Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Biomed Khim ; 70(3): 156-160, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38940204

ABSTRACT

The cytokine profile of primary coronary artery endothelial cells cultivated in the presence of doxorubicin (2 µg/ml and 6 µg/ml) was evaluated using enzyme-linked immunosorbent assay and qPCR. Cultivation of cells in the presence of these concentrations of doxorubicin for 24 h, upregulated expression of the following genes: IL6 (by 2.30 and 2.66 times, respectively), IL1B (by 1.25 and 3.44 times), and CXCL8 (by 6.47 times and 6.42 times), MIF (2.34 and 2.28 times), CCL2 (4.22 and 3.98 times). Under these conditions the following genes were downregulated: IL10, IL1R2, TNF. Cultivation of cells in the presence of doxorubicin (2 µg/ml and 6 µg/ml) for 24 h also increased the secretion of IL-6.


Subject(s)
Coronary Vessels , Doxorubicin , Endothelial Cells , Interleukin-6 , Humans , Doxorubicin/pharmacology , Coronary Vessels/cytology , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Interleukin-6/metabolism , Interleukin-6/genetics , Cells, Cultured , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Cytokines/metabolism , Cytokines/genetics , Gene Expression Regulation/drug effects , Interleukin-8/metabolism , Interleukin-8/genetics , Chemokine CCL2/metabolism , Chemokine CCL2/genetics , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Interleukin-10/metabolism , Interleukin-10/genetics
2.
Redox Biol ; 69: 102979, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38061206

ABSTRACT

BACKGROUND: Empagliflozin (EMPA) ameliorates reactive oxygen species (ROS) generation in human endothelial cells (ECs) exposed to 10 % stretch, but the underlying mechanisms are still unclear. Pathological stretch is supposed to stimulate protein kinase C (PKC) by increasing intracellular calcium (Ca2+), therefore activating nicotinamide adenine dinucleotide phosphate oxidase (NOX) and promoting ROS production in human ECs. We hypothesized that EMPA inhibits stretch-induced NOX activation and ROS generation through preventing PKC activation. METHODS: Human coronary artery endothelial cells (HCAECs) were pre-incubated for 2 h before exposure to cyclic stretch (5 % or 10 %) with either vehicle, EMPA or the PKC inhibitor LY-333531 or PKC siRNA. PKC activity, NOX activity and ROS production were detected after 24 h. Furthermore, the Ca2+ chelator BAPTA-AM, NCX inhibitor ORM-10962 or NCX siRNA, sodium/potassium pump inhibitor ouabain and sodium hydrogen exchanger (NHE) inhibitor cariporide were applied to explore the involvement of the NHE/Na+/NCX/Ca2+ in the ROS inhibitory capacity of EMPA. RESULTS: Compared to 5 % stretch, 10 % significantly increased PKC activity, which was reduced by EMPA and PKC inhibitor LY-333531. EMPA and LY-333531 showed a similar inhibitory capacity on NOX activity and ROS generation induced by 10 % stretch, which was not augmented by combined treatment with both drugs. PKC-ß knockdown inhibits the NOX activation induced by Ca2+ and 10 % stretch. BAPTA, pharmacologic or genetic NCX inhibition and cariporide reduced Ca2+ in static HCAECs and prevented the activation of PKC and NOX in 10%-stretched cells. Ouabain increased ROS generation in cells exposed to 5 % stretch. CONCLUSION: EMPA reduced NOX activity via attenuation of the NHE/Na+/NCX/Ca2+/PKC axis, leading to less ROS generation in HCAECs exposed to 10 % stretch.


Subject(s)
Benzhydryl Compounds , Coronary Vessels , Endothelial Cells , Glucosides , Guanidines , Indoles , Maleimides , Sulfones , Humans , Endothelial Cells/metabolism , Reactive Oxygen Species/metabolism , Coronary Vessels/metabolism , Protein Kinase C/metabolism , Ouabain/metabolism , Oxidative Stress , Sodium-Hydrogen Exchangers/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
3.
J Med Ultrason (2001) ; 51(1): 39-48, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38052761

ABSTRACT

PURPOSE: Here we aimed to develop a minimally invasive treatment for ischemic heart disease and demonstrate that low-intensity pulsed ultrasound (LIPUS) therapy improves myocardial ischemia by promoting myocardial angiogenesis in a porcine model of chronic myocardial ischemia. Studies to date determined the optimal treatment conditions within the range of settings available with existing ultrasound equipment and did not investigate a wider range of conditions. METHODS: We investigated a broad range of five parameters associated with ultrasound irradiation conditions that promote expression of endothelial nitric oxide synthase (eNOS), a key molecule that promotes angiogenesis in human coronary artery endothelial cells (HCAEC). RESULTS: Suboptimal irradiation conditions included 1-MHz ultrasound frequency, 500-kPa sound pressure, 20-min total irradiation time, 32-48-[Formula: see text] pulse duration, and 320-[Formula: see text] pulse repetition time. Furthermore, a proposed index, [Formula: see text], calculated as the product of power and the total number of irradiation cycles applied to cells using LIPUS, uniformly revealed the experimental eNOS expression associated with the various values of five parameters under different irradiation conditions. CONCLUSION: We determined the suboptimal ultrasound irradiation conditions for promoting eNOS expression in HCAEC.


Subject(s)
Myocardial Ischemia , Nitric Oxide Synthase Type III , Humans , Animals , Swine , Nitric Oxide Synthase Type III/metabolism , Endothelial Cells/metabolism , Ultrasonic Waves
4.
Physiol Rep ; 11(24): e15866, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38114067

ABSTRACT

Human bone mesenchymal stem cell-derived extracellular vesicles (HBMSC-EV) have been used successfully in animal models of myocardial ischemia, yet have dampened effects in metabolic syndrome through unknown mechanisms. This study demonstrates the basal differences between non-diabetic human coronary artery endothelial cells (HCAEC) and diabetic HCAEC (DM-HCAEC), and how these cells respond to the treatment of HBMSC-EV. HCAEC and DM-HCAEC were treated with HBMSC-EV for 6 h. Proteomics, western blot analysis, and tube formation assays were performed. Key metabolic, growth, and stress/starvation cellular responses were significantly altered in DM-HCAEC in comparison to that of HCAEC at baseline. Proteomics demonstrated increased phosphorus metabolic process and immune pathways and decreased RNA processing and biosynthetic pathways in DM-HCAEC. Similar to previous in vivo findings, HCAEC responded to the HBMSC-EV with regenerative and anti-inflammatory effects through the upregulation of multiple RNA pathways and downregulation of immune cell activation pathways. In contrast, DM-HCAEC had a significantly diminished response to HBMSC-EV, likely due to the baseline abnormalities in DM-HCAEC. To achieve the full benefits of HBMSC-EV and for a successful transition of this potential therapeutic agent to clinical studies, the abnormalities found in DM-HCAEC will need to be further studied.


Subject(s)
Diabetes Mellitus , Extracellular Vesicles , Mesenchymal Stem Cells , Animals , Humans , Coronary Vessels/metabolism , Endothelial Cells/metabolism , Cells, Cultured , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , Diabetes Mellitus/metabolism
5.
Hematology ; 28(1): 2277502, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37933872

ABSTRACT

OBJECTIVE: High expression of nuclear factor interleukin-3 (NFIL3) and integrin Alpha M (ITGAM) was found in serum samples from Kawasaki disease (KD) patients through bioinformatics analysis. Hence, this study aimed to explore the biological functions of NFIL3 and ITGAM in KD serum-stimulated human coronary artery endothelial cells (HCAECs). METHODS: The differentially-expressed genes in KD were analyzed through bioinformatics analysis. Serum samples were obtained from 18 KD patients and 18 healthy volunteers, followed by detection of NFIL3 and ITGAM levels in KD serum. After HCAECs were transfected with sh-NFIL3, sh-ITGAM, or sh-NFIL3 + oe-ITGAM and underwent 24-h KD serum stimulation, cell viability and apoptosis and the levels of inflammation-related factors were measured. The binding between NFIL3 and ITGAM was validated by dual-luciferase and chromatin immunoprecipitation (ChIP) assays. RESULTS: NFIL3 and ITGAM were up-regulated in serum from KD patients and KD serum-stimulated HCAECs. Down-regulation of NFIL3 or ITGAM inhibited KD serum-induced cell apoptosis and inflammatory response of HCAECs and promoted cell viability. Mechanistically, NFIL3 promoted ITGAM transcription level. Up-regulation of ITGAM reversed the improvement of NFIL3 down-regulation on KD serum-induced HCAEC injury. CONCLUSION: NFIL3 aggravated KD serum-induced HCAEC injury by promoting ITGAM transcription, which provided new insights into the treatment of KD.


Subject(s)
Coronary Vessels , Mucocutaneous Lymph Node Syndrome , Humans , Coronary Vessels/metabolism , Endothelial Cells/metabolism , Mucocutaneous Lymph Node Syndrome/metabolism , CD11b Antigen/metabolism , Interleukin-3/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism
6.
Nutr Metab Cardiovasc Dis ; 32(10): 2424-2438, 2022 10.
Article in English | MEDLINE | ID: mdl-36096977

ABSTRACT

BACKGROUND AND AIMS: Low-grade chronic inflammation was reported to serve as a distinctive pathophysiologic feature of coronary artery disease (CAD), the leading cause of death around the world. Herein, the current study aimed to explore whether and how microRNA-34c-5p (miR-34c-5p), a miRNA enriched in extracellular vesicles (EVs) originated from the activated platelet (PLT-EVs), affects the inflammation of human coronary artery endothelial cells (HCAECs). METHODS AND RESULTS: HCAECs were established as an in vitro cell model using oxidized low-density lipoprotein (ox-LDL). miR-34c-5p, an abundant miRNA in PLT-EVs, can be transferred to HCAECs and target PODXL by binding to its 3'UTR. Gain- and loss-of-function experiments of miR-34c-5p and podocalyxin (PODXL) were performed in ox-LDL-induced HCAECs. Subsequently, HCAECs were subjected to co-culture with PLT-EVs, followed by detection of the expression patterns of key pro-inflammatory factors. Either miR-34c-5p mimic or PLT-EVs harboring miR-34c-5p attenuated the ox-LDL-evoked inflammation in HCAECs by suppressing interleukin-1ß (IL-1ß), IL-6 and tumor necrosis factor-α (TNF-α). By blocking the P38 MAPK signaling pathway, miR-34c-5p-mediated depletion of PODXL contributed to protection against ox-LDL-induced inflammation. In vitro findings were further validated by findings observed in ApoE knock-out mice. Additionally, miR-34c-5p in PLT-EVs showed an athero-protective role in the murine model. CONCLUSION: Altogether, our findings highlighted that miR-34c-5p in PLT-EVs could alleviate inflammation response in HCAECs by targeting PODXL and inactivation of the P38 MAPK signaling pathway.


Subject(s)
Extracellular Vesicles , MicroRNAs , 3' Untranslated Regions , Animals , Apolipoproteins E/genetics , Coronary Vessels/metabolism , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/prevention & control , Interleukin-1beta , Interleukin-6/metabolism , Lipoproteins, LDL/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Sialoglycoproteins , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Bioengineered ; 13(5): 11469-11486, 2022 05.
Article in English | MEDLINE | ID: mdl-35535400

ABSTRACT

Coronary artery disease (CAD) represents a fatal public threat. The involvement of extracellular vesicles (EVs) in CAD has been documented. This study explored the regulation of embryonic stem cells (ESCs)-derived EVs-hnRNPU-actin complex in human coronary artery endothelial cell (HCAEC) growth. Firstly, in vitro HCAEC hypoxia models were established. EVs were extracted from ESCs by ultracentrifugation. HCAECs were treated with EVs and si-VEGF for 24 h under hypoxia, followed by assessment of cell proliferation, apoptosis, migration, and tube formation. Uptake of EVs by HCAECs was testified. Additionally, hnRNPU, VEGF, and RNA Pol II levels were determined using Western blotting and CHIP assays. Interaction between hnRNPU and actin was evaluated by Co-immunoprecipitation assay. HCAEC viability and proliferation were lowered, apoptosis was enhanced, wound fusion was decreased, and the number of tubular capillary structures was reduced under hypoxia, whereas ESC-EVs treatment counteracted these effects. Moreover, EVs transferred hnRNPU into HCAECs. EVs-hnRNPU-actin complex increased RNA Pol II level on the VEGF gene promoter and promoted VEGF expression in HCAECs. Inhibition of hnRNPU or VEGF both annulled the promotion of EVs on HCAEC growth. Collectively, ESC-EVs-hnRNPU-actin increased RNA Pol II phosphorylation and VEGF expression, thus promoting HCAEC growth.


Subject(s)
Actins , Endothelial Cells , Extracellular Vesicles , Heterogeneous-Nuclear Ribonucleoprotein U , RNA Polymerase II , Actins/metabolism , Cell Proliferation/genetics , Coronary Vessels/cytology , Endothelial Cells/metabolism , Extracellular Vesicles/genetics , Extracellular Vesicles/metabolism , Heterogeneous-Nuclear Ribonucleoprotein U/genetics , Heterogeneous-Nuclear Ribonucleoprotein U/metabolism , Humans , Hypoxia/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
8.
Hum Cell ; 35(2): 486-497, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34978047

ABSTRACT

Cardiovascular disease (CVD) is a leading non-communicable disease with a high fatality rate worldwide. Hypertension, a common cardiovascular condition, is a significant risk factor for the development of heart failure because the activation of the renin-angiotensin system (RAS) is considered to be the major promoting reason behind myocardial fibrosis (MF). In this study, Angiotensin II (Ang II) stimulation-induced endothelial to mesenchymal transition (End-MT) in HCAECs, including the decrease of CD31 level, the increase of α-SMA, collagen I, slug, snail, and TGF-ß1 levels, and the promotion of Smad2/3 phosphorylation. Meanwhile, the c-Ski level was reduced in Ang II-stimulated HCAECs. In HCAECs, Ang II-induced changes could be partially attenuated by c-Ski overexpression. miR-214-3p directly targeted c-Ski and inhibited c-Ski expression. Moreover, miR-214-3p inhibition reduced Ang II-caused End-MT in HCAECs. miR-214-3p overexpression further enhanced Ang II-induced End-MT, while c-Ski overexpression could markedly reverse the effects of miR-214-3p overexpression. In the Ang II-induced mouse cardiac hypertrophic model, Ang II-caused increase of cellular cross-sectional area and cardiac fibrosis were partially ameliorated by LV-c-Ski; when mice were co-treated with LV-c-Ski and agomir-214-3p, the beneficial effects of LV-c-Ski were reversed. In conclusion, the miR-214-3p/c-Ski axis modulated Ang II-induced End-MT in HCAECs and cardiac hypertrophy and fibrosis in the mice model.


Subject(s)
DNA-Binding Proteins , Endothelial Cells , MicroRNAs , Proto-Oncogene Proteins , Animals , Coronary Vessels/metabolism , Endothelial Cells/metabolism , Fibrosis , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Signal Transduction
9.
Front Bioeng Biotechnol ; 9: 806151, 2021.
Article in English | MEDLINE | ID: mdl-34881239

ABSTRACT

The coverage of stents with healthy endothelium is crucial to the success of cardiovascular stent implantation. Immobilizing bioactive molecules on stents is an effective strategy to generate such stents. Glycogen synthase kinase-3ß inhibitor (GSKi) is a bioactive molecule that can effectively accelerate vascular endothelialization. In this work, GSKi was covalently conjugated on 316L stainless steel through polydopamine to develop a stable bioactive surface. Fourier transform infrared spectroscopy (FTIR), scanning electron microscopy (SEM) and water contact angle results revealed the successful introduction of GSKi onto 316L stainless steel. The GSKi coating did not obviously affect the hemocompatibility of plates. The adhesion and proliferation of human coronary artery endothelial cells (HCAECs) on stainless steel was significantly promoted by the addition of GSKi. In summary, this work provides a universal and stable strategy of immobilizing GSKi on the stent surface. This method has the potential for widespread application in the modification of vascular stents.

10.
Mol Cell Biochem ; 476(12): 4245-4263, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34351574

ABSTRACT

Kawasaki disease (KD) causes cardiovascular system injury in children. However, the pathogenic mechanisms of KD have not been well defined. Recently, strong correlation between aberrant microRNAs and KD nosogenesis has been revealed. A role of microRNA-197-3p (miR-197-3p) in the pathogenesis of KD is identified in the present study. Cell proliferation assay showed human coronary artery endothelial cells (HCAECs) were suppressed by serum from KD patients, which was correlated with high levels of miR-197-3p in both KD serum and HCAECs cultured with KD serum. The inhibition of HCAECs by miR-197-3p was confirmed by cells expressing miR-197-3p mimic and miR-197-3p inhibitor. Comparative proteomics analysis and Ingenuity Pathway Analysis (IPA) revealed TIMP3 as a potential target of miR-197-3p, which was demonstrated by western blot and dual-luciferase reporter assays. Subsequently, by detecting the endothelium damage markers THBS1, VWF, and HSPG2, the role of miR-197-3p/TIMP3 in KD-induced damage to HCAECs was confirmed, which was further validated by a KD mouse model in vivo. The expressions of miR-197-3p and its target, TIMP3, are dramatically variational in KD serum and HCAECs cultured with KD serum. Increased miR-197-3p induces HCAECs abnormal by restraining TIMP3 expression directly. Hence, dysregulation of miR-197-3p/TIMP3 expression in HCAECs may be an important mechanism in cardiovascular endothelium injury in KD patients, which offers a feasible therapeutic target for KD treatment.


Subject(s)
Coronary Artery Disease/pathology , Endothelial Cells/pathology , MicroRNAs/genetics , Mucocutaneous Lymph Node Syndrome/pathology , Proteome/metabolism , Tissue Inhibitor of Metalloproteinase-3/antagonists & inhibitors , Animals , Apoptosis/physiology , Cells, Cultured , Child, Preschool , Coronary Artery Disease/genetics , Coronary Artery Disease/immunology , Coronary Artery Disease/metabolism , Endothelial Cells/immunology , Endothelial Cells/metabolism , Female , Humans , Infant , Male , Mice , Mice, Inbred C57BL , MicroRNAs/blood , Mucocutaneous Lymph Node Syndrome/etiology , Mucocutaneous Lymph Node Syndrome/metabolism , Proteome/analysis , Tissue Inhibitor of Metalloproteinase-3/genetics , Tissue Inhibitor of Metalloproteinase-3/metabolism
11.
Front Cardiovasc Med ; 8: 650212, 2021.
Article in English | MEDLINE | ID: mdl-34095248

ABSTRACT

Background: This study investigated the association between long non-coding RNAs (lncRNAs) and coronary heart disease (CHD) and further elucidated the potential biological roles of lncRNAs in CHD pathogenesis. Methods: A case-control study (590 patients and 590 controls) was conducted from February 2017 and March 2019 in Fuzhou, China. Environmental factors were investigated using questionnaires and physical examinations. Five representative lncRNAs were screened using lncRNA microarray (peripheral blood in 5 cases and 5 controls) and further verified by quantitative real-time polymerase chain reaction (peripheral blood leukocyte in 100 cases and 100 controls). Oxidized low-density lipoprotein (oxLDL) was used to induce a human coronary artery endothelial cell (HCAECs) injury model, and loss of function was used to elucidate the role of lncRNA ENST00000609755.1 (lnc-MICALL2-2) in oxLDL-induced HCAECs injury. Results: A total of 320 lncRNAs were found dysregulated in CHD patients (fold change> 2, p < 0.05). The results of a discovery microarray, population verification and HCAEC experiments suggested the lnc-MICALL2-2 is upregulated in CHD subjects and in an oxLDL-induced HCAECs injury model. Conversely, lnc-MICALL2-2 inhibition in vitro attenuated the effects of oxLDL on HCAECs morphology, proliferation, and apoptosis. Conclusion: Elevated expression of lnc-MICALL2-2 is an independent risk factor for CHD, and knockdown subsequently confers protection against early pathological processes of oxLDL-induced CHD.

12.
J Adv Res ; 26: 123-135, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33133688

ABSTRACT

INTRODUCTION: In atherosclerotic lesions, extensive inflammation of the vessel wall contributes to plaque instability. Long noncoding RNAs (lncRNAs) play important roles in diverse biological processes in atherosclerosis. OBJECTIVES: Here, we aim to identify the functional role and regulatory mechanisms of lncRNA hypoxia-inducible factor 1 alpha-antisense RNA 2 (HIF1A-AS2) in atherosclerotic inflammation. METHODS: An atherosclerotic mouse model was induced in ApoE-/- mice by high fat diet (HFD). Endothelial cells (ECs), human aortic smooth muscle cells (SMCs) or human coronary artery endothelial cells (HCAECs) were exposed to ox-LDL to develop the in vitro model. The effects of lncRNA HIF1A-AS2 on inflammation were evaluated by determining levels of inflammatory factors tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß) and interleukin-6 (IL-6) and levels of adhesion molecules vascular cell adhesion molecule 1 (VCAM-1), intercellular adhesion molecule 1 (ICAM-1), and macrophage cationic peptide 1 (MCP-1). RESULTS: It was established that lncRNA HIF1A-AS2 and ATF2 were highly expressed in atherosclerotic ApoE-/- mice. Downregulating lncRNA HIF1A-AS2 in ox-LDL-exposed ECs, SMCs and HCAECs inhibited inflammation by reducing levels of pro-inflammatory factors and adhesion molecules. LncRNA HIF1A-AS2 bound to the transcription factor USF1 to elevate ATF2 expression. USF1 overexpression counteracted the suppressive effect of lncRNA HIF1A-AS2 silencing on ox-LDL-induced inflammation. Knockdown of lncRNA HIF1A-AS2 or ATF2 could also attenuate inflammation in atherosclerotic mice. Collectively, the present study demonstrates that downregulation of lncRNA HIF1A-AS2 represses the binding of USF1 to the ATF2 promoter region and then inhibits ATF2 expression, thereby suppressing atherosclerotic inflammation. CONCLUSION: This study suggests lncRNA HIF1A-AS2 as an promising therapeutic target for atherosclerosis.

13.
Mol Med Rep ; 22(3): 2052-2062, 2020 09.
Article in English | MEDLINE | ID: mdl-32582964

ABSTRACT

The purpose of the present study was to explore the potential molecular signaling pathway mediated by the statin rosuvastatin in cultured human coronary artery endothelial cells (HCAECs) induced by CoCl2. CoCl2 was used to induce the apoptosis of HCAECs. Myocardial infarction rats were established and received statin or PBS treatment. Reverse transcription­quantitative PCR, western blotting, ELISA, TUNEL assay and immunohistochemistry were used to analyze the role of statin treatment. The results showed that rosuvastatin treatment decreased apoptosis of HCAECs induced by CoCl2 by increasing anti­apoptosis Bcl­xl and Bcl­2 expression, and decreasing pro­apoptosis Bax, Bad, caspase­3 and caspase­9 expression. The myocardial ischemia rat model demonstrated that rosuvastatin treatment decreased the mitochondrial reactive oxygen species, inflammation, mitochondrial damage, lipid catabolism, heart failure and the myocardial infarction areas, but improved the cardiac function indicators, right and left ventricular ejection fraction and increased expression levels of Janus kinase (JAK) and signal transducer and activator of transcription (STAT)3 in myocardial tissue. In conclusion, the results of the current study revealed that the statin rosuvastatin presents cardioprotective effects by activation of the JAK2/STAT3 signaling pathway.


Subject(s)
Cobalt/adverse effects , Coronary Vessels/cytology , Myocardial Ischemia/drug therapy , Rosuvastatin Calcium/administration & dosage , Animals , Apoptosis/drug effects , Cell Line , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation/drug effects , Humans , Male , Myocardial Ischemia/etiology , Myocardial Ischemia/metabolism , Rats , Reactive Oxygen Species/metabolism , Rosuvastatin Calcium/pharmacology , Signal Transduction/drug effects
14.
J Cell Mol Med ; 24(2): 1189-1199, 2020 01.
Article in English | MEDLINE | ID: mdl-31758636

ABSTRACT

Myeloid-derived growth factor (MYDGF) is a novel protein secreted by bone marrow cells that features important physiological functions. In recent years, MYDGF has gained considerable interest due to their extensive beneficial effect on cardiac repair and protects cardiomyocytes from cell death. However, its precise molecular mechanisms have not been well elucidated. The purpose of this study was to produce sufficient amount of biologically active recombinant human (rh) MYDGF more economically and effectively by using in vitro molecular cloning techniques to study its clinical application. The prokaryotic expression system of Escherichia coli was established for the preparation of rhMYDGF. Finally, a large amount of high biologically active and purified form of recombinant protein was obtained. Moreover, we investigated the potential mechanism of rhMYDGF-mediated proliferation and survival in human coronary artery endothelial cells (HCAECs). Mechanistically, the results suggested that MAPK/STAT3 and the cyclin D1 signalling pathways are indispensable for rhMYDGF-mediated HCAEC proliferation and survival. Therefore, this study successfully established a preparation protocol for biologically active rhMYDGF and it may be a most economical way to produce high-quality active rhMYDGF for future clinical application.


Subject(s)
Cell Proliferation , Endothelium, Vascular/cytology , Escherichia coli/metabolism , Interleukins/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Cells, Cultured , Endothelium, Vascular/metabolism , Escherichia coli/genetics , Escherichia coli/growth & development , Humans , Interleukins/genetics , Interleukins/isolation & purification , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification
15.
Inflammation ; 42(4): 1413-1425, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31011929

ABSTRACT

Serum amyloid A (SAA) is an acute-phase protein with important, pathogenic role in the development of atherosclerosis. Since dysfunctional endothelium represents a key early step in atherogenesis, we aimed to determine whether induced human coronary artery endothelial cells (HCAEC) modulate SAA1/2/4 expression and influence intracellular location and intercellular transport of SAA1. HCAEC were stimulated with 1 ng/ml IL-1ß, 10 ng/ml IL-6, and/or 1 µM dexamethasone for 24 h. QPCR, Western blots, ELISA, and immunofluorescent labeling were performed for detection of SAA1/2/4 mRNA and protein levels, respectively. In SAA1 transport experiments, FITC- or Cy3-labeled SAA1 were added to HCAEC separately, for 24 h, followed by a combined incubation of SAA1-FITC and SAA1-Cy3 positive cells, with IL-1ß and analysis by flow cytometry. IL-1ß upregulated SAA1 (119.9-fold, p < 0.01) and SAA2 (9.3-fold; p < 0.05) mRNA expression levels, while mRNA expression of SAA4 was not affected. Intracellular SAA1 was found mainly as a monomer, while SAA2 and SAA4 formed octamers as analyzed by Western blots. Within HCAEC, SAA1/2/4 located mostly to the perinuclear area and tunneling membrane nanotubes. Co-culturing of SAA1-FITC and SAA1-Cy3 positive cells for 48 h showed a significantly higher percentage of double positive cells in IL-1ß-stimulated (mean ± SD; 60 ± 4%) vs. non-stimulated cells (48 ± 2%; p < 0.05). IL-1ß induces SAA1 expression in HCAEC and promotes its intercellular exchange, suggesting that direct communication between cells in inflammatory conditions could ultimately lead to faster development of atherosclerosis in coronary arteries.


Subject(s)
Coronary Vessels/cytology , Endothelial Cells/metabolism , Interleukin-1beta/pharmacology , Serum Amyloid A Protein/metabolism , Biological Transport , Cells, Cultured , Coronary Artery Disease/etiology , Humans
16.
BMC Mol Biol ; 20(1): 8, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30885136

ABSTRACT

BACKGROUND: Myocyte enhancer factor 2A (MEF2A) plays an important role in cell proliferation, differentiation and survival. Functional deletion or mutation in MEF2A predisposes individuals to cardiovascular disease mainly caused by vascular endothelial dysfunction. However, the effect of the inhibition of MEF2A expression on human coronary artery endothelial cells (HCAECs) is unclear. In this study, expression of MEF2A was inhibited by specific small interference RNA (siRNA), and changes in mRNA profiles in response to MEF2A knockdown were analyzed using an Agilent human mRNA array. RESULTS: Silencing of MEF2A in HCAECs accelerated cell senescence and suppressed cell proliferation. Microarray analysis identified 962 differentially expressed genes (DEGs) between the MEF2A knockdown group and the negative control group. Annotation clustering analysis showed that the DEGs were preferentially enriched in gene ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways related to proliferation, development, survival, and inflammation. Furthermore, 61 of the 578 downregulated DEGs have at least one potential MEF2A binding site in the proximal promoter and were mostly enriched in the GO terms "reproduction" and "cardiovascular." The protein-protein interaction network analyzed for the downregulated DEGs and the DEGs in the GO terms "cardiovascular" and "aging" revealed that PIK3CG, IL1B, IL8, and PRKCB were included in hot nodes, and the regulation of the longevity-associated gene PIK3CG by MEF2A has been verified at the protein level, suggesting that PIK3CG might play a key role in MEF2A knockdown induced HCAEC senescence. CONCLUSIONS: MEF2A knockdown accelerates HCAEC senescence, and the underlying molecular mechanism may be involved in down-regulation of the genes related with cell proliferation, development, inflammation and survival, in which PIK3CG may play a key role.


Subject(s)
Cellular Senescence/genetics , Coronary Vessels/cytology , Endothelial Cells , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Class Ib Phosphatidylinositol 3-Kinase/genetics , Endothelial Cells/cytology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation , Humans , Inflammation/genetics , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/physiology
17.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-851266

ABSTRACT

Objective: To investigate the effect of berberine on the apoptosis of human coronary artery endothelial cells (HCAECs) induced by intermittent high glucose in vitro. Methods HCAECs were isolated, identified, cultured, and divided into five groups: Group A was the normal glucose group (5.5 mmol/L glucose, NG); Group B was the persistent high glucose group (25 mmol/L glucose, PHG); Group C was the intermittent high glucose group (5.5 mmol/L and 25 mmol/L glucose fluctuated every 24 h, IHG); Group D was in a volatile hyperosmotic environment (5.5 mmol/L and 25 mmol/L mannitol alternating every 24 h to maintain the same osmotic pressure as IHG, OC); Group E was the fluctuation of hyperglycemia + berberine intervention group (5.5 mmol/L and 25 mmol/L glucose + 50 μmol/L berberine fluctuated every 24 h, IHG + BBR). The cells of each group were changed every 24 h, and the culture was stopped after 7 d, the cell viability and apoptosis were detected in each group. The expression of the apoptosis related protein Caspase-3 was determined by qRT-PCR and Western blotting. Results Compared with the persistent high glucose group, the apoptosis of HCAECs in the intermittent high glucose group was more significant (P < 0.05), but the apoptosis of HCAECs in the fluctuation of hyperglycemia + berberine intervention group was more significantly reduced than that in the intermittent high glucose group (P < 0.05). Berberine significantly reduced the expression of Caspase-3 induced by intermittent high glucose (P < 0.05) Conclusion Intermittent high glucose decreased the activity of HCAECs more easily than the persistent high glucose,and promoted the apoptosis of HCAECs. But berberine significantly reduced the apoptosis of HCAECs under intermittent high glucose, which had a significant protective effect on HCAECs.

18.
J Biomed Mater Res B Appl Biomater ; 106(1): 88-95, 2018 Jan.
Article in English | MEDLINE | ID: mdl-27875036

ABSTRACT

Biodegradable polymer poly (dl-lactide) (PDLLA) has been used as drug coating material for drug-eluting stents due to its excellent biocompatibility and sustained drug release ability. However, the uniform thin layer drug eluting coating on a stent not only inhibits the blood vessel's smooth muscle cell overgrowth but also delay the endotheliation process which is often associated with the occurrence of acute thrombosis. Therefore, in this study, we developed a novel coating method using PDLLA nanoparticles (NPs) as a coating to overcome this issue. The average 300 nm sized sirolimus-loaded PDLLA nanoparticles were prepared by a conventional emulsion solvent evaporation method. A low temperature plasma polymerization technology to graft hydrophilic polymers on to poly (l-lactide) stent was used to increase the surface coating efficiency of nanoparticles on the stent. Results showed that sirolimus-loaded nanoparticles can be successfully coated on to the stents with sustained drug release properties. In vitro cell culture study showed the drug loaded nanoparticle coating effectively inhibited the proliferation of smooth muscle cells while still allowed a faster proliferation of endothelial cells, suggesting that the new NP coated bioresorbable stents have the potential to reduce both the occurrence of in-stent restenosis and acute thrombosis. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 106B: 88-95, 2018.


Subject(s)
Absorbable Implants , Coated Materials, Biocompatible/chemistry , Endothelial Cells/metabolism , Graft Occlusion, Vascular/prevention & control , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nanoparticles/chemistry , Polyesters/chemistry , Stents , Blood Vessel Prosthesis , Cells, Cultured , Delayed-Action Preparations , Humans
19.
Cardiovasc Ther ; 36(2)2018 Apr.
Article in English | MEDLINE | ID: mdl-29283509

ABSTRACT

INTRODUCTION: Cardiovascular disease is the main cause of mortality and morbidity in the industrialized world. Incretin-mimetic compounds such as exenatide are currently used in the treatment of type 2 diabetes. AIMS: We investigated the effects of incretin drugs on apoptosis, adhesion molecule expression, and concentration of extracellular matrix (ECM) metalloproteinases under inflammatory conditions within the context of atherosclerotic plaque formation of both human coronary artery endothelial cells (hCAECs) and human aortic endothelial cells (hAoECs). TNF-α-stimulated hCAEC and hAoEC were treated with exenatide (1 and 10 nmol/L) and GLP-1 (10 and 100 nmol/L) then evaluated for caspase 3/7 activity and assayed for protein levels of adhesion molecules sICAM-1, sVCAM-1, and P-selectin. Concentrations of matrix metalloproteinases (MMPs) MMP-1, MMP-2, MMP-9, and their inhibitors-tissue inhibitor of metalloproteinases (TIMPs), TIMP-1, TIMP-2 were also measured to evaluate the effects on extracellular matrix turnover within an inflammatory environment. Intracellular signaling pathways were evaluated via transfection of endothelial cells with a GFP vector under the NF-κB promoter. RESULTS: Our experimental data suggest that GLP-1 receptor (GLP-1R) agonists downregulate activation of NF-κB and adhesion molecules ICAM and VCAM, but not P-selectin, in both endothelial cell lines. Exendin-4 and GLP-1 modulate the expression of MMPs and TIMPs, with statistically significant effects observed at high concentrations of both incretins. Expressive modulation may be mediated by NF-κB as observed by activation of the vector when stimulated under inflammatory conditions. CONCLUSION: These findings indicate that GLP-1 analogs have anti-inflammatory properties in endothelial cells that may play an important role in preventing atherosclerosis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cell Adhesion Molecules/metabolism , Endothelial Cells/drug effects , Matrix Metalloproteinases/metabolism , Peptides/pharmacology , Tissue Inhibitor of Metalloproteinases/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Venoms/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Cell Adhesion Molecules/immunology , Cells, Cultured , Dose-Response Relationship, Drug , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Exenatide , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , NF-kappa B/metabolism , Signal Transduction/drug effects
20.
Chongqing Medicine ; (36): 1009-1011,1015, 2018.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-691897

ABSTRACT

Objective To explore the molecular mechanisms of the effect of eluting stent drug rapamycin for injuring human coronary artery endothelial cells(HCAECs)by using the proteomics method.Methods HCAECs were treated with rapamycin,and the differentially expressed proteins were analyzed by two dimension fluorescence differential gel electrophoresis(2D-DIGE).The changed proteins were identified by MALDI-ToF-ToF.Results At least 85 differential protein spots were found,including 49 up-regulated and 36 down-regulated protein spots.Twenty-six proteins were identified by MALDI-ToF-ToF,including the endoplasmic reticulum protein,mitochondrial protein,molecular chaperones,ubiquitin system related protein,structural protein and oxidative stress related proteins,etc.Conclusion The changes of specific proteins of HCAECs injury induced by rapamycin are investigated by the proteomic method.

SELECTION OF CITATIONS
SEARCH DETAIL
...