29 results on '"Tang, Chao-Ke"'
Search Results
2. HAND2-AS1: A novel therapeutic target for atherosclerosis.
- Author
-
Zeng GG, Li H, and Tang CK
- Subjects
- Cell Proliferation, Humans, Atherosclerosis drug therapy, MicroRNAs, RNA, Long Noncoding
- Published
- 2022
- Full Text
- View/download PDF
3. Long non-coding RNA PCA3 inhibits lipid accumulation and atherosclerosis through the miR-140-5p/RFX7/ABCA1 axis.
- Author
-
Zhao ZW, Zhang M, Liao LX, Zou J, Wang G, Wan XJ, Zhou L, Li H, Qin YS, Yu XH, and Tang CK
- Subjects
- ATP Binding Cassette Transporter 1 genetics, Atherosclerosis genetics, Atherosclerosis pathology, Humans, MicroRNAs genetics, RNA, Long Noncoding genetics, Regulatory Factor X Transcription Factors genetics, THP-1 Cells, ATP Binding Cassette Transporter 1 metabolism, Atherosclerosis metabolism, Lipid Metabolism, MicroRNAs metabolism, RNA, Long Noncoding metabolism, Regulatory Factor X Transcription Factors metabolism, Signal Transduction
- Abstract
Objective: The purpose of this study was to explore the role of long noncoding RNA (lncRNA) prostate cancer antigen 3 (PCA3) in atherosclerosis and the underlying mechanism., Methods: The Gene Expression Omnibus (GEO) datasets were used to divide differentially expressed lncRNAs, microRNAs (miRNAs), and mRNAs. The expression of PCA3, miR-140-5p, RFX7 and ABCA1 were determined by qPCR or Western blot in ox-LDL-treated macrophages. Macrophage lipid accumulation s was evaluated using the Oil Red O staining and high-performance liquid chromatography. Target relationships among PCA3, miR-140-5p, RFX7, and ABCA1 promoter area were validated via dual-luciferase reporter gene assay or chromatin immunoprecipitation assay. The apoE
-/- mouse model in vivo was designed to evaluate the effect of PCA3 on the reverse cholesterol transport (RCT) and atherosclerosis., Results: PCA3 was down-regulated in foam cells, whereas miR-140-5p was highly expressed. Overexpression of PCA3 promoted ABCA1-mediated cholesterol efflux and reduced lipid accumulation in macrophages. Besides, RFX7 bound to the ABCA1 promoter and increased ABCA1 expression. Targeted relationships and interactions on the expression between miR-140-5p and PCA3 or RFX7 were elucidated. PCA3 up-regulated ABCA1 expression by binding to miR-140-5p to up-regulate RFX7 and ABCA1 expression in macrophages. PCA3 promoted RCT and impeded the progression of atherosclerosis by sponging miR-140-5p in apoE-/- mice. Meanwhile, miR-140-5p also inhibit ABCA1 expression via downregulation of RFX7 to impede RCT and aggravate atherosclerosis., Conclusions: lncRNA PCA3 promotes ABCA1-mediated cholesterol efflux to inhibit atherosclerosis through sponging miR-140-5p and up-regulating RFX7., (Copyright © 2021 Elsevier B.V. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
4. The Long Noncoding RNA Metastasis-Associated Lung Adenocarcinoma Transcript-1 Regulates CCDC80 Expression by Targeting miR-141-3p/miR-200a-3p in Vascular Smooth Muscle Cells.
- Author
-
Gong D, Zhao ZW, Zhang Q, Yu XH, Wang G, Zou J, Zheng XL, Zhang DW, Yin WD, and Tang CK
- Subjects
- Binding Sites, Cells, Cultured, Extracellular Matrix Proteins genetics, Gene Expression Regulation, Humans, Lipoprotein Lipase genetics, Lipoprotein Lipase metabolism, MicroRNAs genetics, Promoter Regions, Genetic, RNA, Long Noncoding genetics, Extracellular Matrix Proteins metabolism, MicroRNAs metabolism, Muscle, Smooth, Vascular metabolism, Myocytes, Smooth Muscle metabolism, RNA, Long Noncoding metabolism
- Abstract
Objective: Our previous study showed that Coiled-Coil Domain Containing 80 (CCDC80) accelerates the development of atherosclerosis by decreasing lipoprotein lipase (LPL) expression and activity in apoE knockout mice. However, the regulatory mechanism for CCDC80 expression is unclear. This study was designed to evaluate whether noncoding RNAs involved the regulation of CCDC80 expression in vascular smooth muscle cells., Methods and Results: Bioinformatics prediction and luciferase reporter gene results showed that miR-141-3p/200a-3p bound to the 3'UTR of CCDC80. Furthermore, miR-141-3p/200a-3p mimics decreased the expression of CCDC80 but increased LPL expression. Opposite results were observed with miR-141-3p/200a-3p inhibitors. We also found that lncRNA metastasis-associated lung adenocarcinoma transcript-1 (MALAT1) interacted with the sequences of miR-141-3p/200a-3p and decreased their expression. RT-qPCR and western blotting results showed that MALAT1 overexpression increased CCDC80 expression and decreased LPL expression, while MALAT1 knockdown displayed an opposite phenotype. The effects of both MALAT1 overexpression and knockdown were blocked by miR-141-3p/200a-3p mimics or inhibitors., Conclusions: Thus, we demonstrated that lncRNA MALAT1 regulates CCDC80 and LPL expression through miR-141-3p/200a-3p.
- Published
- 2020
- Full Text
- View/download PDF
5. IL-8 negatively regulates ABCA1 expression and cholesterol efflux via upregulating miR-183 in THP-1 macrophage-derived foam cells.
- Author
-
Tang XE, Li H, Chen LY, Xia XD, Zhao ZW, Zheng XL, Zhao GJ, and Tang CK
- Subjects
- Atherosclerosis metabolism, Atherosclerosis pathology, Foam Cells pathology, Humans, THP-1 Cells, ATP Binding Cassette Transporter 1 biosynthesis, Cholesterol metabolism, Foam Cells metabolism, Gene Expression Regulation, Interleukin-8 metabolism, MicroRNAs metabolism
- Abstract
Objective: Previous studies suggest that IL-8 has an important role in the regulation of cholesterol efflux, but whether miRNAs are involved in this process is still unknown. The purpose of this study is to explore whether IL-8 promotes cholesterol accumulation by enhancing miR-183 expression in macrophages and its underlying mechanism., Methods and Results: Treatment of THP-1 macrophage-derived foam cells with IL-8 decreased ABCA1 expression and cholesterol efflux. Using bioinformatics analyses and dual-luciferase reporter assays, we found that miR-183 was highly conserved during evolution and directly inhibited ABCA1 protein and mRNA expression by targeting ABCA1 3'UTR. MiR-183 directly regulated endogenous ABCA1 expression levels. Furthermore, IL-8 enhanced the expression of miR-183 and decrease ABCA1 expression. Cholesterol transport assays confirmed that IL-8 dramatically inhibited apolipoprotein AI-mediated ABCA1-dependent cholesterol efflux by increasing miR-183 expression. In contrast, treatment with anti-IL-8 antibody reversed these effects., Conclusion: IL-8 enhances the expression of miR-183, which then inhibits ABCA1 expression and cholesterol efflux. Our studies suggest that the IL-8-miR-183-ABCA1 axis may play an intermediary role in the development of atherosclerosis., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
6. Krüppel-like factor 14 inhibits atherosclerosis via mir-27a-mediated down-regulation of lipoprotein lipase expression in vivo.
- Author
-
Xie W, Li L, Gong D, Zhang M, Lv YC, Guo DM, Zhao ZW, Zheng XL, Zhang DW, Dai XY, Yin WD, and Tang CK
- Subjects
- Animals, Atherosclerosis pathology, Down-Regulation, Gene Expression Regulation, Enzymologic, Gynostemma, Homeostasis, Lipid Metabolism, Lipids chemistry, Lipoproteins, LDL metabolism, Macrophages metabolism, Male, Mice, Mice, Knockout, ApoE, Plant Extracts pharmacology, RAW 264.7 Cells, Transfection, Atherosclerosis metabolism, Kruppel-Like Transcription Factors metabolism, Lipoprotein Lipase metabolism, MicroRNAs metabolism
- Abstract
Background and Aims: Krüppel-like factor 14 (KLF14) is known to play a role in atherosclerosis, but the underlying mechanisms are still largely unknown. The aim of our study was to explore the effects of KLF14 on lipid metabolism and inflammatory response, providing a potential target for lowering the risk of atherosclerosis-causing disease., Methods and Results: mRNA and protein levels of KLF14 were significantly decreased in oxidized low-density lipoprotein (oxLDL)-treated macrophages and in the atherosclerotic lesion area. Chromatin immunoprecipitation (ChIP) and luciferase reporter gene assays were used to confirm that KLF14 positively regulated miR-27a expression by binding to its promoter. We also found that KLF14 could restored appropriate cellular lipid homeostasis and inflammatory responses via negatively regulating lipoprotein lipase (LPL) expression in THP1-derived macrophages through miR-27a. In addition, gypenosides (GP), a KLF14 activator, delayed the development of atherosclerosis in apolipoprotein E deficient (apoE
-/- ) mice., Conclusions: KLF14 plays an antiatherogenic role via the miR-27a-dependent down-regulation of LPL and subsequent inhibition of proinflammatory cytokine secretion and lipid accumulation., (Copyright © 2019 Elsevier B.V. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
7. Nobiletin reduces LPL-mediated lipid accumulation and pro-inflammatory cytokine secretion through upregulation of miR-590 expression.
- Author
-
He PP, Shen QQ, Wen M, Zou JQ, Wang Y, Yang JX, Hu LZ, Zheng XL, Chen YS, Su H, Liu J, Ouyang XP, and Tang CK
- Subjects
- Atherosclerosis drug therapy, Atherosclerosis genetics, Atherosclerosis metabolism, Cardiotonic Agents pharmacology, Cytokines metabolism, Down-Regulation drug effects, Drug Development, Humans, Inflammation Mediators metabolism, Lipoprotein Lipase antagonists & inhibitors, Macrophages drug effects, Macrophages metabolism, THP-1 Cells, Up-Regulation drug effects, Flavones pharmacology, Lipid Metabolism drug effects, Lipoprotein Lipase metabolism, MicroRNAs genetics, MicroRNAs metabolism
- Abstract
Nobiletin has protective effects on cardiovascular diseases, but the mechanism is not clear. In this study, we examined whether nobiletin affects the expression of miR-590/LPL and its relative effects on lipid accumulation and pro-inflammatory cytokine secretion in human THP-1 macrophages. RT-qPCR analysis showed that nobiletin increased the expression of miR-590. Western blot analysis showed that nobiletin-suppressed LPL expression was enhanced by miR-590 mimic and abrogated by miR-590 inhibitor. Oil Red O staining and high-performance liquid chromatography assays showed that nobiletin attenuated lipid accumulation in macrophages. Treatment with nobiletin and miR-590 mimic decreased cellular lipid accumulation, whereas treatment with miR-590 inhibitor increased cellular lipid accumulation. ELISA illustrated that nobiletin alleviated pro-inflammatory cytokine secretion in macrophages as measured by, which was reduced by miR-590 mimic and increased by miR-590 inhibitor. In conclusion, nobiletin may alleviate lipid accumulation and secretion of pro-inflammatory cytokines by enhancing the inhibitory effect of miR-590 on LPL expression, suggesting a promising strategy for potential drug development for atherosclerosis., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
8. MicroRNA-377 Inhibits Atherosclerosis by Regulating Triglyceride Metabolism Through the DNA Methyltransferase 1 in Apolipoprotein E-Knockout Mice.
- Author
-
Chen LY, Xia XD, Zhao ZW, Gong D, Ma XF, Yu XH, Zhang Q, Wang SQ, Dai XY, Zheng XL, Zhang DW, Yin WD, and Tang CK
- Subjects
- Animals, Aorta pathology, Atherosclerosis genetics, Atherosclerosis pathology, DNA (Cytosine-5-)-Methyltransferase 1 genetics, Human Umbilical Vein Endothelial Cells metabolism, Human Umbilical Vein Endothelial Cells pathology, Humans, Lipoprotein Lipase genetics, Lipoprotein Lipase metabolism, Mice, Mice, Knockout, ApoE, MicroRNAs genetics, Plaque, Atherosclerotic genetics, Plaque, Atherosclerotic pathology, Receptors, Lipoprotein biosynthesis, Receptors, Lipoprotein genetics, Aorta metabolism, Atherosclerosis metabolism, DNA (Cytosine-5-)-Methyltransferase 1 metabolism, MicroRNAs metabolism, Plaque, Atherosclerotic metabolism, Triglycerides metabolism
- Abstract
Background: Lipoprotein lipase (LPL) plays an important role in triglyceride metabolism. It is translocated across endothelial cells to reach the luminal surface of capillaries by glycosylphosphatidylinositol-anchored high-density lipoprotein binding protein 1 (GPIHBP1), where it hydrolyzes triglycerides in lipoproteins. MicroRNA 377 (miR-377) is highly associated with lipid levels. However, how miR-377 regulates triglyceride metabolism and whether it is involved in the development of atherosclerosis remain largely unexplored. Methods and Results: The clinical examination displayed that miR-377 expression was markedly lower in plasma from patients with hypertriglyceridemia compared with non-hypertriglyceridemic subjects. Bioinformatics analyses and a luciferase reporter assay showed that DNA methyltransferase 1 (DNMT1) was a target gene of miR-377. Moreover, miR-377 increased LPL binding to GPIHBP1 by directly targeting DNMT1 in human umbilical vein endothelial cells (HUVECs) and apolipoprotein E (ApoE)-knockout (KO) mice aorta endothelial cells (MAECs). In vivo, hematoxylin-eosin (H&E), Oil Red O and Masson's trichrome staining showed that ApoE-KO mice treated with miR-377 developed less atherosclerotic plaques, accompanied by reduced plasma triglyceride levels., Conclusions: It is concluded that miR-377 upregulates GPIHBP1 expression, increases the LPL binding to GPIHBP1, and reduces plasma triglyceride levels, likely through targeting DNMT1, inhibiting atherosclerosis in ApoE-KO mice.
- Published
- 2018
- Full Text
- View/download PDF
9. Visceral adipose tissue-derived serine protease inhibitor accelerates cholesterol efflux by up-regulating ABCA1 expression via the NF-κB/miR-33a pathway in THP-1 macropahge-derived foam cells.
- Author
-
Gao JH, Zeng MY, Yu XH, Zeng GF, He LH, Zheng XL, Zhang DW, Ouyang XP, and Tang CK
- Subjects
- ATP Binding Cassette Transporter 1 genetics, Base Sequence, Cell Line, Down-Regulation, Foam Cells drug effects, Humans, Lipid Metabolism, MicroRNAs genetics, Signal Transduction, ATP Binding Cassette Transporter 1 metabolism, Cholesterol metabolism, Foam Cells metabolism, Intra-Abdominal Fat metabolism, Macrophages cytology, MicroRNAs metabolism, NF-kappa B metabolism, Serpins metabolism, Up-Regulation
- Abstract
Atherosclerosis is a dyslipidemia disease characterized by foam cell formation driven by the accumulation of lipids. Visceral adipose tissue-derived serine protease inhibitor (vaspin) is known to suppress the development of atherosclerosis via its anti-inflammatory properties, but it is not yet known whether vaspin affects cholesterol efflux in THP-1 macrophage-derived foam cells. Here, we investigated the effects of vaspin on ABCA1 expression and cholesterol efflux, and further explored the underlying mechanism. We found that vaspin decreased miR-33a levels, which in turn increased ABCA1 expression and cholesteorl efflux. We also found that inhibition of NF-κB reduced miR-33a expression and vaspin suppressed LPS-mediated NF-κB phosphorylation. Our findings suggest that vaspin is not only a regular of inflammasion but also a promoter of cholesterol efflux., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
10. Interferon-stimulated gene 15 promotes cholesterol efflux by activating autophagy via the miR-17-5p/Beclin-1 pathway in THP-1 macrophage-derived foam cells.
- Author
-
Huang C, Yu XH, Zheng XL, Ou X, and Tang CK
- Subjects
- Base Sequence, Biological Transport, Foam Cells metabolism, HEK293 Cells, Humans, Intracellular Space metabolism, Autophagy, Beclin-1 genetics, Cholesterol metabolism, Cytokines metabolism, Foam Cells cytology, MicroRNAs genetics, Ubiquitins metabolism
- Abstract
Macrophage autophagy contributes to the hydrolysis of cholesteryl ester into free cholesterol mainly for ATP-binding cassette transporter A1 (ABCA1)-dependent efflux. Interferon-stimulated gene 15 (ISG15) has been shown to regulate autophagy in multiple types of cells. The present study aimed to examine the effects of ISG15 on autophagy and cholesterol efflux in THP-1 macrophage-derived foam cells and to explore the underlying molecular mechanisms. Our results showed that overexpression of ISG15 promoted autophagy and cholesterol efflux and inhibited lipid accumulation without impact on ABCA1 expression. Inhibition of autophagy by 3-methyladenine (3-MA) abrogated the enhancing effects of ISG15 on cholesterol efflux. Both bioinformatics analysis and dual luciferase reporter assay identified Beclin-1 as a direct target of miR-17-5p. Moreover, ISG15 overexpression markedly decreased miR-17-5p levels and upregulated Beclin-1 expression. ISG15-induced enhancement of autophagy and cholesterol efflux was reversed by pretreatment with either miR-17-5p mimic or Beclin-1 siRNA. In conclusion, these findings suggest that ISG15 reduces miR-17-5p levels and thereby promotes Beclin-1-mediated autophagy, resulting in increased cholesterol efflux from THP-1 macrophage-derived foam cells., (Copyright © 2018 Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
11. MicroRNA-134 Promotes the Development of Atherosclerosis Via the ANGPTL4/LPL Pathway in Apolipoprotein E Knockout Mice.
- Author
-
Ye Q, Tian GP, Cheng HP, Zhang X, Ou X, Yu XH, Tan RQ, Yang FY, Gong D, Huang C, Pan YJ, Zhang J, Chen LY, Zhao ZW, Xie W, Li L, Zhang M, Xia XD, Zheng XL, and Tang CK
- Subjects
- Animals, Atherosclerosis metabolism, Cholesterol metabolism, Cytokines metabolism, Foam Cells metabolism, Inflammation, Lipids chemistry, Lipoprotein Lipase metabolism, Macrophages metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, ApoE, Angiopoietin-Like Protein 4 blood, Angiopoietin-Like Protein 4 genetics, Atherosclerosis genetics, MicroRNAs blood, MicroRNAs genetics
- Abstract
Aims: Atherosclerosis is the most common cause of cardiovascular disease, such as myocardial infarction and stroke. Previous study revealed that microRNA (miR)-134 promotes lipid accumulation and proinflammatory cytokine secretion through angiopoietin-like 4 (ANGPTL4)/lipid lipoprotein (LPL) signaling in THP-1 macrophages., Methods: ApoE KO male mice on a C57BL/6 background were fed a high-fat/high-cholesterol Western diet, from 8 to 16 weeks of age. Mice were divided into four groups, and received a tail vein injection of miR-134 agomir, miR-134 antagomir, or one of the corresponding controls, respectively, once every 2 weeks after starting the Western diet. After 8 weeks we measured aortic atherosclerosis, LPL Activity, mRNA and protein levels of ANGPTL4 and LPL, LPL/ low-density lipoprotein receptor related protein 1 Complex Formation, proinflammatory cytokine secretion and lipid levels., Results: Despite this finding, the influence of miR-134 on atherosclerosis in vivo remains to be determined. Using the well-characterized mouse atherosclerosis model of apolipoprotein E knockout, we found that systemic delivery of miR-134 agomir markedly enhanced the atherosclerotic lesion size, together with a significant increase in proinflammatory cytokine secretion and peritoneal macrophages lipid contents. Moreover, overexpression of miR-134 decreased ANGPTL4 expression but increased LPL expression and activity in both aortic tissues and peritoneal macrophages, which was accompanied by increased formation of LPL/low-density lipoprotein receptor-related protein 1 complexes in peritoneal macrophages. However, an opposite effect was observed in response to miR-134 antagomir., Conclusions: These findings suggest that miR-134 accelerates atherogenesis by promoting lipid accumulation and proinflammatory cytokine secretion via the ANGPTL4/LPL pathway. Therefore, targeting miR-134 may offer a promising strategy for the prevention and treatment of atherosclerotic cardiovascular disease.
- Published
- 2018
- Full Text
- View/download PDF
12. MicroRNA-182 Promotes Lipoprotein Lipase Expression and Atherogenesisby Targeting Histone Deacetylase 9 in Apolipoprotein E-Knockout Mice.
- Author
-
Cheng HP, Gong D, Zhao ZW, He PP, Yu XH, Ye Q, Huang C, Zhang X, Chen LY, Xie W, Zhang M, Li L, Xia XD, Ouyang XP, Tan YL, Wang ZB, Tian GP, Zheng XL, Yin WD, and Tang CK
- Subjects
- Animals, Computational Biology, Cytokines drug effects, HEK293 Cells, Histone Deacetylases, Humans, Inflammation metabolism, Lipid Metabolism drug effects, Macrophages, Mice, Mice, Knockout, ApoE, THP-1 Cells, Atherosclerosis chemically induced, Lipoprotein Lipase drug effects, MicroRNAs pharmacology, Repressor Proteins antagonists & inhibitors
- Abstract
Background: Lipoprotein lipase (LPL) expressed in macrophages plays an important role in promoting the development of atherosclerosis or atherogenesis. MicroRNA-182 (miR-182) is involved in the regulation of lipid metabolism and inflammation. However, it remains unclear how miR-182 regulates LPL and atherogenesis.Methods and Results:Using bioinformatics analyses and a dual-luciferase reporter assay, we identified histone deacetylase 9 (HDAC9) as a target gene of miR-182. Moreover, miR-182 upregulated LPL expression by directly targetingHDAC9in THP-1 macrophages. Hematoxylin-eosin (H&E), Oil Red O and Masson's trichrome staining showed that apolipoprotein E (ApoE)-knockout (KO) mice treated with miR-182 exhibited more severe atherosclerotic plaques. Treatment with miR-182 increased CD68 and LPL expression in atherosclerotic lesions in ApoE-KO mice, as indicated by double immunofluorescence staining in the aortic sinus. Increased miR-182-induced increases in LPL expression in ApoE-KO mice was confirmed by real-time quantitative polymerase chain reaction and western blotting analyses. Treatment with miR-182 also increased plasma concentrations of proinflammatory cytokines and lipids in ApoE-KO mice., Conclusions: The results of the present study suggest that miR-182 upregulates LPL expression, promotes lipid accumulation in atherosclerotic lesions, and increases proinflammatory cytokine secretion, likely through targetingHDAC9, leading to an acceleration of atherogenesis in ApoE-KO mice.
- Published
- 2017
- Full Text
- View/download PDF
13. MicroRNA-186 promotes macrophage lipid accumulation and secretion of pro-inflammatory cytokines by targeting cystathionine γ-lyase in THP-1 macrophages.
- Author
-
Yao Y, Zhang X, Chen HP, Li L, Xie W, Lan G, Zhao ZW, Zheng XL, Wang ZB, and Tang CK
- Subjects
- 3' Untranslated Regions, Alleles, HEK293 Cells, Humans, Hydrogen Sulfide chemistry, Inflammation, Interleukin-6 metabolism, Leukocytes, Mononuclear cytology, Lipoprotein Lipase metabolism, Macrophages metabolism, MicroRNAs genetics, Myocytes, Cardiac metabolism, Signal Transduction, THP-1 Cells, Cystathionine gamma-Lyase genetics, Cystathionine gamma-Lyase metabolism, Cytokines metabolism, Lipids chemistry, MicroRNAs physiology
- Abstract
Background and Aims: Several studies suggest that cardiomyocyte-enriched miR-186 is involved in cardiac injury and myocardial infarction, and also plays an important role in atherosclerotic diseases, but the underlying mechanism is unknown. Cystathionine-γ-lyase (CSE) is the predominant enzyme to produce H2S in the cardiovascular system. Here, miR-186 was identified to bind to the 3'UTR of CSE. In this study, we aimed at exploring whether miR-186 affects lipid accumulation and secretion of pro-inflammatory cytokines by targeting CSE and its underlying mechanism in human THP-1 macrophages and peripheral blood monocyte-derived macrophages (PBMDM). PBMDM just as a control group for the comparison with the THP-1 macrophages., Methods: MiR-186 target genes, CSE 3'UTR sequence and free energy were predicted and analyzed by bioinformatics analyses and dual-luciferase reporter assays. The expression of CSE mRNA and protein were measured by real-time quantitative PCR and western blot analyses. The lipid accumulation in THP-1 macrophages was detected by high performance liquid chromatography (HPLC). The effects of miR-186 on secretion of IL-6, IL-1β and TNF-α were examined by ELISA. Endogenous H2S was detected by spectrophotometry. Using small interfering RNA (siRNA) approach to decrease the expression of CSE protein and mRNA., Results: We found that miR-186 directly inhibited CSE protein and mRNA expression through targeting CSE 3'UTR by bioinformatics analyses and dual-luciferase reporter assays. HPLC assays showed that miR-186 increased the lipid accumulation in human THP-1 macrophages. We also showed that miR-186 enhanced secretion of pro-inflammatory cytokines in human THP-1 macrophages. Using siRNA approach, we found that CSE siRNA could inhibit the miR-186 inhibitor-induced decrease in the expression of LPL protein and mRNA in human THP-1 macrophages, which was accompanied a decrease in the level of H2S., Conclusions: MicroRNA-186 promotes macrophage lipid accumulation and pro-inflammatory cytokine secretion by targeting cystathionine γ-lyase in THP-1 macrophages., (Copyright © 2016 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
14. MicroRNA-27 Prevents Atherosclerosis by Suppressing Lipoprotein Lipase-Induced Lipid Accumulation and Inflammatory Response in Apolipoprotein E Knockout Mice.
- Author
-
Xie W, Li L, Zhang M, Cheng HP, Gong D, Lv YC, Yao F, He PP, Ouyang XP, Lan G, Liu D, Zhao ZW, Tan YL, Zheng XL, Yin WD, and Tang CK
- Subjects
- Animals, Apolipoproteins E genetics, Atherosclerosis blood, Atherosclerosis genetics, Cell Line, Chemokine CCL2 blood, Chromatography, High Pressure Liquid, Enzyme-Linked Immunosorbent Assay, Humans, Inflammation blood, Inflammation genetics, Interleukin-1beta blood, Interleukin-6 blood, Lipid Metabolism genetics, Lipid Metabolism physiology, Lipoprotein Lipase genetics, Macrophages metabolism, Male, Mice, Mice, Knockout, MicroRNAs genetics, RAW 264.7 Cells, Real-Time Polymerase Chain Reaction, Receptors, Scavenger metabolism, Tumor Necrosis Factor-alpha blood, Apolipoproteins E deficiency, Atherosclerosis metabolism, Atherosclerosis prevention & control, Inflammation metabolism, Lipoprotein Lipase pharmacokinetics, MicroRNAs metabolism
- Abstract
Atherosclerotic lesions are lipometabolic disorder characterized by chronic progressive inflammation in arterial walls. Previous studies have shown that macrophage-derived lipoprotein lipase (LPL) might be a key factor that promotes atherosclerosis by accelerating lipid accumulation and proinflammatory cytokine secretion. Increasing evidence indicates that microRNA-27 (miR-27) has beneficial effects on lipid metabolism and inflammatory response. However, it has not been fully understood whether miR-27 affects the expression of LPL and subsequent development of atherosclerosis in apolipoprotein E knockout (apoE KO) mice. To address these questions and its potential mechanisms, oxidized low-density lipoprotein (ox-LDL)-treated THP-1 macrophages were transfected with the miR-27 mimics/inhibitors and apoE KO mice fed high-fat diet were given a tail vein injection with miR-27 agomir/antagomir, followed by exploring the potential roles of miR-27. MiR-27 agomir significantly down-regulated LPL expression in aorta and peritoneal macrophages by western blot and real-time PCR analyses. We performed LPL activity assay in the culture media and found that miR-27 reduced LPL activity. ELISA showed that miR-27 reduced inflammatory response as analyzed in vitro and in vivo experiments. Our results showed that miR-27 had an inhibitory effect on the levels of lipid both in plasma and in peritoneal macrophages of apoE KO mice as examined by HPLC. Consistently, miR-27 suppressed the expression of scavenger receptors associated with lipid uptake in ox-LDL-treated THP-1 macrophages. In addition, transfection with LPL siRNA inhibited the miR-27 inhibitor-induced lipid accumulation and proinflammatory cytokines secretion in ox-LDL-treated THP-1 macrophages. Finally, systemic treatment revealed that miR-27 decreased aortic plaque size and lipid content in apoE KO mice. The present results provide evidence that a novel antiatherogenic role of miR-27 was closely related to reducing lipid accumulation and inflammatory response via downregulation of LPL gene expression, suggesting a potential strategy to the diagnosis and treatment of atherosclerosis.
- Published
- 2016
- Full Text
- View/download PDF
15. MicroRNA-134 actives lipoprotein lipase-mediated lipid accumulation and inflammatory response by targeting angiopoietin-like 4 in THP-1 macrophages.
- Author
-
Lan G, Xie W, Li L, Zhang M, Liu D, Tan YL, Cheng HP, Gong D, Huang C, Zheng XL, Yin WD, and Tang CK
- Subjects
- Angiopoietin-Like Protein 4, Cell Line, Enzyme Activation, Humans, Macrophages enzymology, Signal Transduction immunology, Angiopoietins immunology, Inflammation immunology, Lipid Metabolism immunology, Lipoprotein Lipase immunology, Macrophages immunology, MicroRNAs immunology
- Abstract
Angiopoietin-like 4 (Angptl4), a secreted protein, is an important regulator to irreversibly inhibit lipoprotein lipase (LPL) activity. Macrophage LPL contributes to foam cell formation via a so-called"molecular bridge" between lipoproteins and receptors on cell surface. It has been reported that macrophage ANGPTL4 suppresses LPL activity, foam cell formation and inflammatory gene expression to reduce atherosclerosis development. Recently, some studies demonstrated that microRNA-134 is upregulated in atherosclerotic macrophages. Here we demonstrate that miR-134 directly binds to 3'UTR of ANGPTL4 mRNA to suppression the expression of ANGPTL4. To investigate the potential roles of macrophage miR-134, THP-1 macrophages were transfected with miR-134 mimics or inhibitors. Our results showed that LPL activity and protein were dramatically increased. We also found that miR-134 activated LPL-mediated lipid accumulation. Collectively, our findings indicate that miR-134 may regulate lipid accumulation and proinfiammatory cytokine secretion in macrophages by targeting the ANGPTL4 gene. Our results have also suggested a promising and potential therapeutic target for atherosclerosis., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
16. MiR-486 regulates cholesterol efflux by targeting HAT1.
- Author
-
Liu D, Zhang M, Xie W, Lan G, Cheng HP, Gong D, Huang C, Lv YC, Yao F, Tan YL, Li L, Zheng XL, and Tang CK
- Subjects
- Cell Line, Down-Regulation physiology, Humans, ATP Binding Cassette Transporter 1 metabolism, Cholesterol metabolism, Histone Acetyltransferases metabolism, Macrophages metabolism, MicroRNAs metabolism
- Abstract
Rationale: Excessive cholesterol accumulation in macrophages is a major factor of foam cell formation and development of atherosclerosis. Previous studies suggested that miR-486 plays an important role in cardiovascular diseases, but the underlying mechanism is still unknown., Objective: The purpose of this study is to determine whether miR-486 regulates ATP-binding cassette transporter A1 (ABCA1) mediated cholesterol efflux, and also explore the underlying mechanism., Methods and Results: Based on bioinformatics analysis and luciferase reporter assay, we transfected miR-486 mimic and miR-486 inhibitor into THP-1 macrophage-derived foam cells, and found that miR-486 directly bound to histone acetyltransferase-1 (HAT1) 3'UTR, and downregulated its mRNA and protein expression. In addition, our studies through transfection with wildtype HAT1 or shHAT1 (short hairpin HAT1) revealed that HAT1 could promote the expression of ABCA1 at both mRNA and protein levels. At the same time, the acetylation levels of the lysines 5 and 12 of histone H4 were upregulated after overexpression with HAT1. Meanwhile, the results of liquid scintillation counter and high performance liquid chromatography (HPLC) showed that miR-486 promoted cholesterol accumulation in THP-1 macrophages., Conclusion: These data indicated that miR-486 aggravate the cholesterol accumulation in THP-1 cells by targeting HAT1., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
17. Cystathionine γ-lyase(CSE)/hydrogen sulfide system is regulated by miR-216a and influences cholesterol efflux in macrophages via the PI3K/AKT/ABCA1 pathway.
- Author
-
Gong D, Cheng HP, Xie W, Zhang M, Liu D, Lan G, Huang C, Zhao ZW, Chen LY, Yao F, Tan YL, Li L, Xia XD, Zheng XL, Wang ZB, and Tang CK
- Subjects
- Cell Line, Humans, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction physiology, ATP Binding Cassette Transporter 1 metabolism, Cholesterol metabolism, Cystathionine gamma-Lyase metabolism, Hydrogen Sulfide metabolism, Macrophages metabolism, MicroRNAs metabolism
- Abstract
This study was designed to evaluate whether CSE/H2S system, which is regulated by miR-216a, regulated ABCA1-mediated cholesterol efflux and cholesterol contents in THP-1 macrophages-derived foam cells. Our qPCR and western blotting results showed that CSE/H2S significantly up-regulated the expression of ATP-binding cassette transporter A1 (ABCA1) mRNA and protein via PI3K/AKT pathway in foam cells derived from human THP-1 macrophages. The miR-216a directly targeted 3' untranslated region of CSE. It significantly reduced CSE and ABCA1 expression, and also decreased the phosphorylation of PI3K and AKT. Additionally, cholesterol efflux decreased, and cholesterol levels increased in THP-1 macrophage-derived foam cells in response to treatment with miR-216a. Our study demonstrates that CSE/H2S system is regulated by miR-216a, and regulates ABCA1-mediated cholesterol efflux and cholesterol levels through the PI3K/AKT pathway., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
18. MicroRNA-590 Inhibits Lipoprotein Lipase Expression and Prevents Atherosclerosis in apoE Knockout Mice.
- Author
-
He PP, OuYang XP, Li Y, Lv YC, Wang ZB, Yao F, Xie W, Tan YL, Li L, Zhang M, Lan G, Gong D, Cheng HP, Zhong HJ, Liu D, Huang C, Li ZX, Zheng XL, Yin WD, and Tang CK
- Subjects
- Animals, Aorta enzymology, Aorta pathology, CD36 Antigens metabolism, Cytokines blood, Enzyme Repression, Lipid Metabolism, Lipoprotein Lipase metabolism, Macrophages, Peritoneal enzymology, Male, Mice, Knockout, MicroRNAs metabolism, RNA Interference, Apolipoproteins E genetics, Atherosclerosis enzymology, Lipoprotein Lipase genetics, MicroRNAs genetics
- Abstract
Recent studies have suggested that miR-590 may play critical roles in cardiovascular disease. This study was designed to determine the effects of miR-590 on lipoprotein lipase (LPL) expression and development of atherosclerosis in apolipoprotein E knockout (apoE-/-) mice and explore the potential mechanisms. En face analysis of the whole aorta revealed that miR-590 significantly decreased aortic atherosclerotic plaque size and lipid content in apoE-/- mice. Double immunofluorescence staining in cross-sections of the proximal aorta showed that miR-590 agomir reduced CD68 and LPL expression in macrophages in atherosclerotic lesions. MiR-590 agomir down-regulated LPL mRNA and protein expression as analyzed by RT-qPCR and western blotting analyses, respectively. Consistently, miR-590 decreased the expression of CD36 and scavenger receptor A1 (SRA1) mRNA and protein. High-performance liquid chromatography (HPLC)analysis confirmed that treatment with miR-590 agomir reduced lipid levels either in plasma orinabdominal cavity macrophages of apoE-/- mice. ELISA analysis showed that miR-590 agomir decreased plasma levels of pro-inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α), monocyte chemotactic protein-1 (MCP-1), interleukin-1β (IL-1β)and interleukin-6 (IL-6). In contrast, treatment with miR-590 antagomir prevented or reversed these effects. Taken together, these results reveal a novel mechanism of miR-590 effects, and may provide new insights into the development of strategies for attenuating lipid accumulation and pro-inflammatory cytokine secretion.
- Published
- 2015
- Full Text
- View/download PDF
19. Diosgenin inhibits atherosclerosis via suppressing the MiR-19b-induced downregulation of ATP-binding cassette transporter A1.
- Author
-
Lv YC, Yang J, Yao F, Xie W, Tang YY, Ouyang XP, He PP, Tan YL, Li L, Zhang M, Liu D, Cayabyab FS, Zheng XL, and Tang CK
- Subjects
- ATP Binding Cassette Transporter 1 genetics, ATP Binding Cassette Transporter 1 metabolism, Animals, Aorta drug effects, Aorta metabolism, Aorta pathology, Aortic Diseases genetics, Aortic Diseases metabolism, Aortic Diseases pathology, Apolipoproteins E deficiency, Apolipoproteins E genetics, Atherosclerosis genetics, Atherosclerosis metabolism, Atherosclerosis pathology, Cell Line, Disease Models, Animal, Dose-Response Relationship, Drug, Foam Cells metabolism, Foam Cells pathology, Gene Expression Regulation, Humans, Macrophages, Peritoneal metabolism, Macrophages, Peritoneal pathology, Male, Mice, Inbred C57BL, Mice, Knockout, MicroRNAs genetics, Transfection, Aortic Diseases prevention & control, Atherosclerosis prevention & control, Cholesterol metabolism, Diosgenin pharmacology, Foam Cells drug effects, Hypolipidemic Agents pharmacology, Macrophages, Peritoneal drug effects, MicroRNAs metabolism
- Abstract
Rationale: Diosgenin (Dgn), a structural analogue of cholesterol, has been reported to have the hypolipidemic and antiatherogenic properties, but the underlying mechanisms are not fully understood. Given the key roles of macrophages in cholesterol metabolism and atherogenesis, it is critical to investigate macrophage cholesterol efflux and development of atherosclerotic lesion after Dgn treatment., Objective: This study was designed to evaluate the potential effects of Dgn on macrophage cholesterol metabolism and the development of aortic atherosclerosis, and to explore its underlying mechanisms., Methods and Results: Dgn significantly up-regulated the expression of ATP-binding cassette transporter A1 (ABCA1) protein, but didn't affect liver X receptor α levels in foam cells derived from human THP-1 macrophages and mouse peritoneal macrophages (MPMs) as determined by western blotting. The miR-19b levels were markedly down-regulated in Dgn-treated THP-1 macrophages/MPM-derived foam cells. Cholesterol transport assays revealed that treatment with Dgn alone or together with miR-19b inhibitor notably enhanced ABCA1-dependent cholesterol efflux, resulting in the reduced levels of total cholesterol, free cholesterol and cholesterol ester as determined by high-performance liquid chromatography. The fecal 3H-sterol originating from cholesterol-laden MPMs was increased in apolipoprotein E knockout mice treated with Dgn or both Dgn and antagomiR-19b. Treatment with Dgn alone or together with antagomiR-19b elevated plasma high-density lipoprotein levels, but reduced plasma low-density lipoprotein levels. Accordingly, aortic lipid deposition and plaque area were reduced, and collagen content and ABCA1 expression were increased in mice treated with Dgn alone or together with antagomiR-19b. However, miR-19b overexpression abrogated the lipid-lowering and atheroprotective effects induced by Dgn., Conclusion: The present study demonstrates that Dgn enhances ABCA1-dependent cholesterol efflux and inhibits aortic atherosclerosis progression by suppressing macrophage miR-19b expression., (Copyright © 2015 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
20. MicroRNA-590 attenuates lipid accumulation and pro-inflammatory cytokine secretion by targeting lipoprotein lipase gene in human THP-1 macrophages.
- Author
-
He PP, Ouyang XP, Tang YY, Liao L, Wang ZB, Lv YC, Tian GP, Zhao GJ, Huang L, Yao F, Xie W, Tang YL, Chen WJ, Zhang M, Li Y, Wu JF, Peng J, Liu XY, Zheng XL, Yin WD, and Tang CK
- Subjects
- 3' Untranslated Regions genetics, Base Sequence, Blotting, Western, Cell Line, Tumor, Gene Expression, HEK293 Cells, Humans, Inflammation Mediators metabolism, Lipoprotein Lipase metabolism, Lipoproteins, LDL pharmacology, Macrophages drug effects, RNA Interference, Reverse Transcriptase Polymerase Chain Reaction, Sequence Homology, Nucleic Acid, Cytokines metabolism, Lipids analysis, Lipoprotein Lipase genetics, Macrophages metabolism, MicroRNAs genetics
- Abstract
Background: Accumulating evidence suggests that microRNA-590 (miR-590) has protective effects on cardiovascular diseases, but the mechanism is unknown. Interestingly, previous studies from our laboratory and others have shown that macrophage-derived lipoprotein lipase (LPL) might accelerate atherosclerosis by promoting lipid accumulation and inflammatory response. However, the regulation of LPL at the post-transcriptional level by microRNAs has not been fully understood. In this study, we explored whether miR-590 affects the expression of LPL and its potential subsequent effects on lipid accumulation and pro-inflammatory cytokine secretion in human THP-1 macrophages., Methods and Results: Using bioinformatics analyses and dual-luciferase reporter assays, we found that miR-590 directly inhibited LPL protein and mRNA expression by targeting LPL 3'UTR. LPL Activity Assays showed that miR-590 reduced LPL activity in the culture media. Oil Red O staining and high-performance liquid chromatography assays showed that miR-590 had inhibitory effects on the lipid accumulation in human THP-1 macrophages. We also illustrated that miR-590 alleviated pro-inflammatory cytokine secretion in human THP-1 macrophages as measured by ELISA. With the method of small interfering RNA, we found that LPL siRNA can inhibit the miR-590 inhibitor-induced increase in lipid accumulation and secretion of pro-inflammatory cytokines in oxLDL-treated human THP-1 macrophages., Conclusions: MiR-590 attenuates lipid accumulation and pro-inflammatory cytokine secretion by targeting LPL gene in human THP-1 macrophages. Therefore, targeting miR-590 may offer a promising strategy to treat atherosclerotic cardiovascular diseases., (Copyright © 2014 Elsevier B.V. and Société française de biochimie et biologie Moléculaire (SFBBM). All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
21. MicroRNA-19b promotes macrophage cholesterol accumulation and aortic atherosclerosis by targeting ATP-binding cassette transporter A1.
- Author
-
Lv YC, Tang YY, Peng J, Zhao GJ, Yang J, Yao F, Ouyang XP, He PP, Xie W, Tan YL, Zhang M, Liu D, Tang DP, Cayabyab FS, Zheng XL, Zhang DW, Tian GP, and Tang CK
- Subjects
- ATP Binding Cassette Transporter 1 genetics, ATP Binding Cassette Transporter 1 physiology, Animals, Aortic Diseases genetics, Aortic Diseases metabolism, Apolipoprotein A-I metabolism, Apolipoproteins E deficiency, Atherosclerosis genetics, Atherosclerosis metabolism, Base Sequence, Cell Line, Cholesterol Esters metabolism, Collagen analysis, Foam Cells metabolism, Macrophages, Peritoneal metabolism, Male, Mice, Mice, Knockout, Molecular Sequence Data, Plaque, Atherosclerotic metabolism, RNA, Messenger metabolism, Sequence Homology, Nucleic Acid, ATP Binding Cassette Transporter 1 antagonists & inhibitors, Aortic Diseases etiology, Atherosclerosis etiology, Cholesterol metabolism, Macrophages metabolism, MicroRNAs physiology
- Abstract
Rationale: Macrophage accumulation of cholesterol leads to foam cell formation which is a major pathological event of atherosclerosis. Recent studies have shown that microRNA (miR)-19b might play an important role in cholesterol metabolism and atherosclerotic diseases. Here, we have identified miR-19b binding to the 3'UTR of ATP-binding cassette transporter A1 (ABCA1) transporters, and further determined the potential roles of this novel interaction in atherogenesis., Objective: To investigate the molecular mechanisms involved in a miR-19b promotion of macrophage cholesterol accumulation and the development of aortic atherosclerosis., Methods and Results: We performed bioinformatics analysis using online websites, and found that miR-19b was highly conserved during evolution and directly bound to ABCA1 mRNA with very low binding free energy. Luciferase reporter assay confirmed that miR-19b bound to 3110-3116 sites within ABCA1 3'UTR. MiR-19b directly regulated the expression levels of endogenous ABCA1 in foam cells derived from human THP-1 macrophages and mouse peritoneal macrophages (MPMs) as determined by qRT-PCR and western blot. Cholesterol transport assays revealed that miR-19b dramatically suppressed apolipoprotein AI-mediated ABCA1-dependent cholesterol efflux, resulting in the increased levels of total cholesterol (TC), free cholesterol (FC) and cholesterol ester (CE) as revealed by HPLC. The excretion of (3)H-cholesterol originating from cholesterol-laden MPMs into feces was decreased in mice overexpressing miR-19b. Finally, we evaluated the proatherosclerotic role of miR-19b in apolipoprotein E deficient (apoE(-/-)) mice. Treatment with miR-19b precursor reduced plasma high-density lipoprotein (HDL) levels, but increased plasma low-density lipoprotein (LDL) levels. Consistently, miR-19b precursor treatment increased aortic plaque size and lipid content, but reduced collagen content and ABCA1 expression. In contrast, treatment with the inhibitory miR-19b antisense oligonucleotides (ASO) prevented or reversed these effects., Conclusion: MiR-19b promotes macrophage cholesterol accumulation, foam cell formation and aortic atherosclerotic development by targeting ABCA1., (Copyright © 2014 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
22. Chlamydia pneumoniae negatively regulates ABCA1 expression via TLR2-Nuclear factor-kappa B and miR-33 pathways in THP-1 macrophage-derived foam cells.
- Author
-
Zhao GJ, Mo ZC, Tang SL, Ouyang XP, He PP, Lv YC, Yao F, Tan YL, Xie W, Shi JF, Wang Y, Zhang M, Liu D, Tang DP, Zheng XL, Tian GP, and Tang CK
- Subjects
- Cholesterol metabolism, Foam Cells microbiology, Humans, Macrophages metabolism, ATP Binding Cassette Transporter 1 biosynthesis, Chlamydophila pneumoniae physiology, Foam Cells metabolism, MicroRNAs physiology, NF-kappa B physiology, Toll-Like Receptor 2 physiology
- Abstract
Objectives: ATP-binding cassette transporter A1 (ABCA1) is critical in exporting cholesterol from macrophages and plays a protective role in the development of atherosclerosis. This study was to determine the effects and potential mechanisms of Chlamydia pneumoniae (C. pneumoniae) on ABCA1 expression and cellular cholesterol efflux in THP-1 macrophage-derived foam cells., Methods and Results: C. pneumoniae significantly decreased the expression of ABCA1 and reduced cholesterol efflux. Furthermore, we found that C. pneumoniae suppressed ABCA1 expression via up-regulation of miR-33s. The inhibition of C. pneumoniae-induced NF-κB activation decreased miR-33s expression and enhanced ABCA1 expression. In addition, C. pneumoniae increased Toll-like receptor 2 (TLR2) expressions, inhibition of which by siRNA could also block NF-κB activation and miR-33s expression, and promot the expression of ABCA1., Conclusion: Taken together, these results reveal that C. pneumoniae may negatively regulate ABCA1 expression via TLR2-NF-κB and miR-33 pathways in THP-1 macrophage-derived foam cells, which may provide new insights for understanding the effects of C. pneumoniae on the pathogenesis of atherosclerosis., (Copyright © 2014 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
23. MicroRNA-27a/b regulates cellular cholesterol efflux, influx and esterification/hydrolysis in THP-1 macrophages.
- Author
-
Zhang M, Wu JF, Chen WJ, Tang SL, Mo ZC, Tang YY, Li Y, Wang JL, Liu XY, Peng J, Chen K, He PP, Lv YC, Ouyang XP, Yao F, Tang DP, Cayabyab FS, Zhang DW, Zheng XL, Tian GP, and Tang CK
- Subjects
- Cells, Cultured, Esterification, Humans, Hydrolysis, Cholesterol metabolism, Macrophages metabolism, MicroRNAs physiology
- Abstract
Rationale: Macrophage cholesterol homeostasis maintenance is the result of a balance between influx, endogenous synthesis, esterification/hydrolysis and efflux. Excessive accumulation of cholesterol leads to foam cell formation, which is the major pathology of atherosclerosis. Previous studies have shown that miR-27 (miR-27a and miR-27b) may play a key role in the progression of atherosclerosis., Objective: We set out to investigate the molecular mechanisms of miR-27a/b in intracellular cholesterol homeostasis., Methods and Results: In the present study, our results have shown that the miR-27 family is highly conserved during evolution, present in mammals and directly targets the 3' UTR of ABCA1, LPL, and ACAT1. apoA1, ABCG1 and SR-B1 lacking miR-27 bind sites should not be influenced by miR-27 directly. miR-27a and miR-27b directly regulated the expression of endogenous ABCA1 in different cells. Treatment with miR-27a and miR-27b mimics reduced apoA1-mediated cholesterol efflux by 33.08% and 44.61% in THP-1 cells, respectively. miR-27a/b also regulated HDL-mediated cholesterol efflux in THP-1 macrophages and affected the expression of apoA1 in HepG2 cells. However, miR-27a/b had no effect on total cellular cholesterol accumulation, but regulated the levels of cellular free cholesterol and cholesterol ester. We further found that miR-27a/b regulated the expression of LPL and CD36, and then affected the ability of THP-1 macrophages to uptake Dil-oxLDL. Finally, we identified that miR-27a/b regulated cholesterol ester formation by targeting ACAT1 in THP-1 macrophages., Conclusion: These findings indicate that miR-27a/b affects the efflux, influx, esterification and hydrolysis of cellular cholesterol by regulating the expression of ABCA1, apoA1, LPL, CD36 and ACAT1., (Copyright © 2014 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
24. NF-κB suppresses the expression of ATP-binding cassette transporter A1/G1 by regulating SREBP-2 and miR-33a in mice.
- Author
-
Zhao GJ, Tang SL, Lv YC, Ouyang XP, He PP, Yao F, Tang YY, Zhang M, Tang YL, Tang DP, Cayabyab FS, Tian GP, and Tang CK
- Subjects
- ATP Binding Cassette Transporter 1 antagonists & inhibitors, ATP Binding Cassette Transporter, Subfamily G, Member 1, ATP-Binding Cassette Transporters antagonists & inhibitors, Animals, Cell Line, Lipoproteins antagonists & inhibitors, Macrophages metabolism, Male, Mice, Mice, Knockout, MicroRNAs antagonists & inhibitors, Random Allocation, Sterol Regulatory Element Binding Protein 2 antagonists & inhibitors, ATP Binding Cassette Transporter 1 biosynthesis, ATP-Binding Cassette Transporters biosynthesis, Gene Expression Regulation, Lipoproteins biosynthesis, MicroRNAs physiology, NF-kappa B metabolism, Sterol Regulatory Element Binding Protein 2 physiology
- Published
- 2014
- Full Text
- View/download PDF
25. The effects of miR-467b on lipoprotein lipase (LPL) expression, pro-inflammatory cytokine, lipid levels and atherosclerotic lesions in apolipoprotein E knockout mice.
- Author
-
Tian GP, Tang YY, He PP, Lv YC, Ouyang XP, Zhao GJ, Tang SL, Wu JF, Wang JL, Peng J, Zhang M, Li Y, Cayabyab FS, Zheng XL, Zhang DW, Yin WD, and Tang CK
- Subjects
- Animals, Atherosclerosis prevention & control, Gene Expression Regulation, Enzymologic drug effects, Inflammation prevention & control, Lipid Metabolism drug effects, Lipoprotein Lipase biosynthesis, Male, Mice, Mice, Knockout, Treatment Outcome, Apolipoproteins E genetics, Atherosclerosis immunology, Cytokines immunology, Inflammation immunology, Lipid Metabolism immunology, Lipoprotein Lipase immunology, MicroRNAs pharmacology
- Abstract
Atherosclerosis is a lipid disorder disease characterized by chronic blood vessel wall inflammation driven by the subendothelial accumulation of macrophages. Studies have shown that lipoprotein lipase (LPL) participates in lipid metabolism, but it is not yet known whether post-transcriptional regulation of LPL gene expression by microRNAs (miRNAs) occurs in vivo. Here, we tested that miR-467b provides protection against atherosclerosis by regulating the target gene LPL which leads to reductions in LPL expression, lipid accumulation, progression of atherosclerosis and production of inflammatory cytokines in apolipoprotein E knockout (apoE(-/-)) mice. Treatment of apoE(-/-) mice with intra-peritoneal injection of miR-467b agomir led to decreased blood plasma levels of total cholesterol (TC), triglyceride (TG), low density lipoprotein cholesterol (LDL-C), high density lipoprotein cholesterol (HDL-C), tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), IL-1β and monocyte chemotactic protein-1 (MCP-1). Using Western blots and real time PCR, we determined that LPL expression in aorta and abdominal cavity macrophages were significantly down-regulated in the miR-467b agomir group. Furthermore, systemic treatment with miR-467b antagomir accelerated the progression of atherosclerosis in the aorta of apoE(-/-) mice. The present study showed that miR-467b protects apoE(-/-) mice from atherosclerosis by reducing lipid accumulation and inflammatory cytokine secretion via downregulation of LPL expression. Therefore, targeting miR-467b may offer a promising strategy to treat atherosclerotic vascular disease., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
26. Antagonism of betulinic acid on LPS-mediated inhibition of ABCA1 and cholesterol efflux through inhibiting nuclear factor-kappaB signaling pathway and miR-33 expression.
- Author
-
Zhao GJ, Tang SL, Lv YC, Ouyang XP, He PP, Yao F, Chen WJ, Lu Q, Tang YY, Zhang M, Fu Y, Zhang DW, Yin K, and Tang CK
- Subjects
- ATP Binding Cassette Transporter 1 metabolism, Animals, Apolipoproteins E deficiency, Apolipoproteins E metabolism, Atherosclerosis blood, Atherosclerosis metabolism, Atherosclerosis pathology, Biological Transport drug effects, Body Weight drug effects, Cell Line, Cell Nucleus drug effects, Cell Nucleus metabolism, Gene Expression Regulation drug effects, Humans, Lipids blood, Macrophages drug effects, Macrophages metabolism, Male, Mice, MicroRNAs metabolism, Models, Biological, Pentacyclic Triterpenes, Protein Transport drug effects, Signal Transduction drug effects, Signal Transduction genetics, Betulinic Acid, ATP Binding Cassette Transporter 1 antagonists & inhibitors, Cholesterol metabolism, Lipopolysaccharides pharmacology, MicroRNAs genetics, NF-kappa B metabolism, Triterpenes antagonists & inhibitors, Triterpenes pharmacology
- Abstract
ATP-binding cassette transporter A1 (ABCA1) is critical in exporting cholesterol from macrophages and plays a protective role in the development of atherosclerosis. The purpose of this study was to investigate the effects of betulinic acid (BA), a pentacyclic triterpenoid, on ABCA1 expression and cholesterol efflux, and to further determine the underlying mechanism. BA promoted ABCA1 expression and cholesterol efflux, decreased cellular cholesterol and cholesterol ester content in LPS-treated macrophages. Furthermore, we found that BA promoted ABCA1 expression via down-regulation of miR-33s. The inhibition of LPS-induced NF-κB activation further decreased miR-33s expression and enhanced ABCA1 expression and cholesterol efflux when compared with BA only treatment. In addition, BA suppressed IκB phosphorylation, p65 phosphorylation and nuclear translocation, and the transcription of NF-κB-dependent related gene. Moreover, BA reduced atherosclerotic lesion size, miR-33s levels and NF-κB activation, and promoted ABCA1 expression in apoE(-/-) mice. Taken together, these results reveal a novel mechanism for the BA-mediated ABCA1 expression, which may provide new insights for developing strategies for modulating vascular inflammation and atherosclerosis.
- Published
- 2013
- Full Text
- View/download PDF
27. MicroRNA-33 in atherosclerosis etiology and pathophysiology.
- Author
-
Chen WJ, Zhang M, Zhao GJ, Fu Y, Zhang DW, Zhu HB, and Tang CK
- Subjects
- Animals, Atherosclerosis etiology, Bile Acids and Salts metabolism, Cardiovascular System, Cell Cycle, Cell Differentiation, Cell Proliferation, Fatty Acids metabolism, Homeostasis, Humans, Inflammation, Insulin metabolism, Lipids, Risk Factors, Atherosclerosis genetics, Atherosclerosis physiopathology, MicroRNAs metabolism
- Abstract
MicroRNAs are a group of endogenous, small non-coding RNA molecules that can induce translation repression of target genes within metazoan cells by specific base pairing with the mRNA of target genes. Recently, microRNA-33 has been discovered as a key regulator in the initiation and progression of atherosclerosis. This review highlights the impact of microRNA-33-mediated regulation in the major cardiometabolic risk factors of atherosclerosis including lipid metabolism (HDL biogenesis and cholesterol homeostasis, fatty acid, phospholipid and triglyceride, bile acids metabolism), inflammatory response, insulin signaling and glucose/energy homeostasis, cell cycle progression and proliferation, and myeloid cell differentiation. Understanding the etiology and pathophysiology of microRNA-33 in atherosclerosis may provide basic knowledge for the development of novel therapeutic targets for ameliorating atherosclerosis and cardiovascular disease., (Copyright © 2012 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
28. MicroRNA-467b targets LPL gene in RAW 264.7 macrophages and attenuates lipid accumulation and proinflammatory cytokine secretion.
- Author
-
Tian GP, Chen WJ, He PP, Tang SL, Zhao GJ, Lv YC, Ouyang XP, Yin K, Wang PP, Cheng H, Chen Y, Huang SL, Fu Y, Zhang DW, Yin WD, and Tang CK
- Subjects
- 3' Untranslated Regions genetics, Animals, Base Sequence, Blotting, Western, Cell Line, Chemokine CCL2 metabolism, Chromatography, High Pressure Liquid, HEK293 Cells, Humans, Inflammation Mediators metabolism, Interleukin-1beta metabolism, Interleukin-6 metabolism, Lipid Metabolism drug effects, Lipoprotein Lipase metabolism, Lipoproteins, LDL pharmacology, Macrophages cytology, Macrophages drug effects, Mice, MicroRNAs metabolism, RNA Interference, Reverse Transcriptase Polymerase Chain Reaction, Sequence Homology, Nucleic Acid, Tumor Necrosis Factor-alpha metabolism, Cytokines metabolism, Lipids analysis, Lipoprotein Lipase genetics, Macrophages metabolism, MicroRNAs genetics
- Abstract
LPL (lipoprotein lipase) is a rate-limiting enzyme involved in the hydrolysis of triglycerides. Previous studies have shown that microRNA (miR)-467b regulates hepatic LPL expression and plays a role in the progression of steatosis or abnormal lipid retention in obese mice. Macrophage-derived LPL has been shown to promote atherosclerosis. However, if miR-476b influences macrophage LPL expression and the subsequent effects are unknown. Here, we utilized oxLDL-treatment RAW 264.7 macrophages that were transfected with miR-467b mimics or inhibitors to investigate the potential roles of macrophage miR-476b. We found that miR-467b significantly decreased lipid accumulation and IL-6, IL-1β, TNF-α and MCP-1 secretions. Furthermore, our studies suggested an additional explanation for the regulatory mechanism of miR-467b on its functional target, LPL in RAW 264.7 macrophages. Thus, our findings indicate that miR-467b may regulate lipid accumulation and proinflammatory cytokine secretion in oxLDL-stimulated RAW 264.7 macrophages by targeting the LPL gene., (Copyright © 2012 Elsevier Masson SAS. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
29. The magic and mystery of microRNA-27 in atherosclerosis.
- Author
-
Chen WJ, Yin K, Zhao GJ, Fu YC, and Tang CK
- Subjects
- Animals, Atherosclerosis metabolism, Atherosclerosis pathology, Atherosclerosis physiopathology, Atherosclerosis therapy, Blood Vessels pathology, Blood Vessels physiopathology, CCAAT-Enhancer-Binding Proteins metabolism, Cell Cycle Proteins metabolism, Core Binding Factor Alpha 2 Subunit metabolism, Disease Progression, Gene Expression Regulation, Humans, Inflammation metabolism, Inflammation pathology, Inflammation physiopathology, Lipid Metabolism, Receptors, G-Protein-Coupled metabolism, Atherosclerosis genetics, Blood Vessels metabolism, Inflammation genetics, MicroRNAs metabolism
- Abstract
Atherosclerosis (As) is now widely appreciated to represent a chronic inflammatory reaction of the vascular wall in response to dyslipidemia and endothelial distress involving the inflammatory recruitment of leukocytes and the activation of resident vascular cells. MicroRNAs (miRNAs) are a group of endogenous, small (~22 nucleotides in length) non-coding RNA molecules, which function specifically by base pairing with mRNA of genes, thereby induce translation repressions of the genes within metazoan cells. Recently, the function of miR-27, one of the miRNAs, in the initiation and progression of atherosclerosis has been identified. In vivo and in vitro studies suggest that miR-27 may serve as a diagnostic and prognostic marker for atherosclerosis. More recently, studies have identified important roles for miR-27 in angiogenesis, adipogenesis, inflammation, lipid metabolism, oxidative stress, insulin resistance and type 2 diabetes, etc. In this review, we focus on the role of miR-27 in the development of vulnerable atherosclerotic plaques, potential as a disease biomarker and novel therapeutic target in atherosclerosis., (Copyright © 2012 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.