Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Heliyon ; 10(4): e25569, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38384527

ABSTRACT

Background: Although dilated cardiomyopathy (DCM) is a prevalent form of cardiomyopathy, the molecular mechanisms underlying its pathogenesis and progression remain poorly understood. It is possible to identify and validate DCM-associated genes, pathways, and miRNAs using bioinformatics analysis coupled with clinical validation methods. Methods: Our analysis was performed using 3 mRNA datasets and 1 miRNA database. We employed several approaches, including gene ontology (GO) analysis, KEGG pathway enrichment analysis, protein-protein interaction networks analysis, and analysis of hub genes to identify critical genes and pathways linked to DCM. We constructed a regulatory network for DCM that involves interactions between miRNAs and mRNAs. We also validated the differently expressed miRNAs in clinical samples (87 DCM ,83 Normal) using qRT-PCR.The miRNAs' clinical value was evaluated by receiver operating characteristic curves (ROCs). Results: 78 differentially expressed genes (DEGs) and 170 differentially expressed miRNAs (DEMs) were associated with DCM. The top five GO annotations were collagen-containing extracellular matrix, cell substrate adhesion, negative regulation of cell differentiation, and inflammatory response. The most enriched KEGG pathways were the Neurotrophin signaling pathway, Thyroid hormone signaling pathway, Wnt signaling pathway, and Axon guidance. In the PPI network, we identified 10 hub genes, and in the miRNA-mRNA regulatory network, we identified 8 hub genes and 15 miRNAs. In the clinical validation, we found 13 miRNAs with an AUC value greater than 0.9. Conclusion: Our research offers novel insights into the underlying mechanisms of DCM and has implications for identifying potential targets for diagnosis and treatment of this condition.

2.
Sci Rep ; 14(1): 686, 2024 01 06.
Article in English | MEDLINE | ID: mdl-38182722

ABSTRACT

High altitude exposure increases the risk of myocardial ischemia (MI) and subsequent cardiovascular death. Machine learning techniques have been used to develop cardiovascular disease prediction models, but no reports exist for high altitude induced myocardial ischemia. Our objective was to establish a machine learning-based MI prediction model and identify key risk factors. Using a prospective cohort study, a predictive model was developed and validated for high-altitude MI. We consolidated the health examination and self-reported electronic questionnaire data (collected between January and June 2022 in 920th Joint Logistic Support Force Hospital of china) of soldiers undergoing high-altitude training, along with the health examination and second self-reported electronic questionnaire data (collected between December 2022 and January 2023) subsequent to their completion on the plateau, into a unified dataset. Participants were subsequently allocated to either the training or test dataset in a 3:1 ratio using random assignment. A predictive model based on clinical features, physical examination, and laboratory results was designed using the training dataset, and the model's performance was evaluated using the area under the receiver operating characteristic curve score (AUC) in the test dataset. Using the training dataset (n = 2141), we developed a myocardial ischemia prediction model with high accuracy (AUC = 0.86) when validated on the test dataset (n = 714). The model was based on five laboratory results: Eosinophils percentage (Eos.Per), Globulin (G), Ca, Glucose (GLU), and Aspartate aminotransferase (AST). Our concise and accurate high-altitude myocardial ischemia incidence prediction model, based on five laboratory results, may be used to identify risks in advance and help individuals and groups prepare before entering high-altitude areas. Further external validation, including female and different age groups, is necessary.


Subject(s)
Coronary Artery Disease , Myocardial Ischemia , Female , Humans , Cohort Studies , Altitude , Prospective Studies , Myocardial Ischemia/diagnosis , Myocardial Ischemia/etiology , Machine Learning
3.
Annu Int Conf IEEE Eng Med Biol Soc ; 2019: 6298-6301, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31947282

ABSTRACT

In this paper, we proposed and validated a novel and accurate pipeline for automatically segmenting flaky corneal ulcer areas from fluorescein staining images. The ulcer area was segmented within the cornea by employing a joint method of Otsu and Gaussian Mixture Modeling (GMM). In the GMM based segmentation, the total number of Gaussians was determined intelligently using an information theory based algorithm. And the fluorescein staining images were processed in the HSV color model rather than the original RGB color model, aiming to improve the segmentation results' robustness and accuracy. In the Otsu based segmentation, the images were processed in the grayscale space with Gamma correction being conducted before the Otsu binarization. Afterwards, morphological operations and median filtering were employed to further improve the Otsu segmentation result. The GMM and Otsu segmentation results were then intersected, for which post-processing was conducted by identifying and filling holes through a fast algorithm using priority queues of pixels. The proposed pipeline has been validated on a total of 150 clinical images. Accurate ulcer segmentation results have been obtained, with the mean Dice Similarity Coefficient (DSC) being 0.88 when comparing the automatic segmentation result with the manually-delineated gold standard. For images in the RGB color space, the mean DSC was 0.83, being much lower than that of the images in the HSV color space.


Subject(s)
Corneal Ulcer/diagnostic imaging , Image Processing, Computer-Assisted , Algorithms , Color , Humans , Normal Distribution
4.
Biochem Biophys Res Commun ; 509(2): 359-366, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30591217

ABSTRACT

Cell death and inflammation play critical roles in atherosclerosis. Pyroptosis, a novel proinflammatory programmed cell death process, participates in atherosclerosis pathogenesis. Recently, MALAT1 was identified as a pyroptosis-related long noncoding RNA (lncRNA). Here, we investigated the potential role and underlying mechanism of lncRNA MALAT1 in endothelial cells pyroptosis. We first established an endothelial cell pyroptosis model by stimulating EA.hy926 human endothelial cells (EA.hy926 cells) with high glucose. Then, we investigated lncRNA MALAT1 expression and found that it was upregulated in high glucose-treated EA.hy926 cells. Furthermore, lncRNA MALAT1 knockdown significantly inhibited high glucose-induced pyroptosis in EA.hy926 cells, which may critically influence atherosclerosis. Moreover, miR-22 was a target of lncRNA MALAT1 and was negatively correlated with lncRNA MALAT1. NLRP3 expression was significantly suppressed by transfection with a MALAT1-targeting antisense oligonucleotide (ASO). Ultimately, miR-22 overexpression abrogated the effect of MALAT1 on high glucose-induced EA.hy926 cells pyroptosis. Together, our results suggest that lncRNA MALAT1 promotes high glucose-induced pyroptosis of endothelial cells partly by affecting NLRP3 expression through competitively binding miR-22. Our findings indicate a new regulatory mechanism for endothelial cells pyroptosis under high-glucose stress, providing a novel therapeutic target for atherosclerosis.


Subject(s)
Endothelial Cells/drug effects , Glucose/pharmacology , MicroRNAs/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Pyroptosis/genetics , RNA, Long Noncoding/genetics , Base Sequence , Binding Sites , Binding, Competitive , Cell Line , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Regulation , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , MicroRNAs/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oligoribonucleotides, Antisense/genetics , Oligoribonucleotides, Antisense/metabolism , Pyroptosis/drug effects , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/metabolism , Signal Transduction
5.
Clin Chem Lab Med ; 56(9): 1447-1457, 2018 08 28.
Article in English | MEDLINE | ID: mdl-29683797

ABSTRACT

BACKGROUND: Measurement uncertainty (MU) is a parameter associated with the result of a measurement that characterizes its dispersion. We report results for estimating MU following the application of a top-down procedure using only proficiency test data to establish uncertainty levels for various analytes. METHODS: Data were obtained from 142 laboratories participating in the Beijing Center for Clinical Laboratory (BCCL) proficiency testing/external quality assessment (PT/EQA) schemes. The 24-month study included six selected PT shipments to obtain estimates for 50th percentile (median) and 90th percentile MUs and to compare those estimates to usual analytic goals. The number of laboratory participants varied for each trial. The expanded uncertainty (U) was calculated using a cover factor of k=2 for a confidence interval of 95%. All reproducibility, method and laboratory biases came from the PT/EQA data. RESULTS: The median U (k=2) ranged from 3.2% (plasma sodium, indirect ion selective electrode) to 32.8% (triglycerides, free glycerol blanking) for clinical chemistry analyte means from participants in the same method group. Immunoassay analyte median U results ranged from 11.3% (CA125 tumor marker, Roche) to 33.8% (prostate-specific antigen [PSA], Abbott). The range for median U was 3.5% (red blood cell [RBC], Abx) to 30.3% (fibrinogen [FBG], other) for hematology and coagulation analytes. The MUs for most analytes satisfied quality requirements. CONCLUSIONS: The use of PT/EQA data, when available, provides an effective means for estimating uncertainties associated with quantitative measurements. Thus, medical laboratories can calculate their own MUs. Proficiency testing organizers can provide participants with an additional MU estimate using only EQA data, which may be updated at the end of each survey.


Subject(s)
Blood Coagulation Tests/standards , Clinical Chemistry Tests/standards , Immunoassay/standards , Laboratories/standards , Uncertainty , Quality Assurance, Health Care , Quality Control , Reproducibility of Results
6.
RSC Adv ; 8(27): 15036-15043, 2018 Apr 18.
Article in English | MEDLINE | ID: mdl-35541353

ABSTRACT

A new cycloartane triterpene, yunnanterpene G (1), containing an oxaspiro[5.4]decane moiety, was purified from the roots of Cimicifuga foetida. The new structure was determined from spectroscopic data and the X-ray diffraction method. Biological evaluations revealed that compound 1 significantly inhibited the mRNA expression of the atherosclerosis-related adhesion molecule CD147 (extracellular matrix metalloproteinase inducer, EMMPRIN), and proteolytic enzymes matrix metalloproteinase 2 (MMP-2), MMP-9 and MMP-14, in a dose-dependent manner in phorbol-12-myristate-13-acetate-induced human monocytic THP-1 cells by quantitative real-time PCR method. At the same time, the migration ability of the induced THP-1 cells was potently inhibited. Furthermore, western blot experiments showed that compound 1 at 25 µM strongly suppressed phosphorylation of NF-κB p65 and p38 MAPK in the differentiated THP-1 cells.

7.
Cell Physiol Biochem ; 43(4): 1436-1448, 2017.
Article in English | MEDLINE | ID: mdl-29017169

ABSTRACT

BACKGROUND/AIMS: Autophagy, an evolutionary conserved biological process, is activated in cells to cope with various types of stress. MicroRNAs control several activities related to autophagy. However, the role of autophagy-related microRNAs during atherosclerosis is far from known. MicroRNA-155 was identified to be a crucial regulator of atherosclerosis. The objectives of the study were to analyze the effect of microRNA-155 on autophagic signaling and explore its mechanism in human endothelial cells under ox-LDL stress. METHODS: The study included human endothelial cells surrogate EA.hy926 lines (EA.hy926 cells). The expression of microRNA-155 was analyzed by quantitative reverse transcription polymerase chain reaction (qRT-PCR). The effect of microRNA-155 on endothelial autophagy was observed along with the expression levels of Rheb, LC3B, Beclin1, and P62/SQSTM1 by western blotting (WB) and immunofluorescence through microRNA-155 overexpression or inhibition. Bioinformatics analysis and Luciferase reporter assay were used to explore the target gene of microRNA-155. Cell viability and apoptosis were examined by 3-[4,5-dimethylthiazol-2-yl]-5- [3-carboxy-methoxyphenyl]-2-[4-sulfophenyl]-2H-tetrazolium inner salt (MTS) assay and TdT-mediated dUTP Nick-End Labeling (TUNEL) apoptosis assay. RESULTS: MicroRNA-155 expression was significantly increased under ox-LDL stress. MicroRNA-155 increased autophagic activity, while inhibition of it alleviated ox-LDL-induced autophagy in EA.hy926 endothelial cells. In addition, dual-luciferase reporter assays showed that microRNA-155 suppressed Rheb transcription. MicroRNA-155 increased autophagic activity in EA.hy926 cells via inhibition of Rheb-mediated mTOR/P70S6kinase/4EBP signaling pathway. Furthermore, we demonstrated that microRNA-155 could regulate not only autophagy but also apoptosis in EA.hy926 cells. CONCLUSIONS: MicroRNA-155 works as a regulator of endothelial function under ox-LDL stress, making it a potential candidate for the novel therapeutic strategies against atherosclerotic diseases.


Subject(s)
Autophagy , Endothelial Cells/metabolism , Lipoproteins, LDL/metabolism , MicroRNAs/genetics , Ras Homolog Enriched in Brain Protein/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Apoptosis , Atherosclerosis/genetics , Atherosclerosis/metabolism , Cell Line , Cell Survival , Endothelial Cells/cytology , Humans , Up-Regulation
8.
Exp Ther Med ; 13(3): 835-844, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28450907

ABSTRACT

Extracellular matrix metalloproteinase inducer (EMMPRIN) reportedly has a key regulatory role in matrix metalloproteinase (MMP) activities and the progression of atherosclerosis. Statins, which are anti-atherosclerotic pharmacological agents, are widely applied in clinical settings. The aim of the present study was to investigate the pharmaceutical effect of atorvastatin on EMMPRIN expression in atherosclerotic plaques. An atherosclerotic mouse model was established using apoliprotein E-deficient (ApoE-/-) mice raised on a high-fat diet. Additionally, a low (5 mg/kg/day) or high dosage (10 mg/kg/day) of atorvastatin suspension was administered orally for eight weeks, beginning on week 7 or 11 respectively. The effects of atorvastatin on atherosclerotic plaque formation and EMMPRIN expression were subsequently determined. The THP-1 cell line was used to investigate the effect of atorvastatin on EMMPRIN expression in vitro. The results demonstrated that the high-fat diet led to vulnerable plaques (VPs) and increased EMMPRIN expression in VPs in ApoE-/- mice. Atorvastatin treatment decreased EMMPRIN expression in the aortas and plaques of ApoE-/- mice. In vitro, oxidized low-density lipoprotein (ox-LDL) induced the expression of cyclooxygenase-2 (COX-2) and EMMPRIN in THP-1 macrophages, and atorvastatin inhibited ox-LDL-induced expression of PGE2, EMMPRIN and COX-2 in THP-1 macrophages. Therefore, the present data indicated that atorvastatin treatment reduces the vulnerability of atherosclerotic plaques and expression of EMMPRIN, and that the inhibitory effect of atorvastatin on EMMPRIN may occur via the COX-2/PGE2 signaling pathway in macrophages.

9.
Mediators Inflamm ; 2016: 8060182, 2016.
Article in English | MEDLINE | ID: mdl-27843203

ABSTRACT

Inflammation response plays a critical role in all phases of atherosclerosis (AS). Increased evidence has demonstrated that miR-155 mediates inflammatory mediators in macrophages to promote plaque formation and rupture. However, the precise mechanism of miR-155 remains unclear in AS. Here, we also found that miR-155 and PDCD4 were elevated in the aortic tissue of atherosclerotic mice and ox-LDL treated RAW264.7 cells. Further studies showed that miR-155 not only directly inhibited SOCS1 expression, but also increased the expression of p-STAT and PDCD4, as well as the production of proinflammation mediators IL-6 and TNF-α. Downregulation of miR-155 and PDCD4 and upregulation of SOCS1 obviously decreased the IL-6 and TNF-α expression. In addition, inhibition of miR-155 levels in atherosclerotic mice could notably reduce the IL-6 and TNF-α level in plasma and aortic tissue, accompanied with increased p-STAT3 and PDCD4 and decreased SOCS1. Thus, miR-155 might mediate the inflammation in AS via the SOCS1-STAT3-PDCD4 axis. These results provide a rationale for intervention of intracellular miR-155 as possible antiatherosclerotic targets.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Atherosclerosis/metabolism , Inflammation/metabolism , MicroRNAs/metabolism , RNA-Binding Proteins/metabolism , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 1 Protein/metabolism , Animals , Aorta/metabolism , Apolipoproteins E/metabolism , Male , Mice , RAW 264.7 Cells , RNA, Small Interfering/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
10.
Coron Artery Dis ; 25(4): 304-10, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24525789

ABSTRACT

OBJECTIVES: This study aimed to investigate the association between microRNA-155 (miR-155) and the severity and extent of coronary stenotic lesions. PATIENTS AND METHODS: We measured the miR-155 expression by real-time PCR in 110 consecutive patients undergoing coronary angiography for suspected coronary artery disease. The severity and extent of coronary stenotic lesions were evaluated on the basis of coronary angiography findings by the Gensini score. RESULTS: The miR-155 expression was significantly lower in 56 patients with coronary heart disease than those in 54 controls (P<0.01). The level of miR-155 in peripheral blood mononuclear cells or plasma was lower in patients with unstable angina pectoris and acute myocardial infarction than in patients with chest pain syndrome, whereas no statistically significant differences were observed between patients with stable angina pectoris and chest pain syndrome. Spearman's correlation analysis showed that the expression of miR-155 in plasma correlated positively with the expression in peripheral blood mononuclear cells. The levels of miR-155 in the patients with diseased vessels of two and three or more were significantly lower than in those with diseased vessel of zero and one. The levels of miR-155 were not significantly different among groups with diseased vessels of zero and one. miR-155 were associated negatively with Gensini scores (r = -0.663, P<0.001). The miR-155 expression was correlated significantly to age (r = -0.227), hypertension (r = -0.440), total cholesterol (r = 0.239), high-density lipoprotein cholesterol (r = 0.280), low-density lipoprotein cholesterol (r = -0.315), tobacco use (r = -0.363), angiotensin-converting enzyme inhibitor (r = -0.250), statins (r = -0.368), and high-sensitivity C-reactive protein (r = -0.515). CONCLUSION: miR-155 expression is associated inversely with complicated proatherogenic metabolic risk factors, and the severity of coronary stenotic lesions calculated by Gensini scores.


Subject(s)
Coronary Angiography , Coronary Stenosis/diagnosis , Coronary Vessels/diagnostic imaging , Genetic Testing , MicroRNAs/blood , Aged , Angina, Unstable/diagnostic imaging , Angina, Unstable/genetics , Case-Control Studies , Coronary Stenosis/blood , Coronary Stenosis/diagnostic imaging , Coronary Stenosis/genetics , Disease Progression , Female , Genetic Markers , Genetic Testing/methods , Humans , Male , Middle Aged , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/genetics , Predictive Value of Tests , Real-Time Polymerase Chain Reaction , Severity of Illness Index
11.
Mol Cell Biochem ; 382(1-2): 253-61, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23797321

ABSTRACT

Macrophage apoptosis is a prominent feature of advanced atherosclerotic plaques. Here, we examined the hypothesis that the apoptotic machinery is regulated by microRNA-155 (miR-155). Constitutive expression of miR-155 was detected in RAW264.7 cells, which was increased following stimulation with oxidized low-density lipoprotein (OxLDL) in a dose- and time-dependent manner. OxLDL-treated RAW264.7 cells showed a marked time- and dose-dependent increase in apoptosis, which was suppressed in the presence of mimics and increased with antagonists of miR-155. Bioinformatics analysis revealed Fas-associated death domain-containing protein (FADD) as a putative target of miR-155. Luciferase reporter assay and Western blot further disclosed that miR-155 inhibits FADD expression by directly targeting the 3'-UTR region. We propose that miR-155 attenuates the macrophage apoptosis, at least in part, through FADD regulation, since forced expression of FADD blocked the ability of miR-155 to inhibit apoptosis. Our results collectively suggest that miR-155 attenuates apoptosis of OxLDL-mediated RAW264.7 cells by targeting FADD, supporting a possible therapeutic role in atherosclerosis.


Subject(s)
Apoptosis/drug effects , Lipoproteins, LDL/pharmacology , Macrophages/cytology , Macrophages/metabolism , MicroRNAs/metabolism , 3' Untranslated Regions/genetics , Animals , Apoptosis/genetics , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Cytoprotection/drug effects , Cytoprotection/genetics , Fas-Associated Death Domain Protein/metabolism , Gene Expression Regulation/drug effects , Macrophages/drug effects , Mice , MicroRNAs/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
Int J Cardiol ; 168(4): 3248-53, 2013 Oct 09.
Article in English | MEDLINE | ID: mdl-23642811

ABSTRACT

BACKGROUND: Extracellular matrix metalloproteinase inducer (EMMPRIN), a 58-kDa cell surface glycoprotein, has been identified as a key receptor for transmitting cellular signals mediating metalloproteinase activities, as well as inflammation and oxidative stress. Clinical evidence has revealed that EMMPRIN is expressed in human atherosclerotic plaque; however, the relationship between EMMPRIN and atherosclerosis is unclear. To evaluate the functional role of EMMPRIN in atherosclerosis, we treated apolipoprotein E-deficient (ApoE(-/-)) mice with an EMMPRIN function-blocking antibody. METHODS AND RESULTS: EMMPRIN was found to be up-regulated in ApoE(-/-) mice fed a 12-week high-fat diet in contrast to 12 weeks of normal diet. Administration of a function-blocking EMMPRIN antibody (100 µg, twice per week for 4 weeks) to ApoE(-/-) mice, starting after 12 weeks of high-fat diet feeding caused attenuated and more stable atherosclerotic lesions, less reactive oxygen stress generation on plaque, as well as down-regulation of circulating interleukin-6 and monocyte chemotactic protein-1 in ApoE(-/-) mice. The benefit of EMMPRIN functional blockage was associated with reduced metalloproteinases proteolytic activity, which delayed the circulating monocyte transmigrating into atherosclerotic lesions. CONCLUSION: EMMPRIN antibody intervention ameliorated atherosclerosis in ApoE(-/-) mice by the down-regulation of metalloproteinase activity, suggesting that EMMPRIN may be a viable therapeutic target in atherosclerosis.


Subject(s)
Antibodies, Blocking/pharmacology , Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Basigin/metabolism , Animals , Antibodies, Blocking/therapeutic use , Atherosclerosis/etiology , Atherosclerosis/prevention & control , Basigin/immunology , Basigin/physiology , Diet, High-Fat/adverse effects , Diet, High-Fat/methods , Down-Regulation/genetics , Male , Metalloproteases/antagonists & inhibitors , Metalloproteases/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Proteolysis , Random Allocation
13.
Can J Cardiol ; 29(8): 997-1005, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23395513

ABSTRACT

BACKGROUND: Krüppel-like factor 2 (KLF2) and protease-activated receptor-1 (PAR-1) are 2 novel factors that play important roles in inflammation and coagulation, yet their relationship with vulnerable plaques and statin remains uncertain. The purpose of this study was to explore the expression of KLF2 and PAR-1 in vulnerable plaques of ApoE gene knockout (ApoE(-/-)) mice and the pharmaceutical effect of statin on the expression of KLF2 and PAR-1. METHODS: ApoE(-/-) mice were randomized into an early-treatment group and a late-treatment group. Each group was randomized into 4 subgroups: normal diet control, high-fat diet control, high-dose statin, and low-dose statin. After 8 weeks of intervention with statin, the aortas were harvested to determine KLF2 and PAR-1 expression by semiquantitative reverse transcription-polymerase chain reaction, Western blotting, and immunohistochemical analyses. The relative area of plaque, the ratio of fibrous cap thickness to tunica intima/media thickness, and the vulnerability index of atherosclerotic plaques were calculated to evaluate plaque vulnerability. RESULTS: KLF2 increased whereas PAR-1 decreased in vulnerable plaques at messenger RNA, protein, and histologic levels. Atorvastatin increased KLF2 and decreased PAR-1 expression. CONCLUSIONS: Plaque vulnerability correlated negatively with KLF2 but positively with PAR-1. Upregulation of KLF2 and downregulation of PAR-1 could be direct or indirect effects of statin therapy.


Subject(s)
Heptanoic Acids/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Kruppel-Like Transcription Factors/physiology , Plaque, Atherosclerotic/drug therapy , Pyrroles/therapeutic use , Receptor, PAR-1/physiology , Animals , Apolipoproteins E/genetics , Atorvastatin , Blotting, Western , Disease Models, Animal , Immunohistochemistry , Mice , Mice, Knockout , Plaque, Atherosclerotic/pathology , Reverse Transcriptase Polymerase Chain Reaction
14.
Biochem Biophys Res Commun ; 398(2): 315-20, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20599714

ABSTRACT

Knockdown of stromal interaction molecule 1 (STIM1) significantly suppresses neointima hyperplasia after vascular injury. Endothelial progenitor cells (EPCs) are the major source of cells that respond to endothelium repair and contribute to re-endothelialization by reducing neointima formation after vascular injury. We hypothesized that the effect of STIM1 on neointima hyperplasia inhibition is mediated through its effect on the biological properties of EPCs. In this study, we investigated the effects of STIM1 on the proliferation and migration of EPCs and examined the effect of STIM1 knockdown using cultured rat bone marrow-derived EPCs. STIM1 was expressed in EPCs, and knockdown of STIM1 by adenoviral delivery of small interfering RNA (siRNA) significantly suppressed the proliferation and migration of EPCs. Furthermore, STIM1 knockdown decreased store-operated channel entry 48h after transfection. Replenishment with recombinant human STIM1 reversed the effects of STIM1 knockdown. Our data suggest that the store-operated transient receptor potential canonical 1 channel is involved in regulating the biological properties of EPCs through STIM1. STIM1 is a potent regulator of cell proliferation and migration in rat EPCs and may play an important role in the biological properties of EPCs.


Subject(s)
Cell Movement , Cell Proliferation , Endothelial Cells/physiology , Membrane Glycoproteins/physiology , Stem Cells/physiology , Animals , Gene Knockdown Techniques , Humans , Membrane Glycoproteins/genetics , RNA Interference , RNA, Small Interfering/genetics , Rats , Stromal Interaction Molecule 1
15.
Exp Biol Med (Maywood) ; 235(3): 317-25, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20404049

ABSTRACT

Increased Ca(2+) entry through store-operated Ca(2+) channels (SOCCs) plays an essential role in the regulation of hepatocyte growth factor (HGF)-induced cell proliferation. Stromal interaction molecule 1 (STIM1) is thought to transmit endoplasmic reticulum (ER) Ca(2+) store depletion signals to the plasma membrane (PM), causing the opening of SOCCs in the PM. However, the relationship between HGF and STIM1 in endothelial progenitor cell (EPC) proliferation remains uncharacterized. The objective of this study was to evaluate the potential involvement of STIM1 in HGF-induced EPC proliferation. For this purpose, we used cultured rat bone marrow-derived EPCs and found that HGF-induced EPC proliferation at low concentrations. Store-operated Ca(2+) entry (SOCE) was elevated in HGF-treated EPCs, and the SOCC inhibitors 2-aminoethoxydiphenyl borate (2-APB) and BTP-2 inhibited the HGF-induced proliferation response. Moreover, STIM1 mRNA and protein expression levels were increased in response to HGF stimulation and knockdown of STMI1 decreased SOCE and prevented HGF-induced EPC proliferation. In conclusion, our data suggest that HGF-induced EPC proliferation is mediated partly via activation of STIM1.


Subject(s)
Endothelial Cells/cytology , Hepatocyte Growth Factor/metabolism , Membrane Glycoproteins/metabolism , Stem Cells/cytology , Anilides/pharmacology , Animals , Bone Marrow Cells/cytology , Boron Compounds/pharmacology , Calcium/metabolism , Cell Proliferation , Microscopy, Fluorescence/methods , Models, Biological , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley , Stromal Interaction Molecule 1 , Thiadiazoles/pharmacology
16.
Mol Cell Biochem ; 335(1-2): 19-27, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19701608

ABSTRACT

Migration and proliferation of endothelial progenitor cells (EPCs) are the key mechanisms in re-endothelialization after vascular injury. Inhibitor of DNA binding-1 (Id1) function has been linked to the proliferation, migration, and senescence of cells, and studies have shed light on the relationship between Id1 and the biological functions of EPCs. On the basis of the available data concerning Id1 and the behavior of EPCs, we hypothesized that Id1 was an important regulator in modulating the migration and proliferation of EPCs. Culture of spleen-derived EPCs was done as previously described. Id1 was presented at low levels in EPCs. Id1 was localized predominantly in the cytoplasm, and was rapidly upregulated by stimulation with serum and vascular endothelial growth factor. The migration and proliferation of EPCs were extensively improved by overexpression of adenovirus-mediated exogenous Id1 and inhibited by silencing of endogenous Id1 in EPCs. These results suggest that Id1 has a direct role in regulation of the migration and proliferation in EPCs.


Subject(s)
Cell Movement , Cell Proliferation , Endothelial Cells/cytology , Inhibitor of Differentiation Protein 1/physiology , Stem Cells/cytology , Animals , Endothelial Cells/metabolism , Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/metabolism , Male , Mice , Rats , Rats, Sprague-Dawley , Stem Cells/metabolism
17.
Peptides ; 30(7): 1368-73, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19394384

ABSTRACT

Endothelial dysfunction is associated with cardiovascular diseases. The Ca(2+) influx occurring via activation of plasmalemma Ca(2+) channels was shown to be critical in signaling the increase in endothelial permeability in response to a variety of permeability-increasing mediators. It has been reported that angiotensin II (AngII) could induce Ca(2+) signaling in some cells, and transient receptor potential canonical 1 (TRPC1) had an important role in this process. The objective of this study was to examine the mechanism of AngII-induced Ca(2+) entry and vascular endothelial hyperpermeability. Human umbilical vein endothelial cells (HUVECs) exposed to AngII exhibited dose-dependent increase in [Ca(2+)]i and endothelial permeability. Quantitative real-time RT-PCR and Western blotting showed that the level of TRPC1 expression had increased significantly at 12h and at 24h after treatment of HUEVCs with AngII. The expression of p65 was suppressed using an RNAi strategy. The results showed that the NF-kappaB signaling pathway and type-1 receptor of AngII was involved in AngII-induced TRPC1 upregulation. Moreover, knockdown of TRPC1 and NF-kappaB expression attenuates AngII-induced [Ca(2+)]i and endothelial permeability. NF-kappaB and TRPC1 have critical roles in AngII-induced Ca(2+) entry and endothelial permeability.


Subject(s)
Angiotensin II/pharmacology , Calcium/metabolism , Cell Membrane Permeability , Endothelial Cells/drug effects , Gene Expression Regulation/drug effects , NF-kappa B/metabolism , TRPC Cation Channels/metabolism , Blotting, Western , Cell Membrane Permeability/drug effects , Cell Membrane Permeability/genetics , Cells, Cultured , Electrophoretic Mobility Shift Assay , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Gene Knockdown Techniques , Humans , NF-kappa B/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , TRPC Cation Channels/genetics
18.
Cardiovasc Res ; 81(4): 660-8, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19052075

ABSTRACT

AIMS: There is evidence to suggest that stromal interaction molecule 1 (STIM1) functions as a Ca2+ sensor on the endoplasmic reticulum, leading to transduction of signals to the plasma membrane and opening of store-operated Ca2+ channels (SOC). SOC have been detected in vascular smooth muscle cells (VSMCs) and are thought to have an essential role in the regulation of contraction and cell proliferation. We hypothesized that knockdown of STIM1 inhibits VSMC proliferation and suppresses neointimal hyperplasia. METHODS AND RESULTS: We examined the effect of the knockdown of STIM1 using a rat balloon injury model and cultured rat aortic VSMCs. Interestingly, knockdown of rat STIM1 by adenovirus delivery of small interfering RNA (siRNA) significantly suppressed neointimal hyperplasia in a rat carotid artery balloon injury model at 14 days after injury. The re-expression of human STIM1 to smooth muscle reversed the effect of STIM1 knockdown on neointimal formation. Rat aortic VSMCs were used for the in vitro assays. Knockdown of endogenous STIM1 significantly inhibited proliferation and migration of VSMCs. Moreover, STIM1 knockdown induced cell-cycle arrest in G0/G1 and resulted in a marked decrease in SOC. Replenishment with recombinant human STIM1 reversed the effect of siRNA knockdown. These results suggest STIM1 has a critical role in neointimal formation in a rat model of vascular injury. CONCLUSION: STIM1 may represent a novel therapeutic target in the prevention of restenosis after vascular interventions.


Subject(s)
Carotid Artery Injuries/metabolism , Cell Proliferation , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neoplasm Proteins/metabolism , Actins/metabolism , Angioplasty, Balloon/adverse effects , Animals , Calcium/metabolism , Carotid Artery Injuries/etiology , Carotid Artery Injuries/pathology , Cell Cycle , Cell Movement , Cells, Cultured , Disease Models, Animal , Gene Knockdown Techniques , Humans , Hyperplasia , Male , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Muscle, Smooth, Vascular/injuries , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Neoplasm Proteins/genetics , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Rats , Rats, Wistar , Recombinant Proteins/metabolism , Stromal Interaction Molecule 1 , Time Factors , Transduction, Genetic
19.
Zhonghua Xin Xue Guan Bing Za Zhi ; 36(10): 912-5, 2008 Oct.
Article in Chinese | MEDLINE | ID: mdl-19102890

ABSTRACT

OBJECTIVE: To investigate the relationship between the plasma macrophage migration inhibitory factor (MIF), activator protein-1 (AP-1) and MMP-9 concentrations and the severity of coronary artery lesions in coronary heart disease (CHD) patients. METHODS: Patients were divided into normal controls (n = 35), stable angina pectoris (SAP, n = 32) and acute coronary syndrome (ACS, n = 75) according to the coronary angiography (CAG), clinical and laboratory examinations. The CAG severity and extent of coronary lesions were analyzed by means of Gensini coronary score system. Enzyme linked immunosorent assay was used to measure the plasma MIF, AP-1 and MMP-9 concentrations. RESULTS: Plasma MIF, AP-1 and MMP-9 concentrations were significant increased in CHD patients [MIF: (14.97 +/- 2.11) microg/L, AP-1: 1.43 +/- 0.33, MMP-9: (1.48 +/- 0.14) microg/L] compared to those in control group [MIF: (9.07 +/- 1.28) microg/L, AP-1: 0.71 +/- 0.13, MMP-9: (1.01 +/- 0.07) microg/L, all P < 0.05]. The MIF, AP-1 and MMP-9 concentrations in ACS group [MIF: (16.66 +/- 2.56) microg/L, AP-1: 1.56 +/- 0.22, MMP-9: (1.58 +/- 0.14) microg/L] were also significant higher than those in SAP group [MIF: (11.01 +/- 2.12) microg/L, AP-1: 1.04 +/- 0.25, MMP-9: (1.25 +/- 0.07) microg/L, all P < 0.05] and there was significant positive correlation between MIF, AP-1 and MMP-9 concentrations and the Gensini score of coronary artery lesions (all P < 0.05). AP-1 was positively correlated with MMP-9 in CHD patients (P < 0.05). CONCLUSIONS: Plasma MIF, AP-1 and MMP-9 concentrations were positively correlated to the severity of coronary lesions in CHD patients. Higher MIF, AP-1 and MMP-9 concentrations in ACS patients than in SAP patients might suggest higher plaque instability in ACS patients.


Subject(s)
Coronary Artery Disease , Macrophage Migration-Inhibitory Factors , Coronary Angiography , Coronary Artery Disease/blood , Humans , Plaque, Atherosclerotic
20.
Zhonghua Nei Ke Za Zhi ; 47(7): 545-7, 2008 Jul.
Article in Chinese | MEDLINE | ID: mdl-19035162

ABSTRACT

OBJECTIVE: To investigate the relation between activator protein-1 (AP-1) and coronary atherosclerotic changes and the potential role of AP-1 in the stabilization of atherosclerotic plaques in patients with coronary heart disease (CHD). METHOD: 142 patients were included in this study and divided into CHD group (107) and control group (35) according to coronary angiography (CAG). The CHD group was further divided into a stable angina pectoris (SAP) group (32) and an acute coronary syndrome (ACS) group (75) according to the clinical manifestations. In addition, the CHD group was divided into A type group, B type group and C type group according to the standard of ACC/AHA coronary change in 1988. Meanwhile, the CHD group was further divided into light stenosis group, moderate stenosis group and severe stenosis group according to the degree of coronary lesion. The lysate of cells was obtained through lysis of the leucocytes from peripheral blood with cell lysis buffer. The amount of Phospho-c-Jun in lysate was measured with enzyme-linked immunosorbent assay (ELISA). The results were demonstrated with absorbance, which reflects the amount of AP-1. RESULTS: The main coronary changes in the SAP group were A type (68.7%) and the changes were mainly of light degree (53.1%); the main coronary changes in the ACS group were B type (52.0%) or C type (37.3%) and the changes were mainly of heavy degree (66.7%). The absorbance of Phospho-c-Jun in CHD group was significantly higher than that in the control subjects (1.43 +/- 0.33 vs 0.71 +/- 0.13, P < 0.001). The absorbance of Phospho-c-Jun in the ACS group was significantly higher than that in the SAP group (1.56 +/- 0.28 vs 1.14 +/- 0.25, P < 0.001). The absorbance of Phospho-c-Jun increased gradually from A type group to C type group (1.18 +/- 0.27 vs 1.42 +/- 0.26 vs 1.71 +/- 0.27, P < 0.001) and from light stenosis group to severe stenosis group (1.09 +/- 0.20 vs 1.37 +/- 0. 26 vs 1.60 +/- 0.29, P < 0.001). CONCLUSION: There is a significant relationship between AP-1 and coronary atherosclerotic changes. AP-1 may be a factor that can predict coronary arteriosclerotic progression and stability of the plaque.


Subject(s)
Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Transcription Factor AP-1/biosynthesis , Adult , Aged , Coronary Angiography , Female , Genes, jun , Humans , Male , Middle Aged , Oxidative Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL
...