Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 40
1.
Sci Bull (Beijing) ; 2024 Mar 27.
Article En | MEDLINE | ID: mdl-38637226

Currently, clinically available coronary CT angiography (CCTA) derived fractional flow reserve (CT-FFR) is time-consuming and complex. We propose a novel artificial intelligence-based fully-automated, on-site CT-FFR technology, which combines the automated coronary plaque segmentation and luminal extraction model with reduced order 3 dimentional (3D) computational fluid dynamics. A total of 463 consecutive patients with 600 vessels from the updated China CT-FFR study in Cohort 1 undergoing both CCTA and invasive fractional flow reserve (FFR) within 90 d were collected for diagnostic performance evaluation. For Cohort 2, a total of 901 chronic coronary syndromes patients with index CT-FFR and clinical outcomes at 3-year follow-up were retrospectively analyzed. In Cohort 3, the association between index CT-FFR from triple-rule-out CTA and major adverse cardiac events in patients with acute chest pain from the emergency department was further evaluated. The diagnostic accuracy of this CT-FFR in Cohort 1 was 0.82 with an area under the curve of 0.82 on a per-patient level. Compared with the manually dependent CT-FFR techniques, the operation time of this technique was substantially shortened by 3 times and the number of clicks from about 60 to 1. This CT-FFR technique has a highly successful (> 99%) calculation rate and also provides superior prediction value for major adverse cardiac events than CCTA alone both in patients with chronic coronary syndromes and acute chest pain. Thus, the novel artificial intelligence-based fully automated, on-site CT-FFR technique can function as an objective and convenient tool for coronary stenosis functional evaluation in the real-world clinical setting.

3.
Transl Res ; 255: 1-13, 2023 05.
Article En | MEDLINE | ID: mdl-36384204

Cell inflammation and death are closely linked processes contributing to endothelial dysfunction, which plays a critical role in atherogenesis. Activation of the NLRP3 inflammasome causes pyroptosis, the Gasdermin D (GSDMD)-mediated inflammatory cell death. The non-canonical NF-κB pathway has been implicated in inflammation; however, its role in NLRP3 inflammasome-mediated endothelial dysfunction has not been investigated. This study investigated a role for the non-canonical NF-κB pathway in regulating endothelial pyroptosis as it relates to atherogenesis. Immunohistochemistry indicated inflammasome activation in the endothelial cells (EC) of human atherosclerotic arteries. Flow cytometry and Western blot analysis revealed that oxidized low-density lipoprotein (oxLDL) activated the NLRP3 inflammasome, concomitant with the activation of non-canonical NF-κB in primary human aortic EC. Interference of NF-κB inducing kinase (NIK), the key regulator of the non-canonical pathway, significantly attenuated oxLDL- or LPS/ATP-induced NLRP3 inflammasome activation, pyroptosis, IL-1ß, and IL-18 secretion. In contrast, overexpression of NIK exacerbated these responses. Chromatin immunoprecipitation revealed that activation of the non-canonical NF-κB pathway upregulated the transcription factor IRF-1 through RelB/p52 binding to its promoter region at -782/-770. In addition to the known target CASP1, RNA sequencing further identified GSDMD as a target gene of IRF-1. IRF-1 but not RelB/p52 interacted with the GSDMD promoter at -526/-515 and the CASP1 promoter at -11/10 to promote the expression and CASP1-mediated activation of GSDMD. Consistent with the observations in cultured endothelium, endothelial-specific deficiency of NIK or IRF-1 attenuated atherosclerosis in high-fat diet-fed Apoe-null mice. These data demonstrate that the non-canonical NF-κB pathway contributes to NLRP3 inflammasome-mediated endothelial pyroptosis and the development of atherosclerosis through GSDMD activation in a manner dependent on IRF-1. Further investigation may facilitate the identification of specific therapeutic targets for atherosclerotic heart diseases.


Atherosclerosis , NF-kappa B , Mice , Animals , Humans , NF-kappa B/metabolism , Inflammasomes/metabolism , Pyroptosis/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Endothelial Cells/metabolism , Endothelium/metabolism , Inflammation
4.
Front Pharmacol ; 13: 868401, 2022.
Article En | MEDLINE | ID: mdl-35837280

ATP-sensitive potassium channels (KATP channels) play pivotal roles in excitable cells and link cellular metabolism with membrane excitability. The action potential converts electricity into dynamics by ion channel-mediated ion exchange to generate systole, involved in every heartbeat. Activation of the KATP channel repolarizes the membrane potential and decreases early afterdepolarization (EAD)-mediated arrhythmias. KATP channels in cardiomyocytes have less function under physiological conditions but they open during severe and prolonged anoxia due to a reduced ATP/ADP ratio, lessening cellular excitability and thus preventing action potential generation and cell contraction. Small active molecules activate and enhance the opening of the KATP channel, which induces the repolarization of the membrane and decreases the occurrence of malignant arrhythmia. Accumulated evidence indicates that mutation of KATP channels deteriorates the regulatory roles in mutation-related diseases. However, patients with mutations in KATP channels still have no efficient treatment. Hence, in this study, we describe the role of KATP channels and subunits in angiocardiopathy, summarize the mutations of the KATP channels and the functional regulation of small active molecules in KATP channels, elucidate the potential mechanisms of mutant KATP channels and provide insight into clinical therapeutic strategies.

5.
Commun Biol ; 5(1): 726, 2022 07 22.
Article En | MEDLINE | ID: mdl-35869262

Clinically used inhibitors of mammalian target of rapamycin (mTOR) negatively impacts endothelial-dependent vasodilatation (EDD) through unidentified mechanisms. Here we show that either the endothelium-specific deletion of Mtor to inhibit both mTOR complexes, or depletion of Raptor or Rictor to disrupt mTORC1 or mTORC2, causes impaired EDD, accompanied by reduced NO in the serum of mice. Consistently, inhibition of mTOR decreases NO production by human and mouse EC. Specifically, inhibition of mTORC1 suppresses eNOS gene expression, due to impairment in p70S6K-mediated posttranscriptional regulation of the transcription factor KLF2 expression. In contrast to mTORC1 inhibition, a positive-feedback between MAPK (p38 and JNK) activation and Nox2 upregulation contributes to the excessive generation of reactive oxygen species (ROS), which causes eNOS uncoupling and decreased NO bioavailability in mTORC2-inhibited EC. Adeno-associated virus-mediated EC-specific overexpression of KLF2 or suppression of Nox2 restores EDD function in endothelial mTORC1- or mTORC2-inhibited mice.


Nitric Oxide Synthase Type III/metabolism , TOR Serine-Threonine Kinases/metabolism , Vasodilation , Animals , Endothelium/metabolism , Humans , Mammals , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 2 , Mice , Sirolimus/pharmacology
6.
Bioengineered ; 13(4): 9628-9644, 2022 04.
Article En | MEDLINE | ID: mdl-35412431

Endothelial dysfunction is an important mechanism involved in myocardial ischemia-reperfusion (I/R) injury. We aimed to explore the effects of Oxycodone on myocardial I/R injury in vivo and in vitro to reveal its mechanisms related to Sigma-1 Receptor (SIGMAR1). A rat model of I/R-induced myocardial injury was developed. The ischemic area and myocardial histopathological changes after oxycodone addition were evaluated by TTC staining and H&E staining. LDH, CK-MB and cTnI levels were used to assess myocardial function. Then, the endothelial integrity was reflected by the expressions of ZO-1, Claudin-1 and Occludin. Afterward, ELISA, RT-qPCR, western blot and immunofluorescence assays were adopted for the detection of inflammation-related genes. SIGMAR1 expression in myocardial tissues induced by I/R and cardiac microvascular endothelial cells (CMECs) under hypoxic/reoxygenation (H/R) was determined using RT-qPCR and western blotting. Subsequently, after SIGMAR1 silencing or BD1047 addition (a SIGMAR1 antagonist), cell apoptosis and endothelial integrity were analyzed in the presence of Oxycodone in H/R-stimulated CMECs. Results indicated that Oxycodone decreased the ischemic area and improved myocardial function in myocardial I/R injury rat. Oxycodone improved myocardial histopathological injury and elevated endothelial integrity, evidenced by upregulated ZO-1, Claudin-1 and Occludin expressions. Moreover, inflammatory response was alleviated after Oxycodone administration. Molecular docking suggested that SIGMAR1 could directly bind to Oxycodone. Oxycodone elevated SIGMAR1 expression and SIGMAR1 deletion or BD1047 addition attenuated the impacts of Oxycodone on apoptosis and endothelial integrity of CMECs induced by H/R. Collectively, Oxycodone alleviates myocardial I/R injury in vivo and in vitro by binding to SIGMAR1.


Endothelial Cells , Myocardial Reperfusion Injury , Oxycodone , Receptors, sigma , Animals , Apoptosis , Claudin-1/metabolism , Claudin-1/pharmacology , Endothelial Cells/metabolism , Molecular Docking Simulation , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Occludin/metabolism , Occludin/pharmacology , Oxycodone/metabolism , Oxycodone/pharmacology , Rats , Receptors, sigma/metabolism , Sigma-1 Receptor
7.
ESC Heart Fail ; 9(2): 1370-1379, 2022 04.
Article En | MEDLINE | ID: mdl-35128826

AIMS: Heart failure (HF) is a chronic heart disease with a high incidence and mortality. Due to the regulatory complexity of gene coexpression networks, the underlying hub genes regulation in HF remain incompletely appreciated. We aimed to explore potential key modules and genes for HF using weighted gene coexpression network analysis (WGCNA). METHODS AND RESULTS: The expression profiles by high throughput sequencing of heart tissues samples from HF and non-HF samples were obtained from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) between HF and non-HF samples were firstly identified. Then, a coexpression network was constructed to identify key modules and potential hub genes. The biological functions of potential hub genes were analysed by Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses. Finally, a protein-protein interaction (PPI) network was constructed using the STRING online tool. A total of 135 DEGs (133 up-regulated and 2 down-regulated DEGs) between HF and non-HF samples were identified in the GSE135055 and GSE123976 datasets. Moreover, a total of 38 modules were screened based on WGCNA in the GSE135055 dataset, and six potential hub genes (UCK2, ASB1, CCNI, CUX1, IRX6, and STX16) were screened from the key module by setting the gene significance over 0.2 and the module membership over 0.8. Furthermore, 78 potential hub genes were obtained by taking the intersection of the 135 DEGs and all genes in the key module, and enrichment analysis revealed that they were mainly involved in the MAPK and PI3K-AKT signalling pathways. Finally, in a PPI network constructed with the 78 potential hub genes, CUX1 and ASB1 were identified as hub genes in HF because they were also identified as potential hub genes in the WGCNA. CONCLUSIONS: To the best of our knowledge, our study is the first to employ WGCNA to identify the key module and hub genes for HF. Our study identified a module and two genes that might play important roles in HF, which may provide potential biomarkers for the diagnosis of HF and improve our knowledge of the molecular mechanisms underlying HF.


Heart Failure , Phosphatidylinositol 3-Kinases , Biomarkers/metabolism , Cyclin I , Gene Expression Profiling/methods , Gene Regulatory Networks , Heart Failure/genetics , Homeodomain Proteins , Humans , Transcription Factors/genetics
8.
Diabetes Res Clin Pract ; 185: 109776, 2022 Mar.
Article En | MEDLINE | ID: mdl-35149165

BACKGROUND: In diet-induced arterial atherosclerosis, increased KCa3.1 channel was associated with atherosclerotic plaque progression and instability. Macrophages are involved in the formation of atherosclerotic plaques, and the release of inflammatory cytokines and oxygen free radicals promotes plaque progression. However, whether the macrophage KCa3.1 channel facilitates diabetes-accelerated atherosclerosis is still unclear. This study investigated atherosclerotic plaque in ApoE-/- mice regulated by the KCa3.1 channel. METHODS AND RESULTS: In vivo, blocking KCa3.1channel inhibit the development of the atherosclerotic lesion in diabetic ApoE-/- mice fed with a high-fat diet. In vitro, upregulation of KCa3.1 channel level occurred in RAW264.7 cells treated with HG plus ox-LDL in a time-dependent manner. Blocking KCa3.1 significantly reduced the uptake of ox-LDL in mice peritoneal macrophages. Further studies indicated the KCa3.1 siRNA and TRAM-34 (KCa3.1 inhibitor) attenuated the scavenger receptor CD36 expression via inhibiting STAT3 phosphorylation. CONCLUSION: Blockade of macrophage KCa3.1 channel inhibit cellular oxidized low-density lipoprotein accumulation and decrease proinflammation factors expression via STAT3/CD36 axis. This study provided a novel therapeutic target to reduce the risk of atherosclerosis development in diabetic patients.


Atherosclerosis , Diabetes Mellitus , Plaque, Atherosclerotic , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/prevention & control , CD36 Antigens/genetics , CD36 Antigens/metabolism , Humans , Lipoproteins, LDL , Mice , Mice, Knockout , Plaque, Atherosclerotic/etiology , STAT3 Transcription Factor/metabolism
9.
Exp Ther Med ; 23(2): 157, 2022 Feb.
Article En | MEDLINE | ID: mdl-35069838

Myocardial ischemia-reperfusion injury results in elevated reactive oxygen species (ROS) production and causes oxidative stress damage. Therefore, the current study aimed to investigate whether adaptor protein phosphotyrosine interacting with PH domain and leucine zipper 1 (APPL1) could induce the expression of antioxidant enzymes through AMP-activated protein kinase (AMPK) signaling in order to alleviate the injury caused by ischemia/hypoxia-reperfusion. Following induction of hypoxia-reoxygenation (H/R) injury in H9c2 cells, the liver kinase B1 (LKB1)/AMPK/acetyl-CoA carboxylase α (ACC) signaling pathway was investigated using western blot analysis, along with the detection of superoxide dismutase (SOD)2 and SOD3 expression. Additionally, cell viability was detected using a Cell Counting Kit-8 assay and ROS production was analyzed using ROS staining, whereas the expression levels of inflammatory mediators (TNF-α, monocyte chemoattractant protein 1 and IL-1ß), apoptosis mediators [cleaved caspase-3, cleaved poly (ADP-ribose) polymerase and Bcl-2] and nuclear factor erythroid 2-related factor 2 signaling pathway-related proteins were detected via western blot analysis following overexpression of APPL1 alone or in combination with compound C treatment (an AMPK inhibitor). The results indicated that H/R induction upregulated the phosphorylation levels of LKB1, AMPK and ACC, and decreased the expression levels of APPL1 and SOD enzyme activities. APPL1 overexpression increased the phosphorylation levels of LKB1, AMPK and ACC, SOD enzyme activity and cell viability whereas the expression levels of proinflammatory mediators and proapoptotic mediators, and the levels of ROS production were markedly decreased when compared with H/R group with empty plasmid transfection. APPL overexpression-mediated effects were significantly abrogated by compound C. Taken together, the data indicated that APPL1 inhibited ROS production and H/R-induced myocardial injury via the AMPK signaling pathway. Therefore, APPL1 may serve as a potential therapeutic target for myocardial H/R injury.

10.
Bioengineered ; 13(1): 407-417, 2022 01.
Article En | MEDLINE | ID: mdl-34783297

Dysregulated lipid metabolism of macrophages contributes to thrombosis and antiphospholipid syndrome (APS). The long non-coding RNAs (lncRNA) myocardial infarction-associated transcript 2 (Mirt2) has been reported to inhibit inflammation and lipid accumulation; therefore, this study intended to clarify whether Mirt2 served a role in lipid metabolism. THP-1-derived macrophages with or without Mirt2-knockdown or overexpression, were exposed to oxidized low-density lipoprotein (ox-LDL), then cell migration, lipid accumulation, cholesterol efflux and inflammation were assessed using wound healing, oil red staining, commercial kits and western blot assays. Besides, ML385 was used to treat THP-1-derived macrophages to inhibit nuclear factor erythroid-related factor 2 (NRF2) expression. The expression of proteins involved in the above processes were measured by western blot. Results demonstrated that phorbol 12-myristate 13-acetate (PMA) significantly increased Mirt2 expression in THP-1 cells. Mirt2-knockdown enhanced ox-LDL-induced macrophage migration, lipid accumulation, inflammation, and inhibited cholesterol efflux. By contrast, Mirt2 overexpression displayed the opposite effects. Furthermore, Mirt2-knockdown inhibited NRF2 signaling and enhanced mitogen-activated protein kinase (MAPK) signaling, while Mirt2 overexpression displayed the opposite effects. Finally, the NRF2 inhibitor ML385 significantly reversed the above effects of Mirt2. In summary, Mirt2 served an important role in regulating lipid metabolism in macrophages via inhibiting MAPK signaling and activating the NRF2 signaling pathway.


Cholesterol/metabolism , Lipoproteins, LDL/metabolism , Macrophages/cytology , NF-E2-Related Factor 2/metabolism , RNA, Long Noncoding/genetics , Tetradecanoylphorbol Acetate/pharmacology , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Humans , Lipid Metabolism/drug effects , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Macrophages/metabolism , Signal Transduction/drug effects , THP-1 Cells , Up-Regulation
11.
J Inflamm Res ; 14: 6799-6812, 2021.
Article En | MEDLINE | ID: mdl-34924765

BACKGROUND: Atherosclerosis is an aging-related disease, partly attributed to telomerase dysfunction. This study aims to investigate whether telomere dysfunction-related vascular aging is involved in the protection mechanism of melatonin (MLT) in atherosclerosis. METHODS: Young and aged ApoE-/- mice were used to establish atherosclerotic mice model. H&E staining and immunofluorescence assay were performed to detect endothelial cell injury and apoptosis. Inflammatory cytokines and oxidative stress-related factors were determined using corresponding commercial assay kits. Telomerase activity was detected by TRAP assay, and SA-ß-gal staining was conducted to evaluate cellular senescence. HUVECs were treated with H2O2 for 1 h to induce senescence. Western blot was performed to measure protein expression. RESULTS: An obvious vascular endothelial injury, reflected by excessive production of inflammatory cytokines, elevated ROS, MDA and SOD levels, and more apoptotic endothelial cells, was found in atherosclerotic mice, especially in aged mice, which were then greatly suppressed by MLT. In addition, telomere dysfunction and senescence occurred in atherosclerosis, especially in aged mice, while MLT significantly alleviated the conditions. CYP1A1, one of the targeted genes of MLT, was verified to be upregulated in atherosclerotic mice but downregulated by MLT. Furthermore, H2O2 induced a senescence model in HUVECs, which was accompanied with a remarkably increased cell viability loss and apoptosis rate, and a downregulated telomerase activity of HUVECs, and this phenomenon was strengthened by RHPS4, an inhibitor of telomerase activity. However, MLT could partly abolish these changes in H2O2- and RHPS4-treated HUVECs, demonstrating that MLT alleviated vascular endothelial injury by regulating senescence and telomerase activity. CONCLUSIONS: Collectively, this study provided evidence for the protective role of MLT in atherosclerosis through regulating telomere dysfunction-related vascular aging.

12.
Sci Rep ; 11(1): 24279, 2021 12 20.
Article En | MEDLINE | ID: mdl-34930980

With the rapid aging of the population, coronary artery disease (CAD) has become one of the most fatal chronic diseases. However, the genetic mechanism of CAD is still unclear. The purpose of this study is to construct the lncRNA-miRNA-mRNA regulatory network for CAD diseases and systematically identify differentially expressed genes in patients with coronary heart disease. In this study, two lncRNA datasets (GSE69587 and GSE113079) and a microRNA dataset (GSE105449) which contained 393 and 38 CAD samples were selected. In addition, two mRNA datasets which named GSE113079 (98 CAD samples) and GSE9820 (8 CAD samples) were selected to search the differentially expressed genes (DEGs). By comparing the expression data between CAD and control samples, a total of 1111 lncRNAs, 2595 mRNAs and 22 miRNAs were identified. Based on the DEGs, a lncRNA-miRNA-mRNA ceRNA network was constructed to explore the hub nodes in CAD. In the ceRNA network, the lncRNAs KCNQ1OT1 and H19 showed high connectivity with the nine miRNAs. GO and KEGG results showed that genes in ceRNA networks were mainly involved in nitrogen compound metabolic process, PI3K-Akt signaling pathway and retrograde endocannabinoid signaling. These findings will improve the understanding of the occurrence and development mechanism of CAD.


Coronary Artery Disease/genetics , MicroRNAs/genetics , Phosphatidylinositol 3-Kinases/genetics , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , Signal Transduction , Biomarkers/metabolism , Cardiovascular System , Computational Biology , Gene Expression Profiling , Gene Regulatory Networks , Humans , Macrophages/metabolism , MicroRNAs/metabolism , Permeability , Phosphatidylinositol 3-Kinases/metabolism
13.
Mol Med Rep ; 24(5)2021 Nov.
Article En | MEDLINE | ID: mdl-34476498

Gycyrrhizic acid (GA), an inhibitor of high mobility group box 1 (HMGB1), inhibits inflammatory responses and is involved in the occurrence and development of several inflammation­related diseases. However, the role of GA in the atherosclerotic lesions caused by diabetes mellitus (DM) remains unknown. In the present study, Sprague Dawley rats were selected to desi=gn a diabetic atherosclerosis (AS) model. Rats from the DM­AS group were subsequently divided into DM­AS, DM­AS + GA (50 mg/kg) and DM­AS + GA (150 mg/kg) groups. Biochemical analyzers were used to measure levels of blood glucose, fasting insulin, total cholesterol, total triglyceride, low­density lipoprotein and high­density lipoprotein. The number of plaques was recorded after collection of thoracic aortas from the rats. The intimal thickness of arterial tissue was detected by hematoxylin and eosin staining. The expression levels of CD68 and α­smooth muscle actin (α­SMA) were detected by immunohistochemistry. The expression of tumor necrosis factor­α, interleukin (IL)­6 and IL­1ß in the serum of the rats was detected by ELISA. The expression of fatty acid synthetase, sterol regulatory element binding protein 1C, HMGB1 and receptor for advanced glycation end products (RAGE) was detected by western blotting. Reverse transcription quantitative PCR was used to detect the mRNA expression of HMGB1 and RAGE. The results demonstrated that GA treatment could decrease the body weight, blood glucose level and biochemical parameters of AS DM rats in a dose­dependent manner. In addition, GA decreased the intimal thickness of carotid artery and the formation of plaque in rats with diabetic AS. Furthermore, GA inhibited macrophage activation and decreased α­SMA expression in vascular smooth muscle cells, and decreased the expression of proteins (FAS and SREBP­1c) and inflammatory factors. Taken together, the findings from the present study demonstrated that GA may have a therapeutic effect on DM­associated AS. This study provides a theoretical basis for the treatment of diabetic AS.


Atherosclerosis/drug therapy , Diabetes Mellitus, Experimental/complications , Glycyrrhizic Acid/pharmacology , HMGB1 Protein/antagonists & inhibitors , Animals , Atherosclerosis/etiology , Diabetes Mellitus, Experimental/chemically induced , Glycyrrhizic Acid/therapeutic use , HMGB1 Protein/metabolism , Humans , Macrophage Activation/drug effects , Male , Rats , Rats, Sprague-Dawley , Streptozocin/administration & dosage , Streptozocin/toxicity
14.
Am J Respir Cell Mol Biol ; 65(6): 646-657, 2021 12.
Article En | MEDLINE | ID: mdl-34251297

Compromised endothelial-cell (EC) barrier function is a hallmark of inflammatory diseases. mTOR inhibitors, widely applied as clinical therapies, cause pneumonitis through mechanisms that are not yet fully understood. This study aimed to elucidate the EC mechanisms underlying the pathogenesis of pneumonitis caused by mTOR inhibition (mTORi). Mice with EC-specific deletion of mTOR complex components (Mtor, Rptor or Rictor) were administered LPS to induce pulmonary injury. Cultured ECs were treated with pharmacologic inhibitors, siRNA, or overexpression plasmids. EC barrier function was evaluated in vivo with Evans blue assay and in vitro by measurement of transendothelial electrical resistance and albumin flux. mTORi increased basal and TNFα-induced EC permeability, which was caused by myosin light chain (MLC) phosphorylation-dependent cell contraction. Inactivation of mTOR kinase activity by mTORi triggered PKCδ/p38/NF-κB signaling that significantly upregulated TNFα-induced MLCK (MLC kinase) expression, whereas Raptor promoted the phosphorylation of PKCα/MYPT1 independently of its interaction with mTOR, leading to suppression of MLCP (MLC phosphatase) activity. EC-specific deficiency in mTOR, Raptor or Rictor aggravated lung inflammation in LPS-treated mice. These findings reveal that mTORi induces PKC-dependent endothelial MLC phosphorylation, contraction, and hyperpermeability that promote pneumonitis.


Human Umbilical Vein Endothelial Cells/enzymology , MAP Kinase Signaling System/drug effects , MTOR Inhibitors/adverse effects , Pneumonia/enzymology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Humans , Lipopolysaccharides/toxicity , MTOR Inhibitors/pharmacology , Mice , Mice, Knockout , Myosin Light Chains/metabolism , Permeability , Phosphorylation/drug effects , Pneumonia/chemically induced , TOR Serine-Threonine Kinases/metabolism
15.
J Cardiovasc Pharmacol ; 78(3): 388-393, 2021 09 01.
Article En | MEDLINE | ID: mdl-34029271

ABSTRACT: Percutaneous coronary intervention has become the main revascularization strategy for coronary artery disease. Compared with early percutaneous coronary angioplasty and the extensive clinical application of bare metal stents, drug-eluting stents can significantly reduce the stenosis caused by the elastic retraction of plaque and neoatherosclerosis (NA), but there is still a high incidence of in-stent restenosis (ISR), which restricts the clinical efficacy of stent implantation. In-stent neoatherosclerosis (ISNA), defined as atherosclerotic lesions in the neointima, is one of the main causes of late stent failure. ISNA plays an important role in stent thrombosis and ISR. The rate of target lesion revascularization and in-stent thrombosis is high when NA arises. Therefore, it is of great clinical significance to explore the occurrence of NA and its development mechanism after stent implantation to prevent ISR and improve stent implantation efficacy and associated clinical prognosis. In this article, we systematically reviewed the existing clinical research on ISNA and the role of optical coherence tomography in its evaluation.


Coronary Artery Disease/therapy , Coronary Restenosis/etiology , Coronary Thrombosis/etiology , Coronary Vessels/pathology , Neointima , Percutaneous Coronary Intervention/instrumentation , Plaque, Atherosclerotic , Stents , Animals , Coronary Restenosis/diagnostic imaging , Coronary Restenosis/pathology , Coronary Thrombosis/diagnostic imaging , Coronary Thrombosis/pathology , Coronary Vessels/diagnostic imaging , Drug-Eluting Stents , Humans , Percutaneous Coronary Intervention/adverse effects , Predictive Value of Tests , Risk Factors , Tomography, Optical Coherence , Treatment Outcome
16.
Theranostics ; 11(11): 5605-5619, 2021.
Article En | MEDLINE | ID: mdl-33859766

Increased expression of vascular cell adhesion molecule (VCAM)-1 on the activated arterial endothelial cell (EC) surface critically contributes to atherosclerosis which may in part be regulated by epigenetic mechanisms. This study investigated whether and how the clinically available histone deacetylases 1 and 2 (HDAC1/2) inhibitor drug Romidepsin epigenetically modulates VCAM-1 expression to suppress atherosclerosis. Methods: VCAM-1 expression was analyzed in primary human aortic EC (HAEC) treated with Romidepsin or transfected with HDAC1/2-targeting siRNA. Methylation of GATA6 promoter region was examined with methylation-specific PCR assay. Enrichment of STAT3 to GATA6 promoter was detected with chromatin immunoprecipitation. Lys685Arg mutation was constructed to block STAT3 acetylation. The potential therapeutic effect of Romidepsin on atherosclerosis was evaluated in Apoe-/- mice fed with a high-fat diet. Results: Romidepsin significantly attenuated TNFα-induced VCAM-1 expression on HAEC surface and monocyte adhesion through simultaneous inhibition of HDAC1/2. This downregulation of VCAM-1 was attributable to reduced expression of transcription factor GATA6. Romidepsin enhanced STAT3 acetylation and its binding to DNA methyltransferase 1 (DNMT1), leading to hypermethylation of the GATA6 promoter CpG-rich region at +140/+255. Blocking STAT3 acetylation at Lys685 disrupted DNMT1-STAT3 interaction, decreased GATA6 promoter methylation, and reversed the suppressive effects of HDAC1/2 inhibition on GATA6 and VCAM-1 expression. Finally, intraperitoneal administration of Romidepsin reduced diet-induced atherosclerotic lesion development in Apoe-/- mice, accompanied by a reduction in GATA6/VCAM-1 expression in the aorta. Conclusions: HDAC1/2 contributes to VCAM-1 expression and atherosclerosis by suppressing STAT3 acetylation-dependent GATA6 promoter methylation. These findings may provide a rationale for HDAC1/2-targeting therapy in atherosclerotic heart disease.


Atherosclerosis/genetics , Endothelial Cells/metabolism , GATA6 Transcription Factor/genetics , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Promoter Regions, Genetic/genetics , Vascular Cell Adhesion Molecule-1/genetics , Animals , Aorta/metabolism , Apolipoproteins E/genetics , Cells, Cultured , CpG Islands/genetics , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Down-Regulation/genetics , Humans , Male , Methylation , Mice , Mice, Inbred C57BL , Monocytes/metabolism , STAT3 Transcription Factor/genetics , THP-1 Cells/metabolism
17.
Exp Ther Med ; 21(6): 560, 2021 Jun.
Article En | MEDLINE | ID: mdl-33850532

In-stent restenosis (ISR) remains an inevitable problem for some patients receiving drug-eluting stent (DES) implantation. Intimal hyperplasia is an important biological cause of ISR. It has been previously reported that adropin is a potentially protective factor in cardiovascular disease. Therefore, the present study investigated the function of adropin in inhibiting smooth muscle cell (SMC) phenotype modulation and proliferation, causing intimal hyperplasia. A total of 56 patients who visited the hospital consecutively (25 with ISR and 31 without ISR), who were followed up between April 2016 and March 2019, 1 year following DES, were analyzed to evaluate the association between in-stent neointimal volume and adropin serum levels. Rat aorta smooth muscle cells (RASMCs) were used to determine the effects of adropin on their phenotypic modulation and proliferation using western blot, MTT, PCR and immunofluorescence analyses. Adropin serum levels in the ISR group were significantly lower than those in the non-ISR group. Furthermore, linear regression analysis revealed that only adropin levels were negatively associated with neointimal volume in both groups. The overall adropin levels of the 56 patients and the percentages of neointimal volume revealed a strong negative association. In vitro, adropin suppressed angiotensin II (Ang II)-induced phenotypic modulation in RASMCs by restoring variations of osteopontin and α-smooth muscle actin. Furthermore, compared with the Ang II group, adropin markedly decreased the percentage of G2/M-phase cells. Finally, adropin negatively regulated the phenotypic modulation and proliferation of RASMCs via the AMP-activated protein kinase/acetyl-CoA carboxylase (AMPK/ACC) signaling pathway. In conclusion, an independent, negative association was revealed between adropin and intimal hyperplasia; specifically, adropin inhibited the phenotypic modulation and proliferation of RASMCs by activating the AMPK/ACC signaling pathway. Therefore, adropin may be used as a potential predictor and therapeutic target for intimal hyperplasia and ISR.

18.
Front Cardiovasc Med ; 8: 765419, 2021.
Article En | MEDLINE | ID: mdl-35004882

Calcified aortic valve disease (CAVD) was previously regarded as a passive process associated with valve degeneration and calcium deposition. However, recent studies have shown that the occurrence of CAVD is an active process involving complex changes such as endothelial injury, chronic inflammation, matrix remodeling, and neovascularization. CAVD is the ectopic accumulation of calcium nodules on the surface of the aortic valve, which leads to aortic valve thickening, functional stenosis, and ultimately hemodynamic disorders. CAVD has become an important cause of death from cardiovascular disease. The discovery of therapeutic targets to delay or block the progression of CAVD and the clinical application of transcatheter aortic valve implantation (TAVI) provide new ideas for the prevention and treatment of CAVD. This article summarizes the pathogenesis of CAVD and provides insight into the future directions of CAVD diagnosis and treatment.

19.
Gene ; 771: 145354, 2021 Mar 01.
Article En | MEDLINE | ID: mdl-33333215

BACKGROUND: Puerarin shows inhibitory effects on inflammation in chronic heart failure (CHF), but its efficacy in coronary heart disease (CHD) remained vague. METHODS: Rat CHD model was constructed, and serum parameters were determined using a blood liquid biochemical analyzer. Also, contents of creatine kinase (CK), creatine kinase MB isoenzyme (CK-MB), lactate dehydrogenase (LDH) and cardiac troponin (cTnT) were measured using colorimetry. Histological examination was conducted with Hematoxylin-Eosin (H&E) staining, and cardiac function was assessed by Echocardiography. Cell apoptosis was detected using Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay. Relative expressions were measured using quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot as needed. RESULTS: In CHD rats, the levels of TC, LDL and TG and the expressions of matrix metalloproteinase-9 (MMP-9), CD40 ligand (CD40L), tumor necrosis factor-α (TNF-α) and C-reactive protein (CRP) were increased while HDL level was decreased, accompanied with inflammatory cell infiltration and cardiac malfunction. Also, the contents of CK, CK-MB, LDH and cTnT, the percentage of apoptotic cells, the expressions of Bcl-2 associated X protein (Bax), cleaved Caspase-3, TNF-α, Interleukin-ß (IL-ß), IL-6 and Lipoprotein-associated Phospholipase A2 (Lp-PLA2) expressions and the levels of oxidized-(ox-)LDL and malondialdehyde (MDA) were upregulated, while the level of super oxidase dismutase (SOD) and the expressions of B cell lymphoma-2 (Bcl-2) and vascular endothelial growth factor (VEGF) were downregulated. However, Puerarin ameliorated the effects of CHD model construction, suppressed nuclear factor-(NF-)κB expression, and enhanced the expressions of Farnesoid X Receptor (FXR), phosphorylated-AKT (p-AKT) and phosphorylated-signal transducer and activator of transcription 3 (p-STAT3). CONCLUSION: Puerarin alleviated CHD in rats via inhibiting inflammation, providing possible method for CHD treatment.


Anti-Inflammatory Agents/administration & dosage , Coronary Disease/drug therapy , Gene Regulatory Networks/drug effects , Isoflavones/administration & dosage , Animals , Anti-Inflammatory Agents/pharmacology , Coronary Disease/genetics , Creatine Kinase/blood , Disease Models, Animal , Gene Expression Regulation/drug effects , Isoflavones/pharmacology , L-Lactate Dehydrogenase/blood , Male , Rats , Troponin C/blood
20.
Medicine (Baltimore) ; 99(52): e23346, 2020 Dec 24.
Article En | MEDLINE | ID: mdl-33350725

BACKGROUND: Ethylene diamine tetra-acetic acid (EDTA) is a chelating agent which attach to metals such as calcium and enables their elimination. In particular, some researchers suggest chelation with EDTA to treat cardiovascular disease with the hypothesis of moderating calcium to decrease atherosclerotic calcification of arteries. However, chelation with EDTA therapeutic effects in atherosclerotic cardiovascular disease is still unclear. Therefore, we propose to undertake a meta-analysis to assess the curative effects of EDTA chelation therapy in patients with atherosclerotic cardiovascular disease. METHODS: In the current study, we set to perform a systematic literature search using the electronic databases of 4 most commonly used English databases (EMBASE, MEDLINE, Cochrane Library, and ClinicalTrials.gov trials register), as well as 3 most commonly employed Chinese databases (China Nation Knowledge Infrastructure, Wan Fang, and VIP), from the date of database inception until September 30, 2020 to identify relevant randomized controlled studies of the evaluation on curative effects of EDTA chelation therapy in patients with atherosclerotic cardiovascular disease. In the study, 2 authors worked independently to screen search results, chose studies for inclusion, then they extracted pertinent data to evaluate and study quality based on Cochrane Risk of Bias Tool V.2.0. Additionally, we will address discrepancies by consultation with a third author. We also intend to use pooled risk ratio (RR) and pooled mean difference (MD) or pooled standardized mean difference (SMD) with 95% confidence intervals (CI) to approximate the relative strength of curative effects of EDTA chelation therapy in patients with atherosclerotic cardiovascular disease. RESULTS: The results of the current study will systematically assess curative effects of EDTA chelation therapy in patients with atherosclerotic cardiovascular disease. CONCLUSION: The study will infer the currently published evidence to evaluate curative effects of EDTA chelation therapy in patients with atherosclerotic cardiovascular disease, which might be beneficial to these patients. ETHICS AND DISSEMINATION: The present study is a systematic review, hence the pooled results are founded upon the published evidence. Therefore, ethical approval is not necessary for the study. OPEN SCIENCE FRAMEWORK REGISTRATION NUMBER: October 20, 2020.osf.io/tvmk8. (https://osf.io/tvmk8/).


Atherosclerosis/drug therapy , Calcium , Chelation Therapy/methods , Edetic Acid/therapeutic use , Meta-Analysis as Topic , Research Design , Systematic Reviews as Topic/methods , Humans
...