Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Cell Biochem ; 120(8): 13775-13782, 2019 08.
Article in English | MEDLINE | ID: mdl-30938872

ABSTRACT

Atherosclerosis is a complex inflammatory disease that involves disrupted cellular cholesterol levels and formation of foam cells. Studies about long noncoding RNA (lncRNA) have revealed its function in the development of atherosclerosis, by mediating reverse cholesterol transport and formation of foam cells. In this study, we found that oxidized low-density lipoprotein (ox-LDL) markedly decreased lncRNA AC096664.3 in vascular smooth muscle cells (VSMCs) and THP-1 macrophages. We also found that ox-LDL reduced ATP-binding cassette (ABC) G1 through inhibiting lncRNA AC096664.3 in VSMCs. Further experiments showed that the downregulation of lncRNA AC096664.3 reduced ABCG1 expression through inhibiting the expression of peroxisome proliferator-activated receptor-γ (PPAR-γ) and that ox-LDL reduced ABCG1 expression through inhibiting the expression of PPAR-γ. Furthermore, we discovered that ox-LDL inhibited ABCG1 via the lncRNA AC096664.3/PPAR-γ/ABCG1 pathway, which led to an increase in total and free cholesterol in VMSCs. Thus, we confirmed that ox-LDL induces cholesterol accumulation via the lncRNA AC096664.3/PPAR-γ/ABCG1 pathway in VSMCs, indicating a promising novel therapy in protecting against atherosclerosis.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Cholesterol/metabolism , Homeostasis , PPAR gamma/metabolism , RNA, Long Noncoding/metabolism , Signal Transduction , ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cholesterol/genetics , Humans , Lipoproteins, LDL/genetics , Lipoproteins, LDL/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , PPAR gamma/genetics , RNA, Long Noncoding/genetics , THP-1 Cells
2.
DNA Cell Biol ; 38(6): 583-591, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30994379

ABSTRACT

Atherosclerosis is a chronic vascular inflammatory disease that involves diverse cell types and circulating regulatory factors, including intercellular adhesion molecule (ICAM)-1, a proinflammatory cytokine. Lipopolysaccharides (LPS) increase ICAM-1 expression and promote cell adhesion, but the mechanism is not clear. We found that LPS induced time- and dose-regulated upregulation of ICAM-1 expression and downregulation of forkhead box protein C2 (Foxc2) expression in human umbilical vein endothelial cells (HUVECs). Overexpression of Foxc2 significantly inhibited both LPS-induced ICAM-1 expression in HUVECs and LPS-induced adhesion of THP-1 cells to HUVECs. Foxc2 siRNA dramatically increased both LPS-induced ICAM-1 expression and LPS-induced adhesion of THP-1 human monocytes cells to HUVECs. We conclude that Foxc2 inhibited LPS-induced adhesion of THP-1 cells to HUVECs by suppressing ICAM-1 expression in HUVECs.


Subject(s)
Cell Adhesion , Forkhead Transcription Factors/physiology , Human Umbilical Vein Endothelial Cells/metabolism , Intercellular Adhesion Molecule-1/metabolism , Cell Adhesion/drug effects , Cells, Cultured , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Intercellular Adhesion Molecule-1/genetics , Lipopolysaccharides/pharmacology , RNA, Messenger/metabolism
3.
Cell Death Differ ; 26(9): 1670-1687, 2019 09.
Article in English | MEDLINE | ID: mdl-30683918

ABSTRACT

Atherosclerosis is a progressive, chronic inflammation in arterial walls. Long noncoding RNAs (lncRNAs) participate in inflammation, but the exact mechanism in atherosclerosis is unclear. Our microarray analyses revealed that the levels of lncRNA-FA2H-2 were significantly decreased by oxidized low-density lipoprotein (OX-LDL). Bioinformatics analyses indicated that mixed lineage kinase domain-like protein (MLKL) might be regulated by lncRNA-FA2H-2. In vitro experiments showed that lncRNA-FA2H-2 interacted with the promoter of the MLKL gene, downregulated MLKL expression, and the binding sites between -750 and 471 were necessary for lncRNA-FA2H-2 responsiveness to MLKL. Silencing lncRNA-FA2H-2 and overexpression of MLKL could activate inflammation and inhibited autophagy flux. Both lncRNA-FA2H-2 knockdown and overexpression of MLKL could significantly aggravate inflammatory responses induced by OX-LDL. We found that the 3-methyladenine (3-MA) and Atg7-shRNA enhanced inflammatory responses induced by knockdown of lncRNA-FA2H-2 and overexpression of MLKL. We demonstrated that the effects of MLKL on autophagy might be associated with a mechanistic target of rapamycin (mTOR)-dependent signaling pathways. In vivo experiments with apoE knockout mice fed a western diet demonstrated that LncRNA-FA2H-2 knockdown decreased microtubule-associated expression of microtubule-associated protein 1 light chain 3 II and lysosome-associated membrane protein 1, but increased expression of sequestosome 1 (p62), MLKL, vascular cell adhesion molecule-1, monocyte chemoattractant protein-1, and interleukin-6 in atherosclerotic lesions. Our findings indicated that the lncRNA-FA2H-2-MLKL pathway is essential for regulation of autophagy and inflammation, and suggested that lncRNA-FA2H-2 and MLKL could act as potential therapeutic targets to ameliorate atherosclerosis-related diseases.


Subject(s)
Atherosclerosis/metabolism , Autophagy/genetics , Inflammation/metabolism , Lipoproteins, LDL/metabolism , Mixed Function Oxygenases/metabolism , Protein Kinases/metabolism , RNA, Long Noncoding/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Atherosclerosis/genetics , Autophagosomes/metabolism , Autophagosomes/ultrastructure , Autophagy/drug effects , Autophagy-Related Protein 7/genetics , Autophagy-Related Protein 7/metabolism , Chromatin Immunoprecipitation Sequencing , Human Umbilical Vein Endothelial Cells , Humans , Inflammation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , Mixed Function Oxygenases/antagonists & inhibitors , Mixed Function Oxygenases/genetics , Protein Kinases/genetics , RNA, Long Noncoding/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tissue Array Analysis
4.
J Clin Invest ; 129(3): 1115-1128, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30589415

ABSTRACT

Noncoding RNAs are emerging as important players in gene regulation and disease pathogeneses. Here, we show that a previously uncharacterized long noncoding RNA, nexilin F-actin binding protein antisense RNA 1 (NEXN-AS1), modulates the expression of the actin-binding protein NEXN and that NEXN exerts a protective role against atherosclerosis. An expression microarray analysis showed that the expression of both NEXN-AS1 and NEXN was reduced in human atherosclerotic plaques. In vitro experiments revealed that NEXN-AS1 interacted with the chromatin remodeler BAZ1A and the 5' flanking region of the NEXN gene and that it also upregulated NEXN expression. Augmentation of NEXN-AS1 expression inhibited TLR4 oligomerization and NF-κB activity, downregulated the expression of adhesion molecules and inflammatory cytokines by endothelial cells, and suppressed monocyte adhesion to endothelial cells. These inhibitory effects of NEXN-AS1 were abolished by knockdown of NEXN. In vivo experiments using ApoE-knockout mice fed a Western high-fat diet demonstrated that NEXN deficiency promoted atherosclerosis and increased macrophage abundance in atherosclerotic lesions, with heightened expression of adhesion molecules and inflammatory cytokines, whereas augmented NEXN expression deterred atherosclerosis. Patients with coronary artery disease were found to have lower blood NEXN levels than healthy individuals. These results indicate that NEXN-AS1 and NEXN represent potential therapeutic targets in atherosclerosis-related diseases.


Subject(s)
Atherosclerosis/metabolism , Coronary Artery Disease/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Microfilament Proteins/biosynthesis , Plaque, Atherosclerotic/metabolism , RNA, Long Noncoding/metabolism , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Coronary Artery Disease/genetics , Coronary Artery Disease/pathology , Down-Regulation , Human Umbilical Vein Endothelial Cells/pathology , Humans , Mice , Mice, Knockout, ApoE , Microfilament Proteins/genetics , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/pathology , RNA, Long Noncoding/genetics , THP-1 Cells
5.
Immunol Cell Biol ; 96(2): 175-189, 2018 02.
Article in English | MEDLINE | ID: mdl-29363163

ABSTRACT

Atherosclerotic cardiovascular disease is considered as the leading cause of mortality and morbidity worldwide. Accumulating evidence supports an important role for long noncoding RNA (lncRNA) in the pathogenesis of atherosclerosis. Nevertheless, the role of lncRNA in atherosclerosis-associated vascular dysfunction and the underlying mechanism remain elusive. Here, using microarray analysis, we identified a novel lncRNA RP11-714G18.1 with significant reduced expression in human advanced atherosclerotic plaque tissues. We demonstrated in both human vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) that RP11-714G18.1 impaired cell migration, reduced the adhesion of ECs to monocytes, suppressed the neoangiogenesis, decreased apoptosis of VSMCs and promoted nitric oxide production. Mechanistically, RP11-714G18.1 could directly bind to its nearby gene LRP2BP and increased the expression of LRP2BP. Moreover, we showed that RP11-714G18.1 impaired cell migration through LRP2BP-mediated downregulation of matrix metalloproteinase (MMP)1 in both ECs and VSMCs. In atherosclerotic patients, the serum levels of LRP2BP were positively correlated with high-density lipoprotein cholesterol, but negatively correlated with cardiac troponin I. Our study suggests that RP11-714G18.1 may play an athero-protective role by inhibiting vascular cell migration via RP11-714G18.1/LRP2BP/MMP1 signaling pathway, and targeting the pathway may provide new therapeutic approaches for atherosclerosis.


Subject(s)
Carrier Proteins/metabolism , Cell Movement , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , RNA, Long Noncoding/metabolism , Adaptor Proteins, Signal Transducing , Adult , Aged , Aged, 80 and over , Apoptosis/genetics , Base Sequence , Carrier Proteins/blood , Carrier Proteins/genetics , Cell Adhesion/genetics , Cell Cycle/genetics , Cell Movement/genetics , Cholesterol, HDL/metabolism , Female , Gene Expression Regulation , Humans , Low Density Lipoprotein Receptor-Related Protein-2 , Male , Matrix Metalloproteinase 1/metabolism , Middle Aged , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Neovascularization, Physiologic , Nitric Oxide/biosynthesis , Open Reading Frames/genetics , Plaque, Atherosclerotic/blood , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/pathology , RNA, Long Noncoding/genetics , Troponin I/metabolism
6.
DNA Cell Biol ; 35(11): 722-729, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27574949

ABSTRACT

Atherosclerosis is a common pathological basis of cardiovascular disease, which remains the leading cause of mortality. Long noncoding RNAs (lncRNAs) are newly studied non-protein-coding RNAs involved in gene regulation, but how lncRNAs exert regulatory effect on atherosclerosis remains unclear. In this study, we found that lncRNA HOXC cluster antisense RNA 1 (HOXC-AS1) and homeobox C6 (HOXC6) were downregulated in carotid atherosclerosis by performing microarray analysis. The results were verified in atherosclerotic plaques and normal arterial intima tissues by quantitative reverse transcription PCR and western blot analysis. Lentivirus-mediated overexpression of HOXC-AS1 induced HOXC6 expression at mRNA and protein levels in THP-1 macrophages. Besides, oxidized low-density lipoprotein (Ox-LDL) decreased expression of HOXC-AS1 and HOXC6 in a time-dependent manner. Induction of cholesterol accumulation by Ox-LDL could be partly suppressed by overexpression of HOXC-AS1.


Subject(s)
Cholesterol/metabolism , Homeodomain Proteins/metabolism , Lipoproteins, LDL/metabolism , Macrophages/metabolism , RNA, Long Noncoding/genetics , Atherosclerosis/metabolism , Cell Line , Gene Expression Regulation/drug effects , Humans , Lipoproteins, LDL/genetics , Real-Time Polymerase Chain Reaction
7.
Arch Biochem Biophys ; 604: 27-35, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27267730

ABSTRACT

BACKGROUND: Atherosclerosis is a chronic inflammatory disease and represents the leading cause of morbidity and mortality throughout the world. Accumulating evidences have showed that Dihydrocapsaicin (DHC) has been found to exert multiple pharmacological and physiological effects. Nevertheless, the effects and possible mechanism of DHC on proinflammatory response remain largely unexplained. METHODS AND RESULTS: We found that DHC markedly upregulated NFIA and suppressed NF-κB expression in THP-1 macrophages. Up-regulation of proinflammatory cytokines induced by LPS including TNF-α, IL-1ß and IL-6 were markedly suppressed by DHC treatment. We also observed that protein level of NFIA was significantly increased while NF-κB and proinflammatory cytokines were decreased by DHC treatment in apoE(-/-) mice. Lentivirus-mediated overexpression of NFIA suppressed NF-κB and proinflammatory cytokines expression both in THP-1 macrophages and plaque tissues of apoE-/- mice. Moreover, treatment with lentivirus-mediated overexpression of NFIA made the down-regulation of DHC on NF-κB and proinflammatory cytokines expression notably accentuated in THP-1 macrophages and apoE(-/-) mice. In addition, treatment with siRNA targeting NF-κB accentuated the suppression of proinflammatory cytokines by lentivirus-mediated overexpression of NFIA. CONCLUSION: These observations demonstrated that DHC can significantly decrease proinflammatory cytokines through enhancing NFIA and inhibiting NF-κB expression and thus DHC may be a promising candidate as an anti-inflammatory drug for atherosclerosis as well as other disorders.


Subject(s)
Capsaicin/analogs & derivatives , Cytokines/metabolism , Gene Expression Regulation , NF-kappa B/metabolism , NFI Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Anti-Inflammatory Agents/chemistry , Apolipoproteins E/genetics , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Capsaicin/chemistry , Gene Expression Profiling , Humans , Inflammation , Interleukin-6/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , RNA, Small Interfering/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...