge, Junbo
- Abstract
Endothelial cell apoptosis and renin-angiotensin-aldosterone system (RAAS) activation are the major pathological mechanisms for cardiovascular disease and heart failure; however, the interaction and mechanism between them remain unclear. Investigating the role of PTP1B in angiotensin II (Ang II)–induced apoptosis of primary cardiac microvascular endothelial cells (CMECs) may provide direct evidence of the link between endothelial cell apoptosis and RAAS. Isolated rat CMECs were treated with different concentrations of Ang II to induce apoptosis, and an Ang II concentration of 4 nM was selected as the effective dose for the subsequent studies. The CMECs were cultured for 48 h with or without Ang II (4 nM) in the absence or presence of the PTP1B inhibitor TCS 401 (8 μM) and the PI3K inhibitor LY294002 (10 μM). The level of CMEC apoptosis was assessed by TUNEL staining and caspase-3 activity. The protein expressions of PTP1B, PI3K, Akt, p-Akt, Bcl-2, Bax, caspase-3, and cleaved caspase-3 were determined by Western blot (WB). The results showed that Ang II increased apoptosis of CMECs, upregulated PTP1B expression, and inhibited the PI3K/Akt pathway. Furthermore, cotreatment with PTP1B inhibitor significantly decreased the number of apoptotic CMECs induced by Ang II, along with increased PI3K expression, phosphorylation of Akt and the ratio of Bcl-2/Bax, decreased caspase-3 activity, and a cleaved caspase-3/caspase-3 ratio, while treatment with LY294002 partly inhibited the anti-apoptotic effect of the PTP1B inhibitor. Ang II induces apoptosis of primary rat CMECs via regulating the PTP1B/PI3K/Akt pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
26. Long‐chain noncoding RNA GAS5 mediates oxidative stress in cardiac microvascular endothelial cells injury.
- Author
-
Diao, Liwei, Bai, Long, Jiang, Xingpei, Li, Jianjun, and Zhang, Qinghua
- Subjects
- *
NON-coding RNA , *ENDOTHELIAL cells , *OXIDATIVE stress , *VASCULAR endothelial growth factors , *ATP-binding cassette transporters , *CELL migration - Abstract
This study is performed to figure out the role of long‐chain noncoding RNA growth‐arrest specific transcript 5 (GAS5) in homocysteine (HCY)‐induced cardiac microvascular endothelial cells (CMECs) injury. CMECs were cultured and the model of CMECs injury was established by coincubation with HCY. To construct stable overexpression of GAS5 cells, the expression of GAS5, microRNA‐33a‐5p (miR‐33a‐5p) and ATP‐binding cassette transporter A1 (ABCA1), and biological characteristics of cells were determined. The messenger RNA (mRNA) level and secretion of vascular endothelial growth factor (VEGF), activity of reactive oxygen species (ROS) and superoxide dismutase (SOD), and the content of malondialdehyde (MDA) were measured. The binding site between GAS5 and miR‐33a‐5p and between miR‐33a‐5p and ABCA1 was verified. CMECs were successfully cultured. Reduction of GAS5 expression and ABCA1 expression together with increased expression of miR‐33a‐5p was found in CMECs induced by HCY. After overexpression of GAS5, there showed increased proliferative activity, decreased cell apoptosis rate and apoptosis index, enhanced cell migration ability, increased number of lumen formation, increased mRNA expression of VEGF in cells and the secretion in the supernatant, decreased activity of ROS and SOD in cells, and decreased content of ROS in cells. miR‐33a‐5p could promote the enrichment of GAS5 and ABCA1 was the direct target gene of miR‐33a‐5p. Our study suggests that the low expression of GAS5 was observed in HCY‐induced CMECs injury, and the upregulation of GAS5 could attenuate HCY‐induced CMECs injury by mediating oxidative stress, and its mechanism is related to the upregulation of ABCA1 expression by competitively binding with miR‐33a‐5p. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
27. Lutein suppresses ferroptosis of cardiac microvascular endothelial cells via positive regulation of IRF in cardiac hypertrophy.
- Author
-
Liu, Yang, Yang, Guanlin, Huo, Shiqiao, Wu, Jiabi, Ren, Ping, Cao, Yonggang, Gao, Jingquan, Tong, Liquan, and Min, Dongyu
- Subjects
- *
CARDIAC hypertrophy , *LUTEIN , *ENDOTHELIAL cells , *INTERFERON regulatory factors , *GLUTAMATE transporters , *GLUTATHIONE peroxidase - Abstract
Cardiac microvascular dysfunction contributes to cardiac hypertrophy (CH) and can progress to heart failure. Lutein is a carotenoid with various pharmacological properties, such as anti-apoptotic, anti-inflammatory, and antioxidant effects. Limited research has been conducted on the effects of lutein on pressure overload-induced CH. Studies have shown that CH is accompanied by ferroptosis in the cardiac microvascular endothelial cells (CMECs). This study aimed to investigate the effect of lutein on ferroptosis of CMECs in CH. The transcription factor interferon regulatory factor (IRF) is associated with immune system function, tumor suppression, and apoptosis. The results of this study suggested that pressure overload primarily inhibits IRF expression, resulting in endothelial ferroptosis. Administration of lutein increased the expression of IRF, providing protection to endothelial cells during pressure overload. IRF silencing downregulated solute carrier family 7 member 11 (SLC7A11) and glutathione peroxidase 4 (GPX4) expression, leading to the induction of ferroptosis in CMECs. Lutein supplementation suppressed endothelial ferroptosis by upregulating IRF. These data suggest that IRF may function as a transcription factor for SLC7A11 and that lutein represses ferroptosis in CMECs by upregulating IRF expression. Therefore, targeting IRF may be a promising therapeutic strategy for effective cardioprotection in patients with CH and heart failure. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
28. Tongxinluo Reverses the Hypoxia-suppressed Claudin-9 in Cardiac Microvascular Endothelial Cells
- Author
-
Kun Liu, Xiu-Juan Wang, Yan-Ning Li, Bin Li, Jin-Sheng Qi, Jing Zhang, and Yu Wang
- Subjects
Cardiac Microvascular Endothelial Cells ,Claudin-9 ,H3K9 Acetylation ,Hypoxia ,Tongxinluo ,Medicine - Abstract
Background: Claudin-5, claudin-9, and claudin-11 are expressed in endothelial cells to constitute tight junctions, and their deficiency may lead to hyperpermeability, which is the initiating process and pathological basis of cardiovascular disease. Although tongxinluo (TXL) has satisfactory antianginal effects, whether and how it modulates claudin-5, claudin-9, and claudin-11 in hypoxia-stimulated human cardiac microvascular endothelial cells (HCMECs) have not been reported. Methods: In this study, HCMECs were stimulated with CoCl2to mimic hypoxia and treated with TXL. First, the messenger RNA (mRNA) expression of claudin-5, claudin-9, and claudin-11 was confirmed. Then, the protein content and distribution of claudin-9, as well as cell morphological changes were evaluated after TXL treatment. Furthermore, the distribution and content histone H3K9 acetylation (H3K9ac) in the claudin-9 gene promoter, which guarantees transcriptional activation, were examined to explore the underlying mechanism, by which TXL up-regulates claudin-9 in hypoxia-stimulated HCMECs. Results: We found that hypoxia-suppressed claudin-9 gene expression in HCMECs (F = 7.244; P = 0.011) and the hypoxia-suppressed claudin-9 could be reversed by TXL (F = 61.911; P = 0.000), which was verified by its protein content changes (F = 29.142; P = 0.000). Moreover, high-dose TXL promoted the cytomembrane localization of claudin-9 in hypoxia-stimulated HCMECs, with attenuation of cell injury. Furthermore, high-dose TXL elevated the hypoxia-inhibited H3K9ac in the claudin-9 gene promoter (F = 37.766; P = 0.000), activating claudin-9 transcription. Conclusions: The results manifested that TXL reversed the hypoxia-suppressed claudin-9 by elevating H3K9ac in its gene promoter, playing protective roles in HCMECs.
- Published
- 2016
- Full Text
- View/download PDF
29. TLN1 synergizes with ITGA5 to ameliorate cardiac microvascular endothelial cell dysfunction.
- Author
-
Wang X, Mao W, and Ma X
- Subjects
- Humans, Heart, Integrins, Oxidative Stress, Talin, Atherosclerosis, Endothelial Cells
- Abstract
Background: The complex process of atherosclerosis is thought to begin with endothelial cell dysfunction, and advanced atherosclerosis is the underlying cause of coronary artery disease (CAD). Uncovering the underlying mechanisms of CAD-related endothelial cell injury may contribute to the treatment., Materials and Methods: Cardiac microvascular endothelial cells (CMVECs) were treated with oxidised low-density lipoprotein (ox-LDL) to mimic an injury model. The involvement of Talin-1 (TLN1) and integrin alpha 5 (ITGA5) in the proliferation, apoptosis, angiogenesis, inflammatory response, and oxidative stress in CMVECs were assessed., Results: TLN1 overexpression assisted CMVECs in resistance to ox-LDL stimulation, with alleviated cell proliferation and angiogenesis, reduced apoptosis, inflammatory response, and oxidative stress. TLN1 overexpression triggered increased ITGA5, and ITGA5 knockdown reversed the effects of TLN1 overexpression on the abovementioned aspects. Together, TLN1 synergized with ITGA5 to ameliorate the dysfunction in CMVECs., Conclusions: This finding suggests their probable involvement in CAD, and increasing their levels is beneficial to disease relief.
- Published
- 2024
- Full Text
- View/download PDF
30. FOXO6 transcription inhibition of CTRP3 promotes OGD/R-triggered cardiac microvascular endothelial barrier disruption via SIRT1/Nrf2 signalling.
- Author
-
Zheng S, Wang Y, Guo W, and Tan H
- Subjects
- Humans, Forkhead Transcription Factors metabolism, NF-E2-Related Factor 2 metabolism, Signal Transduction, Tumor Necrosis Factors, Endothelial Cells metabolism, Sirtuin 1 genetics, Sirtuin 1 metabolism
- Abstract
Background: C1q/TNF-related protein 3 (CTRP3) has been clarified to display its protective roles in cardiac function. The current study is concentrated on exploring the impacts of CTRP3 on myocardial ischaemia., Materials and Methods: Oxygen and glucose hypoxia/reoxygenation (OGD/R) model was constructed in human cardiac microvascular endothelial cells (HCMECs). Reverse transcription-quantitative polymerase chain reaction and western blot analysis of CTRP3 expression were conducted. CCK-8 assay was to estimate cell activity and lactate dehydrogenase (LDH) assay kit was to test LDH release. TUNEL assay and western blot were to judge apoptosis. Endothelial barrier function was detected by in vitro vascular permeability assay kit. Zonula occludens-1 (ZO-1) expression was evaluated by immunofluorescence assay. The interaction between CTRP3 promoter and Forkhead Box O6 (FOXO6) was predicted by JASPAR database and verified by chromatin immunoprecipitation and luciferase reporter assays. After OGD/R-induced HCMECs were co-transfected with CTRP3 overexpression and FOXO6 overexpression plasmids, the above functional experiments above were conducted again. Lastly, the expression of sirtuin 1 (SIRT1)/nuclear factor erythroid 2-related factor 2 (Nrf2) signalling-related proteins was examined by western blot., Results: CTRP3 was down-regulated in OGD/R-induced HCMECs. CTRP3 enhanced the viability and barrier integrity while reduced the apoptosis and permeability of OGD/R-insulted HCMECs. This process may be regulated by FOXO6 transcription. Also, FOXO6 inhibition-mediated CTRP3 up-regulation activated the SIRT1/Nrf2 signalling., Conclusions: FOXO6 transcription inhibition of CTRP3 promotes OGD/R-triggered cardiac microvascular endothelial barrier disruption via SIRT1/Nrf2 signalling.
- Published
- 2024
- Full Text
- View/download PDF
31. Allicin improves the function of cardiac microvascular endothelial cells by increasing PECAM-1 in rats with cardiac hypertrophy.
- Author
-
Shi, Pilong, Cao, Yonggang, Gao, Jingquan, Fu, Bowen, Ren, Jing, Ba, Lina, Song, Chao, Qi, Hanping, Huang, Wei, Guan, Xueying, and Sun, Hongli
- Abstract
Objective: Cardiac microvascular damage is significantly associated with the development of cardiac hypertrophy (CH). Researchers found that allicin could inhibit CH, but the relationship between cardiac microvessel and the inhibition of allicin on CH has not been reported. We aimed to investigate the effect of allicin on the function of cardiac microvascular endothelial cells (CMECs) in CH rat.Materials and Methods: The hemodynamic parameters were measured by BL-420F biological function experimental system and the indicators of the ventricular structure and function were measured by echocardiographic system. MTT assay was performed to assess the cell viability. Nitrite detection was performed to detect nitric oxide content. The morphology and molecular characteristics were detected by electron micrographs, immunofluorescence, quantitative real-time polymerase chain reaction (qRT-PCR), western blot. Wound healing experiment, analysis of tube formation and shear adaptation were performed to assess CMECs migration ability, angiogenesis and shear-responsiveness respectively.Result: Our findings have identified that microvascular density was decreased by observing the expression of platelet endothelial cell adhesion molecule-1 (PECAM-1) in CH rats. Interestingly, allicin improved the distribution and expression of PECAM-1. Meanwhile, allicin enhanced the migration and angiogenesis ability of CMECs, activated PECAM-1-PI3K-AKT-eNOS signaling pathway, however, the role of allicin was disappear after PECAM-1 was silenced. Allicin decreased the expression of caspase-3 and receptor interacting protein 3 (RIP3), inhibited necroptosis, and increased the levels of Angiopoietin-2 (Ang-2) and platelet-derived growth factor receptor-β (PDGFR-β). Under 10 dyn/cm2 condition, allicin advanced the modification ability of CMECs's shear-adaptation by activating PECAM-1.Conclusion: Allicin provided cardioprotection for CH rats by improving the function of CMECs through increasing the expression of PECAM-1. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
32. Matrine inhibits hypoxia/reoxygenation-induced apoptosis of cardiac microvascular endothelial cells in rats via the JAK2/STAT3 signaling pathway.
- Author
-
Zhao, Xue-Bin, Qin, Yi, Niu, Yu-Ling, and Yang, Jun
- Subjects
- *
HYPOXEMIA , *OXIDATIVE stress , *APOPTOSIS , *ENDOTHELIAL cells , *CARDIOVASCULAR diseases , *MICRORNA , *JAK-STAT pathway ,RISK factors - Abstract
Hypoxia/reoxygenation (H/R) often results in cellular oxidative stress and the subsequent apoptosis of cardiac microvascular endothelial cells (CMECs). More recently, studies have highlighted the therapeutic effects of matrine on various cardiovascular diseases. Thus, the aim of the present study was to investigate the underlying mechanism and effects of matrine on hypoxia/reoxygenation (H/R)-induced apoptosis of CMECs in rats. CMECs from Sprague Dawley (SD) rats were primarily treated with H/R, ld (low-dose, 0.5 mg/mL)-Ma + H/R, md (middle-dose, 1 mg/mL)-Ma + H/R, hd (high-dose, 2 mg/mL)-Ma + H/R, Ma + AG490 + H/R (2 mg/mL matrine and 50 μmol/L AG490, a JAK2/STAT3 signaling pathway inhibitor), and AG490 + H/R in an attempt to identify the underlying regulatory mechanisms of matrine. MTT assay was applied to determine cell viability. Hoechst staining was performed to detect the morphology of apoptotic CMECs, while cell cycle and the rate of apoptosis rate were determined by flow cytometry means. The mRNA and protein expression of the JAK2/STAT3 signaling pathway and apoptosis related genes were determined through the use of RT-qPCR and western blot assay methods respectively. An in vitro angiogenesis assay was employed to evaluate the value of matrine in tube formation. CMECs treated with ld-Ma+H/R, md-Ma+H/R, hd-Ma+H/R and Ma + AG490+H/R exhibited higher cell viability, greater cell ratio at the S phase, higher expression levels of p-JAK2 and p-STAT3, increased tube formation ability, and a lower apoptosis rate, with a lower ratio of cells at the G1 phase and Bax/Bcl-2 ratio. Meanwhile, the rats treated with AG490+H/R exhibited opposite results. Taken together, the key findings of the present study suggest that matrine inhibits the H/R-induced apoptosis of CMECs in rats via the JAK2/STAT3 signaling pathway, highlighting its therapeutic potential for H/R injury. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
33. 高糖环境中培养的小鼠心肌微血管内皮细胞蛋白激酶 с 活性、А 激酶锚定蛋白 150 表达变化
- Author
-
曾超, 霍瑞雪, 钟磊, 曾伟杰, 席玉胜, and 范智文
- Abstract
Objective To observe the activation of protein kinase C (PKC) and expression of А-kinase anchor protein 150 (AKAP150) in cardiac microvascular endothelial cells of mice pretreated with high glucose, and to investigate its significance. Methods The left ventricular cardiac microvascular endothelial cells were isolated from 8-week old male C57BL mice. The cultured cardiac microvascular endothelial cells were randomized into two groups : the control group ( n = 5 ) which was cultured in the normal medium with 5 mmol/L glucose,and the observation group (n = 5) that was cultured in the medium supplemented with 25 mmol/L glucose for 24 h. We used non-radioactive PKC assay to detect PKC activity and Western blotting to measure AKAP150 expression. The subcellular co-localization of AKAP150 and PKC was examined by immunofluorescence. Results The PKC activity of cardiac microvascular endothelial cells in the observation group was 0. 231 ±0. 037,which was higher than that of the control group ( 0. 143 ± 0. 047 )( P < 0. 05 ). The expression of AKAP150 protein in the observation group was 0.527 ±0.080,which was higher than that in the control group (0. 124 土 0. 038 ) ( P <0. 01). The co-localizaiton of AKAP150 and PKC in the observation group was more than that in the control group. Conclusions The PKC activity and AKAP150 expression are both increased in the cardiac microvascular endothelial cells of mice pretreated with high glucose, and the co-localization of PKC and AKAP150 is also significantly increased.The increase of PKC activity and AKAP150 expression may play a major role in the injury of mouse cardiac microvascular endothelial cells induced by high glucose. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
34. BI1 is associated with microvascular protection in cardiac ischemia reperfusion injury via repressing Syk-Nox2-Drp1-mitochondrial fission pathways.
- Author
-
Zhou, Hao, Shi, Chen, Hu, Shunying, Zhu, Hong, Ren, Jun, and Chen, Yundai
- Subjects
APOPTOSIS ,HOMEOSTASIS ,SMALL interfering RNA ,ISCHEMIA ,IMMUNOFLUORESCENCE - Abstract
Background: Mitochondrial fission has been identified as the pathogenesis underlying the development of cardiac microvascular ischemia reperfusion (IR) injury, although the regulatory signaling upstream from fission is far from clear. Bax inhibitor is a novel anti-apoptotic factor, and, however, its role of cardiac microvascular IR injury and mitochondrial homeostasis remains unclear.Methods: The cardiac microvascular IR injury was performed in WT mice and BI1 transgenic (BI
TG ) mice. The alterations of microvascular structure and function were detected via electron microscope, immunohistochemistry and immunofluorescence in vivo. Cardiac microvascular endothelial cells were isolated form WT and BITG mice and underwent hypoxia/reoxygenation injury in vitro. Cellular viability and apoptosis were analyzed via MTT assay and caspase-3 activity. Mitochondrial function, morphology and apoptosis were detected. Signaling pathways were analyzed via inhibitor, siRNA and mutant plasmid.Results: Herein, we demonstrated that Bax inhibitor 1 (BI1) was downregulated following cardiac microvascular IR injury, and its expression correlated negatively with microvascular collapse, endothelial cell apoptosis and mitochondrial damage. However, compared to wild-type mice, BI1 transgenic mice were actually protected from the acute microvascular injury and mitochondrial dysfunction. Functional studies illustrated that reintroduced BI1 directly interacted with and inhibited the Syk pathway, leading to the inactivation of Nox2. Subsequently, less Nox2 was associated with ROS downregulation, inhibiting Drp1 phosphorylated activation. Through repression of the Syk-Nox2-Drp1 signaling axis, BI1 strongly disrupted mitochondrial fission, abolishing mitochondrial apoptosis and thus sustaining endothelial cell viability.Conclusions: In summary, our report illustrates that BI1 functions as a novel microvascular guardian in cardiac IR injury that operates via inhibition of the Syk-Nox2-Drp1-mitochondrial fission signaling axis. Thus, novel therapeutic strategies to regulate the balance between BI1 and mitochondrial fission could provide a survival advantage to microvasculature following IR stress. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
35. Qiliqiangxin attenuates hypoxia‐induced injury in primary rat cardiac microvascular endothelial cells via promoting HIF‐1α‐dependent glycolysis.
- Author
-
Wang, Yanyan, Han, Xueting, Fu, Mingqiang, Wang, Jingfeng, Song, Yu, Liu, Yuan, Zhang, Jingjing, Zhou, Jingmin, and Ge, Junbo
- Subjects
CHINESE medicine ,CARDIOVASCULAR disease treatment ,ENDOTHELIAL cells ,GLYCOLYSIS ,HEART cells - Abstract
Abstract: Protection of cardiac microvascular endothelial cells (CMECs) against hypoxia injury is an important therapeutic strategy for treating ischaemic cardiovascular disease. In this study, we investigated the effects of qiliqiangxin (QL) on primary rat CMECs exposed to hypoxia and the underlying mechanisms. Rat CMECs were successfully isolated and passaged to the second generation. CMECs that were pre‐treated with QL (0.5 mg/mL) and/or HIF‐1α siRNA were cultured in a three‐gas hypoxic incubator chamber (5% CO
2 , 1% O2 , 94% N2 ) for 12 hours. Firstly, we demonstrated that compared with hypoxia group, QL effectively promoted the proliferation while attenuated the apoptosis, improved mitochondrial function and reduced ROS generation in hypoxic CMECs in a HIF‐1α‐dependent manner. Meanwhile, QL also promoted angiogenesis of CMECs via HIF‐1α/VEGF signalling pathway. Moreover, QL improved glucose utilization and metabolism and increased ATP production by up‐regulating HIF‐1α and a series of glycolysis‐relevant enzymes, including glucose transport 1 (GLUT1), hexokinase 2 (HK2), 6‐phosphofructokinase 1 (PFK1), pyruvate kinase M2 (PKM2) and lactate dehydrogenase A (LDHA). Our findings indicate that QL can protect CMECs against hypoxia injury via promoting glycolysis in a HIF‐1α‐dependent manner. Lastly, the results suggested that QL‐dependent enhancement of HIF‐1α protein expression in hypoxic CMECs was associated with the regulation of AMPK/mTOR/HIF‐1α pathway, and we speculated that QL also improved HIF‐1α stabilization through down‐regulating prolyl hydroxylases 3 (PHD3) expression. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
36. Autophagy inhibits high glucose induced cardiac microvascular endothelial cells apoptosis by mTOR signal pathway.
- Author
-
Zhang, Zheng, Zhang, Shenwei, Wang, Yong, Yang, Ming, Zhang, Ning, Jin, Zhitao, Ding, Liping, Jiang, Wei, Yang, Junke, Sun, Zhimin, Qiu, Chunguang, and Hu, Taohong
- Abstract
Cardiac microvascular endothelial cells (CMECs) dysfunction is an important pathophysiological event in the cardiovascular complications induced by diabetes. However, the underlying mechanism is not fully clarified. Autophagy is involved in programmed cell death. Here we investigated the potential role of autophagy on the CMECs injury induced by high glucose. CMECs were cultured in normal or high glucose medium for 6, 12 and 24 h respectively. The autophagy of CMECs was measured by green fluorescence protein (GFP)-LC3 plasmid transfection. Moreover, the apoptosis of CMEC was determined by flow cytometry. Furthermore, 3-Methyladenine (3MA), ATG7 siRNA and rapamycin were administrated to regulate the autophagy state. Moreover, Western blotting assay was performed to measure the expressions of Akt, mTOR, LC3 and p62. High glucose stress decreased the autophagy, whereas increased the apoptosis in CMECs time dependently. Meanwhile, high glucose stress activated the Akt/mTOR signal pathway. Furthermore, autophagy inhibitor, 3-MA and ATG7 siRNA impaired the autophagy and increased the apoptosis in CMECs induced by high glucose stress. Conversely, rapamycin up-regulated the autophagy and decreased the apoptosis in CMECs under high glucose condition. Our data provide evidence that high glucose directly inhibits autophagy, as a beneficial adaptive response to protect CMECs against apoptosis. Furthermore, the autophagy was mediated, at least in part, by mTOR signaling. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
37. Interleukin-37 suppresses the inflammatory response to protect cardiac function in old endotoxemic mice.
- Author
-
Li, Jilin, Zhai, Yufeng, Ao, Lihua, Hui, Haipeng, Fullerton, David A., Dinarello, Charles A., and Meng, Xianzhong
- Subjects
- *
MYOCARDIAL infarction , *TREATMENT of endotoxemia , *HEART function tests , *LIPOPOLYSACCHARIDES , *TUMOR necrosis factors , *TUMOR treatment - Abstract
Myocardial inflammatory responses to endotoxemia are enhanced in old mice, which results in worse cardiac dysfunction. Anti-inflammatory cytokine interleukin (IL)-37 has a broad effect on innate immunoresponses. We hypothesized that IL-37 suppresses myocardial inflammatory responses to protect cardiac function during endotoxemia in old mice. Old (20–24 month) wild-type (WT), and IL-37 transgenic (IL-37tg) mice were treated with lipopolysaccharide (LPS, 0.5 mg/kg, iv) or normal saline (0.1 ml/mouse, iv). Six hours later, left ventricle (LV) function was assessed using a pressure-volume microcatheter. Levels of monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor-α (TNF-α), interleukin (IL)-1β and IL-6 in plasma and myocardial tissue, as well as mononuclear cell density in the myocardium, were examined. Cardiac microvascular endothelial cells isolated from WT and IL-37tg mice were treated with LPS (0.2 µg/ml) for 0.5–24 h. Nuclear factor-kappa B (NF-κB) p65 phosphorylation was examined by immunoblotting, and MCP-1 levels in cell culture supernatant was determined using enzyme-linked immunosorbent assay. LV dysfunction in old WT endotoxemic mice was accompanied by up-regulated MCP-1, myocardial accumulation of mononuclear cells and production of TNF-α, IL-1β and IL-6. Expression of IL-37 suppressed myocardial inflammatory responses to endotoxemia in old mice, resulting in improved LV function. Treatment of old WT endotoxemic mice with recombinant IL-37 also improved LV function. In vitro experiments revealed that cardiac microvascular endothelial cells from IL-37tg mice had attenuated NF-κB activation and MCP-1 production following LPS stimulation. In conclusion, IL-37 is potent to suppress myocardial inflammation and protects against cardiac dysfunction during endotoxemia in old mice. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
38. Quantitative Proteomics Analysis of Ischemia/Reperfusion Injury-Modulated Proteins in Cardiac Microvascular Endothelial Cells and the Protective Role of Tongxinluo.
- Author
-
Li, Qing, Cui, He-He, Yang, Yue-Jin, Li, Xiang-Dong, Chen, Gui-Hao, Tian, Xia-Qiu, Jin, Chen, Dong, Qiu-Ting, Huang, Pei-Sen, and Xu, Jun
- Subjects
- *
PROTEOMICS , *REPERFUSION injury , *ENDOTHELIAL cells , *CHINESE medicine , *TANDEM mass spectrometry - Abstract
Background: The protection of endothelial cells (ECs) against reperfusion injury has received little attention. In this study, we used Tandem Mass Tag (TMT) labeling proteomics to investigate the modulated proteins in an in vitro model of cardiac microvascular endothelial cells (CMECs) subjected to ischemia/reperfusion (I/R) injury and their alteration by traditional Chinese medicine Tongxinluo (TXL). Methods: Human CMECs were subjected to 2 h of hypoxia followed by 2 h of reoxygenation with different concentrations of TXL. Protein expression profiles of CMECs were determined using tandem mass spectrometry. We evaluated several proteins with altered expression in I/R injury and summarized some reported proteins related to I/R injury. Results: TXL dose-dependently decreased CMEC apoptosis, and the optimal concentration was 800 μg/mL. I/R significantly altered proteins in CMECs, and 30 different proteins were detected between a normal group and a hypoxia and serum deprivation group. In I/R injury, TXL treatment up-regulated 6 types of proteins including acyl-coenzyme A synthetase ACSM2B mitochondrial (ACSM2B), cyclin-dependent kinase inhibitor 1B (CDKN1B), heme oxygenase 1 (HMOX1), transcription factor SOX-17 (SOX17), sequestosome-1 isoform 1 (SQSTM1), and TBC1 domain family member 10B (TBC1D10B). Also, TXL down-regulated 5 proteins including angiopoietin-2 isoform c precursor (ANGPT2), cytochrome c oxidase assembly factor 5 (COA5), connective tissue growth factor precursor (CTGF), cathepsin L1 isoform 2 (CTSL), and eukaryotic elongation factor 2 kinase (LOC101930123). These types of proteins mainly had vital functions, including cell proliferation, stress response, and regulation of metabolic process. Conclusions: The study presented differential proteins upon I/R injury through a proteomic analysis. TXL modulated the expression of proteins in CMECs and has a protective role in response to I/R. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
39. Overexpression of microRNA-136-3p Alleviates Myocardial Injury in Coronary Artery Disease via the Rho A/ROCK Signaling Pathway
- Author
-
Hanliang Dan, Yongbo Lin, and Jinguo Lu
- Subjects
Male ,Cardiac function curve ,lcsh:Diseases of the circulatory (Cardiovascular) system ,cardiac microvascular endothelial cells ,030232 urology & nephrology ,microrna-136-3p ,eif5a2 ,medicine.disease_cause ,lcsh:RC870-923 ,Rats, Sprague-Dawley ,Pathogenesis ,Coronary artery disease ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,microRNA ,lcsh:Dermatology ,Animals ,Medicine ,business.industry ,Myocardium ,General Medicine ,lcsh:RL1-803 ,medicine.disease ,lcsh:Diseases of the genitourinary system. Urology ,Rho Factor ,Rats ,MicroRNAs ,Nephrology ,Apoptosis ,lcsh:RC666-701 ,Cancer research ,rho a/rock signaling pathway ,Signal transduction ,Cardiology and Cardiovascular Medicine ,business ,Oxidative stress ,coronary artery disease ,Signal Transduction - Abstract
Objective: Coronary artery disease (CAD) is a cardiovascular disease that poses a fatal threat to human health, and the identification of potential biomarkers may help to delineate its pathophysiological mechanisms. Accumulating evidence has implicated microRNAs (miRNAs) in the pathogenesis and development of cardiovascular diseases. The present study aims to identify the expression of miRNA-136-3p (miR-136-3p) in CAD and further investigate its functional relevance in myocardial injury both in vitro and in vivo. Methods: Initially, CAD models were induced in rats by high-fat diet and intraperitoneal injection of pituitrin. Next, the effect of overexpressed miR-136-3p on cardiac function and pathological damage in myocardial tissue, cardiomyocyte apoptosis, oxidative stress and inflammatory response were assessed in CAD rats. Rat cardiac microvascular endothelial cells (CMECs) were isolated and cultured by the tissue explant method, and the CMEC injury model was induced by homocysteine (HCY). The function of miR-136-3p in vitro was further evaluated. Results: miR-136-3p was poorly expressed in the myocardial tissue of CAD rats and CMEC injury models. In vivo assays indicated that overexpressed miR-136-3p could improve cardiac function and alleviate pathological damage in myocardial tissue, accompanied by reduced oxidative stress and inflammatory response. Moreover,in vitro assays suggested that overexpression of miR-136-3p enhanced proliferation and migration while inhibiting apoptosis of HCY-stressed CMECs. Notably, we revealed that EIF5A2 was a target gene of miR-136-3p, and miR-136-3p inhibited EIF5A2 expression and activation of the Rho A/ROCK signaling pathway. Conclusion: In conclusion, the overexpression of miR-136-3p could potentially impede myocardial injury in vitro and in vivo in CAD through the blockade of the Rho A/ROCK signaling pathway, highlighting a potential miR-136-3p functional relevance in the treatment of CAD.
- Published
- 2020
40. FoxO3α-mediated autophagy contributes to apoptosis in cardiac microvascular endothelial cells under hypoxia.
- Author
-
Wang, Ruian, Yang, Qun, Wang, Xia, Wang, Wei, Li, Jing, Zhu, Juanxia, Liu, Xiaohua, Liu, Jian, and Du, Jianqing
- Subjects
- *
AUTOPHAGY , *FORKHEAD transcription factors , *APOPTOSIS , *ENDOTHELIAL cells , *HYPOXEMIA , *MYOCARDIAL infarction , *PATHOLOGICAL physiology , *CARDIOVASCULAR diseases - Abstract
Hypoxic injury of cardiac microvascular endothelial cells (CMECs) is an important pathophysiological event in myocardial infarction, whereas, the underlying mechanism is still poorly understood. Autophagy, a highly conserved process of cellular degradation, is required for normal cardiac function and also has been implicated in various cardiovascular diseases. Here we investigated the potential role of autophagy in CMEC dysfunction under hypoxia. CMECs were isolated from SD rats. Hypoxia (6–24 h, 1% O 2 ) induced autophagy in CMECs as evidenced by formation of punctate LC3, increased conversion of LC3-I to LC3-II and increased p62 degradation. Importantly, hypoxia-induced apoptosis in CMECs was attenuated by 3-Methyladenine (5 mM), an autophagy inhibitor, and aggravated by rapamycin (1.0 μg/L), an autophagy inducer. Meanwhile, hypoxia increased the nuclear localization of FoxO3α, accompanying with the decreased phosphorylation of FoxO3α and Akt. FoxO3α silencing decreased hypoxia-induced autophagy and the resultant apoptosis. Furthermore, treatment with 3-Methyladenine (10 mg/kg/day) improved the endothelial-dependent diastolic function of coronary artery in rats with myocardial infarction. These results indicated that hypoxia-induced autophagy formation in CMECs is mediated by FoxO3α and contributes to hypoxic injury of hearts. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
41. Substance-p Inhibits Cardiac Microvascular Endothelial Dysfunction Caused by High Glucose-Induced Oxidative Stress
- Author
-
Jiyuan Piao, Hyun Sook Hong, and Do-Young Kim
- Subjects
0301 basic medicine ,Antioxidant ,Endothelium ,Physiology ,medicine.medical_treatment ,Clinical Biochemistry ,cardiac microvascular endothelial cells ,RM1-950 ,030204 cardiovascular system & hematology ,Pharmacology ,medicine.disease_cause ,Biochemistry ,Article ,03 medical and health sciences ,Paracrine signalling ,0302 clinical medicine ,medicine ,oxidative stress ,Endothelial dysfunction ,Molecular Biology ,Protein kinase B ,chemistry.chemical_classification ,Reactive oxygen species ,Cell Biology ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,substance-P ,Therapeutics. Pharmacology ,hyperglycemia ,Signal transduction ,Oxidative stress - Abstract
Diabetes is characterized by high glucose (HG) levels in the blood circulation, leading to exposure of the vascular endothelium to HG conditions. Hyperglycemia causes oxidative stress via excessive reactive oxygen species (ROS) production in the endothelium, which leads to cellular dysfunction and the development of diabetic vascular diseases. Substance-P (SP) is an endogenous peptide involved in cell proliferation and migration by activating survival-related signaling pathways. In this study, we evaluated the role of SP in cardiac microvascular endothelial cells (CMECs) in HG-induced oxidative stress. CMECs were treated with diverse concentrations of glucose, and then the optimal dose was determined. Treatment of CMECs with HG reduced their viability and induced excessive ROS secretion, inactivation of PI3/Akt signaling, and loss of vasculature-forming ability in vitro. Notably, HG treatment altered the cytokine profile of CMECs. However, SP treatment inhibited the HG-mediated aggravation of CMECs by restoring viability, free radical balance, and paracrine potential. SP-treated CMECs retained the capacity to form compact and long stretching-tube structures. Collectively, our data provide evidence that SP treatment can block endothelial dysfunction in hyperglycemia and suggest the possibility of using SP for treating diabetic complications as an antioxidant.
- Published
- 2021
- Full Text
- View/download PDF
42. The secretion patterns and roles of cardiac and circulating arginine vasopressin during the development of heart failure.
- Author
-
Chen, Xuanlan, Lu, Guihua, Tang, Kaiyu, Li, Qinglang, and Gao, Xiuren
- Abstract
Objective The aim of this study is to investigate local cardiac and circulating AVP secretion during heart failure and to determine whether AVP mediates ventricular remodeling. Methods We assessed cardiac function and AVP levels of post-myocardial infarction (MI) heart-failure rats 3 weeks (n = 10), 4 weeks (n = 10), 6 weeks (n = 10), 9 weeks (n = 15) after the proximal left anterior descending coronary artery (LAD) ligation. Ten sham-operated rats were used as the control group. In vitro, cardiac microvascular endothelial cells (CMECs) were initiated from isolated Wistar rat hearts and subjected to Ang II to induce AVP expression and secretion. Besides, the effects of AVP stimulation on CMECs and cardiac fibroblasts (CFs) were studied using methylthiazol tetrazolium assay, Western blotting and real-time PCR. Results With cardiac dysfunction, plasma and local cardiac AVP, aldosterone levels increased over time, peaking at 9 weeks post-MI. AVP levels were negatively correlated with serum Na + and LVEF but positively correlated with LVEDD and myocardial hydroxyproline. In CMECs treated with Ang II, AVP mRNA and protein expression increased. In addition, AVP promoted CFs proliferation and up-regulated the expression of endothelin-1 and connective tissue growth factor. Conclusion CMECs are the cellular sources of elevated local heart AVP stimulated with Ang II/AT1. An intrinsic cardiac AVP system exists. Local cardiac and circulating AVP secretion were enhanced by deteriorating cardiac function. AVP may promote ventricular remodeling. Thus, AVP could be an important mediator of myocardial fibrosis in late-stage heart failure. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
43. FoxO3a suppresses the senescence of cardiac microvascular endothelial cells by regulating the ROS-mediated cell cycle.
- Author
-
Qi, Xu-Feng, Chen, Zhuo-Ying, Xia, Jing-Bo, Zheng, Li, Zhao, Hui, Pi, Long-Quan, Park, Kyu-Sang, Kim, Soo-Ki, Lee, Kyu-Jae, and Cai, Dong-Qing
- Subjects
- *
ENDOTHELIAL cells , *BIOLOGICAL rhythms , *CELL cycle , *EPITHELIAL cells , *ANTIOXIDANTS - Abstract
FoxO3a plays an important role in the aging process and decreases with age. However, the potential regulatory roles of FoxO3a in processes involved in cardiac microvascular endothelial cell (CMEC) senescence, and its underlying molecular mechanisms have not been elucidated. This study demonstrates that FoxO3a is deactivated in senescent CMECs together with the inhibition of proliferation and tube formation. Furthermore, the activation of the antioxidant enzymes catalase and SOD, downstream FoxO3a targets, was significantly decreased, thereby leading to cell cycle arrest in G1-phase by increased ROS generation and subsequently the activation of the p27 Kip1 pathway. However, FoxO3a overexpression in primary low-passage CMECs not only significantly suppressed the senescence process by increasing the activation of catalase and SOD but also markedly inhibited ROS generation and p27 Kip1 activation, although it failed to reverse cellular senescence. Moreover, both cell viability and tube formation were greatly increased by FoxO3a overexpression in primary CMECs during continuous passage. In addition, FoxO3a, deficiency in low-passage CMECs, accelerated the senescence process. Collectively, our data suggest that FoxO3a suppresses the senescence process in CMECs by regulating the antioxidant/ROS/p27 Kip1 pathways, although it fails to reverse the cellular senescent phenotype. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
44. Single-cell analysis for BDNF and TrkB receptors in cardiac microvascular endothelial cells.
- Author
-
Xinlei Bai, Chen Yilin, Xufeng Qi, and Dongqing Cai
- Subjects
- *
BRAIN-derived neurotrophic factor , *ENDOTHELIAL cells , *GENE expression , *PROTEIN-tyrosine kinases , *C-terminal residues , *AMINO acids - Abstract
Recent studies revealed that BDNF-TrkB pathway plays an important role in cardiac microvascular endothelial cells (CMECs) mediated myocardial angiogenesis. Single-cell analysis is a powerful tool for studying gene expression in individuals since cellular heterogeneity and dynamic microenvironments which individual cell will experience. Little is currently known about the expression of BDNF and TrkB receptors at the single CMEC level. Our single-cell analysis of seven randomly selected CMECs for BDNF and TrkB receptors (FL, T1, T2) showed that under an in vitro culture environment, BDNF was expressed in two of the seven selected CMECs. None of the single CMEC expressed TrkB-FL. TrkB-T1 was expressed in all seven selected CMECs, while, TrkB-T2 was expressed in three of these. In addition, none of single CMEC was found to express both BDNF and three TrkB receptors or BDNF and TrkB-FL simultaneously. These results suggest that a stochastic or random expression pattern for BDNF and their receptors might be set in each of the CMEC to response requirement of the time and spatial change, regulation or pathophysiological change. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
45. TXNIP mediates NLRP3 inflammasome activation in cardiac microvascular endothelial cells as a novel mechanism in myocardial ischemia/reperfusion injury.
- Author
-
Liu, Yi, Lian, Kun, Zhang, Lijian, Wang, Rutao, Yi, Fu, Gao, Chao, Xin, Chao, Zhu, Di, Li, Yan, Yan, Wenjun, Xiong, Lize, Gao, Erhe, Wang, Haichang, and Tao, Ling
- Subjects
- *
THIOREDOXIN-interacting protein , *INFLAMMATION , *ENDOTHELIAL cells , *CORONARY disease , *REPERFUSION injury , *GENETIC regulation , *CELLULAR signal transduction - Abstract
NLRP3 inflammasome is necessary for initiating acute sterile inflammation. Recent studies have demonstrated that NLRP3 inflammasome is up-regulated and mediates myocardial ischemia/reperfusion (MI/R) injury. However, the signaling pathways that lead to the activation of NLRP3 inflammasome by MI/R injury have not been fully elucidated. C57BL/6J mice were subjected to 30 min ischemia and 3 or 24 h reperfusion. The ischemic heart exhibited enhanced inflammasome activation as evidenced by increased NLRP3 expression and caspase-1 activity and increased IL-1β and IL-18 production. Intramyocardial NLRP3 siRNA injection or an intraperitoneal injection of BAY 11-7028, an inflammasome inhibitor, attenuated macrophage and neutrophil infiltration and decreased MI/R injury, as measured by cardiomyocyte apoptosis and infarct size. The ischemic heart also exhibited enhanced interaction between Txnip and NLRP3, which has been shown to be a mechanism for activating NLRP3. Intramyocardial Txnip siRNA injection also decreased infarct size and NLRP3 activation. In vitro experiments revealed that NLRP3 was expressed in cardiac microvascular endothelial cells (CMECs), but was hardly expressed in cardiomyocytes. Simulated ischemia/reperfusion (SI/R) stimulated NLRP3 inflammasome activation in CMECs, but not in cardiomyocytes. Moreover, CMECs subjected to SI/R injury increased interactions between Txnip and NLRP3. Txnip siRNA diminished NLRP3 inflammasome activation and SI/R-induced injury, as measured by LDH release and caspase-3 activity in CMECs. ROS scavenger dissociated TXNIP from NLRP3 and inhibited the activation of NLRP3 inflammasome in the CMECs. For the first time, we demonstrated that TXNIP-mediated NLRP3 inflammasome activation in CMECs was a novel mechanism of MI/R injury. Interventions that block Txnip/NLRP3 signaling to inhibit the activation of NLRP3 inflammasomes may be novel therapies for mitigating MI/R injury. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
46. Extracellular Ubiquitin Increases Expression of Angiogenic Molecules and Stimulates Angiogenesis in Cardiac Microvascular Endothelial Cells.
- Author
-
Steagall, Rebecca J., Daniels, Christopher R., Dalal, Suman, Joyner, William L., Singh, Mahipal, and Singh, Krishna
- Subjects
- *
UBIQUITIN , *EXTRACELLULAR matrix , *NEOVASCULARIZATION , *ENDOTHELIAL cells , *MICROCIRCULATION , *HEART diseases - Abstract
Extracellular Ub is an immune modulator that plays a role in suppression of inflammation, organ injury, myocyte apoptosis, and fibrosis. The purpose of this study was to investigate the effects of extracellular Ub on the process of cardiac angiogenesis. CMECs and aortic tissue were isolated from rats to measure changes in angiogenic protein levels and to assess angiogenic responses to extracellular Ub. In CMECs, extracellular Ub increased protein levels of VEGF-A and MMP-2, known angiogenesis regulators. CMECs demonstrated enhanced rearrangement of fibrillar actin and migration in response to Ub treatment. Ub-treated CMECs demonstrated an increase in tube network formation which was inhibited by the CXCR4 receptor antagonist, AMD3100. Methylated Ub, unable to form polyubiquitin chains, enhanced tube network formation. Aortic ring sprouting assays demonstrated that Ub increases microvessel sprouting in the Matrigel. The results of our study suggest a novel role for extracellular Ub in cardiac angiogenesis, providing evidence that extracellular Ub, at least in part acting via the CXCR4 receptor, has the potential to facilitate the process of angiogenesis in myocardial endothelial cells. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
47. NADPH oxidase 4 promotes cardiac microvascular angiogenesis after hypoxia/reoxygenation in vitro.
- Author
-
Wang, Jinyi, Hong, Zhibo, Zeng, Chao, Yu, Qiujun, and Wang, Haichang
- Subjects
- *
NADPH oxidase , *NEOVASCULARIZATION , *MICROCIRCULATION disorders , *HYPOXEMIA , *CORONARY disease , *ENDOTHELIAL cells - Abstract
Abstract: Microvascular endothelial cell dysfunction plays a key role in myocardial ischemia/reperfusion (I/R) injury, wherein reactive oxygen species (ROS)-dependent signaling is intensively involved. However, the roles of the various ROS sources remain unclear. This study sought to investigate the role of NADPH oxidase 4 (Nox4) in the cardiac microvascular endothelium in response to I/R injury. Adult rat cardiac microvascular endothelial cells (CMECs) were isolated and subjected to hypoxia/reoxygenation (H/R). Our results showed that Nox4 was highly expressed in CMECs, was significantly increased at both mRNA and protein levels after H/R injury, and contributed to H/R-stimulated increase in Nox activity and ROS generation. Downregulation of Nox4 by small interfering RNA transfection did not affect cell viability or ROS production under normoxia, but exacerbated H/R injury as evidenced by increased apoptosis and inhibited cell survival, migration, and angiogenesis after H/R. Nox4 inhibition also increased prolyl hydroxylase 2 (PHD2) expression and blocked H/R-induced increases in HIF-1α and VEGF expression. Pretreatment with DMOG, a specific competitive PHD inhibitor, upregulated HIF-1α and VEGF expression and significantly reversed Nox4 knockdown-induced injury. However, Nox2 was scarcely expressed and played a minimal role in CMEC survival and angiogenesis after H/R, though a modest upregulation of Nox2 was observed. In conclusion, this study demonstrated a previously unrecognized protective role of Nox4, a ROS-generating enzyme and the major Nox isoform in CMECs, against H/R injury by inhibiting apoptosis and promoting migration and angiogenesis via a PHD2-dependent upregulation of HIF-1/VEGF proangiogenic signaling. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
48. Qiliqiangxin attenuates hypoxia‐induced injury in primary rat cardiac microvascular endothelial cells via promoting HIF‐1α‐dependent glycolysis
- Author
-
Jingmin Zhou, Yu Song, Jingfeng Wang, Junbo Ge, Yanyan Wang, Mingqiang Fu, Xueting Han, Yuan Liu, and Jingjing Zhang
- Subjects
Male ,Vascular Endothelial Growth Factor A ,0301 basic medicine ,Angiogenesis ,cardiac microvascular endothelial cells ,Apoptosis ,Pharmacology ,Rats, Sprague-Dawley ,Adenosine Triphosphate ,Glycolysis ,Cells, Cultured ,Membrane Potential, Mitochondrial ,education.field_of_study ,biology ,Chemistry ,HIF‐1α ,Cell Hypoxia ,Mitochondria ,Up-Regulation ,Molecular Medicine ,Original Article ,medicine.symptom ,Signal Transduction ,glucose metabolism ,Lactate dehydrogenase A ,Procollagen-Proline Dioxygenase ,Neovascularization, Physiologic ,PKM2 ,Hydroxylation ,03 medical and health sciences ,qiliqiangxin ,medicine ,Animals ,education ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,hypoxia injury ,Glucose transporter ,Endothelial Cells ,Original Articles ,Cell Biology ,Hypoxia (medical) ,Hypoxia-Inducible Factor 1, alpha Subunit ,Glucose ,030104 developmental biology ,Microvessels ,biology.protein ,GLUT1 ,Reactive Oxygen Species ,Drugs, Chinese Herbal - Abstract
Protection of cardiac microvascular endothelial cells (CMECs) against hypoxia injury is an important therapeutic strategy for treating ischaemic cardiovascular disease. In this study, we investigated the effects of qiliqiangxin (QL) on primary rat CMECs exposed to hypoxia and the underlying mechanisms. Rat CMECs were successfully isolated and passaged to the second generation. CMECs that were pre‐treated with QL (0.5 mg/mL) and/or HIF‐1α siRNA were cultured in a three‐gas hypoxic incubator chamber (5% CO2, 1% O2, 94% N2) for 12 hours. Firstly, we demonstrated that compared with hypoxia group, QL effectively promoted the proliferation while attenuated the apoptosis, improved mitochondrial function and reduced ROS generation in hypoxic CMECs in a HIF‐1α‐dependent manner. Meanwhile, QL also promoted angiogenesis of CMECs via HIF‐1α/VEGF signalling pathway. Moreover, QL improved glucose utilization and metabolism and increased ATP production by up‐regulating HIF‐1α and a series of glycolysis‐relevant enzymes, including glucose transport 1 (GLUT1), hexokinase 2 (HK2), 6‐phosphofructokinase 1 (PFK1), pyruvate kinase M2 (PKM2) and lactate dehydrogenase A (LDHA). Our findings indicate that QL can protect CMECs against hypoxia injury via promoting glycolysis in a HIF‐1α‐dependent manner. Lastly, the results suggested that QL‐dependent enhancement of HIF‐1α protein expression in hypoxic CMECs was associated with the regulation of AMPK/mTOR/HIF‐1α pathway, and we speculated that QL also improved HIF‐1α stabilization through down‐regulating prolyl hydroxylases 3 (PHD3) expression.
- Published
- 2018
- Full Text
- View/download PDF
49. Up-regulation of IRF3 is required for docosahexaenoic acid suppressing ferroptosis of cardiac microvascular endothelial cells in cardiac hypertrophy rat.
- Author
-
Shi, Pilong, Song, Chao, Qi, Hanping, Ren, Jing, Ren, Ping, Wu, Jiabi, Xie, Yawen, Zhang, Meitian, Sun, Hongli, and Cao, Yonggang
- Subjects
- *
CARDIAC hypertrophy , *ENDOTHELIAL cells , *HEART cells , *DOCOSAHEXAENOIC acid , *GLUTAMATE transporters , *ARACHIDONIC acid - Abstract
The molecular characteristics of ferroptosis in cardiac hypertrophy have been rarely studied. Especially, there have been no studies to investigate the regulatory mechanisms of docosahexaenoic acid (DHA) on ferroptosis in cardiac hypertrophy. This study was designed to determine the role of ferroptosis in microvascular injury, and investigate the contribution of DHA in suppressing ferroptosis and preventing pressure overload-mediated endothelial damage. Our results indicated that the expression of interferon regulating factor 3 (IRF3) was primarily inhibited by pressure overload and consequently caused endothelial ferroptosis. Nevertheless, administration of DHA increased IRF3 expression and provided a pro-survival advantage for the endothelial system in the context of pressure overload. Experimental studies clearly showed that inhibition of IRF3 down-regulated SLC7A11 expression, and the latter leaded to the increase in the activities of arachidonate 12-lipoxygenase, which obligated cardiac microvascular endothelial cells to undergo ferroptosis via augmenting lipid peroxides. Interestingly, DHA supplementation suppressed endothelial ferroptosis via up-regulation of IRF3. Taken together, our studies identified the IRF3-SLC7A11-arachidonate 12-lipoxygenase axis as a new pathway responsible for pressure overload-mediated microvascular damage via initiating endothelial ferroptosis. In contrast, DHA treatment up-regulated the expression of IRF3 and thus reduced cellular ferroptosis, conferring a protective advantage to the endothelial system in pressure overload. These findings revealed that targeting IRF3 might be a useful therapeutic strategy for cardioprotection in cardiac hypertrophy and heart failure. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
50. Involvement of the FoxO3a pathway in the ischemia/reperfusion injury of cardiac microvascular endothelial cells.
- Author
-
Qi, Xu-Feng, Li, Yun-Jian, Chen, Zhuo-Ying, Kim, Soo-Ki, Lee, Kyu-Jae, and Cai, Dong-Qing
- Subjects
- *
REPERFUSION injury , *ENDOTHELIAL cells , *FORKHEAD transcription factors , *CORONARY disease , *HUMAN cell cycle , *CELLULAR signal transduction , *LABORATORY rats - Abstract
Abstract: FoxO3a, a member of the forkhead transcription factors, has been demonstrated to be involved in myocardial ischemia/reperfusion (I/R) injury. Cardiac microvascular endothelial cells (CMECs) are some of the predominant cells damaged immediately after myocardial I/R injury. Despite the importance of injured CMECs in an ischemic heart, little is known about the involvement of FoxO3a in regulating CMECs injury. Thus, we used rat CMECs following simulated I/R to examine FoxO3a activation and signaling in relation to survival, the cell cycle and apoptosis in CMECs. We found that Akt negatively regulates activation of the FoxO3a pathway by phosphorylating FoxO3a in CMECs as demonstrated with an Akt inhibitor and activator. Upon I/R injury, the FoxO3a pathway was significantly activated in CMECs, which was accompanied by Akt deactivation. In parallel, the I/R of CMECs induced G1-phase arrest through p27Kip1 up-regulation and significant activation of caspase-3. Accordingly, inhibition of the FoxO3a pathway by IGF-1, an Akt activator, could significantly block the I/R-enhanced activation of p27Kip1 and caspase-3 in CMECs. Collectively, our results indicate that the FoxO3a pathway is involved in the I/R injury of CMECs at least in part through the regulation of cell cycle arrest and apoptosis, suggesting that the FoxO3a pathway may be a novel therapeutic target that protects against microvascular endothelial damage in ischemic hearts. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.