Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Nitric Oxide ; 142: 47-57, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38049061

ABSTRACT

BACKGROUND: Endothelial-mesenchymal transition (EndMT) induced by low shear stress plays an important role in the development of atherosclerosis. However, little is known about the correlation between hydrogen sulfide (H2S), a protective gaseous mediator in atherosclerosis and the process of EndMT. METHODS: We constructed a stable low-shear-stress-induced(2 dyn/cm2) EndMT model, acombined with the pretreatment method of hydrogen sulfide slow release agent(GYY4137). The level of MEST was detected in the common carotid artery of ApoE-/- mice with local carotid artery ligation. The effect of MEST on atherosclerosis development in vivo was verified using ApoE-/- mice were given tail-vein injection of endothelial-specific overexpressed and knock-down MEST adeno-associated virus (AAV). RESULTS: These findings confirmed that MEST is up-regulated in low-shear-stress-induced EndMT and atherosclerosis. In vivo experiments showed that MEST gene overexpression significantly promoted EndMT and aggravated the development of atherosclerotic plaques and MEST gene knockdown significantly inhibited EndMT and delayed the process of atherosclerosis. In vitro, H2S inhibits the expression of MEST and EndMT induced by low shear stress and inhibits EndMT induced by MEST overexpression. Knockdown of NFIL3 inhibit the up regulation of MEST and EndMT induced by low shear stress in HUVECs. CHIP-qPCR assay and Luciferase Reporter assay confirmed that NFIL3 binds to MEST DNA, increases its transcription and H2S inhibits the binding of NFIL3 and MEST DNA, weakening NFIL3's transcriptional promotion of MEST. Mechanistically, H2S increased the sulfhydrylation level of NFIL3, an important upstream transcription factors of MEST. In part, transcription factor NFIL3 restrain its binding to MEST DNA by sulfhydration. CONCLUSIONS: H2S negatively regulate the expression of MEST by sulfhydrylation of NFIL3, thereby inhibiting low-shear-stress-induced EndMT and atherosclerosis.


Subject(s)
Atherosclerosis , Hydrogen Sulfide , Mice , Animals , Humans , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/metabolism , Endothelial-Mesenchymal Transition , Atherosclerosis/genetics , Atherosclerosis/metabolism , Endothelium/metabolism , DNA/metabolism , Apolipoproteins E/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Epithelial-Mesenchymal Transition
2.
Nitric Oxide ; 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39305980

ABSTRACT

BACKGROUND: Under normal circumstances, high-density lipoprotein (HDL) is considered to have cardiovascular protective effects, but the impact of oxidized HDL (ox-HDL) on vascular endothelial function remains poorly understood. Mitochondrial function is closely related to endothelial function, and hydrogen sulfide (H2S) is a gas with endothelial protective properties. The novel hydrogen sulfide donor AP39 can target mitochondria to release H2S, but the combined effects of ox-HDL and AP39 on vascular endothelium are not well studied. METHODS: We established a cell model of ox-HDL-induced endothelial cell damage and mitochondrial dysfunction using human umbilical vein endothelial cells (HUVECs) and conducted AP39 pretreatment. The experiments confirmed the functional damage and mitochondrial dysfunction in HUVECs caused by ox-HDL. Additionally, to further explore the role of SIRT1 in AS, we analyzed SIRT1 expression in AS carotid artery tissue. This included the analysis of differentially expressed genes from AS-related datasets, presented through volcano plots and heatmaps, with enrichment analysis of downregulated genes in KEGG pathways and GO functions. Furthermore, we evaluated the differences in SIRT1 expression in coronary arteries with varying degrees of stenosis and in early and late-stage AS carotid artery tissues, and analyzed data from SIRT1 knockout mouse models. RESULTS: The experimental results indicate that AP39 effectively alleviated ox-HDL-induced endothelial cell damage and mitochondrial dysfunction by upregulating SIRT1 expression. MTT and CCK-8 assays showed that ox-HDL treatment led to decreased cell viability and proliferation in HUVECs, reduced eNOS expression, and significantly increased levels of ICAM-1, IL-6, and TNF-α, along with enhanced monocyte adhesion. These findings reveal the damaging effects of ox-HDL on HUVECs. Transcriptomic data indicated that while SIRT1 expression did not significantly differ in coronary arteries with varying degrees of stenosis, it was notably downregulated in AS carotid artery tissues, especially in late-stage AS tissues. KEGG pathway enrichment analysis revealed that SIRT1 downregulated genes were associated with processes such as vascular smooth muscle contraction, while GO analysis showed that these downregulated genes were involved in muscle system processes and muscle contraction functions, further confirming SIRT1's critical role in AS pathology. In transcriptomic data from the SIRT1 knockout mouse model, elevated levels of inflammation-related proteins IL-6 and TNF-α were observed after SIRT1 knockout, along with decreased expression of the chaperone protein PGC-1α. The expression of mitochondrial-related functional proteins Nrf2 and PGC-1α was positively correlated with SIRT1 expression, while inflammation-related proteins ICAM-1, IL-6, IL-20, and TNF-α were negatively correlated with SIRT1 expression. We further discovered that ox-HDL triggered mitochondrial dysfunction, as evidenced by reduced expression of Mfn2, Nrf2, PGC1-α, UCP-1, and SIRT1, corroborating the results from the previous database analysis. Additionally, mitochondrial dysfunction was characterized by decreased mitochondrial membrane potential (MMP), increased mitochondrial ROS levels, and reduced ATP content, further impacting cellular energy metabolism and respiratory function. Subsequent experimental results showed that the addition of AP39 mitigated these adverse effects, as evidenced by decreased levels of ICAM-1, IL-6, and TNF-α, increased eNOS expression, reduced monocyte adhesion, increased mitochondrial H2S content, and upregulated expression of SIRT1 protein associated with mitochondrial function, reduced ROS levels, and increased ATP content. Furthermore, validation experiments using the SIRT1 inhibitor EX527 confirmed that AP39 alleviated ox-HDL-induced endothelial cell damage and mitochondrial dysfunction by upregulating SIRT1 expression. CONCLUSION: Ox-HDL can induce damage and mitochondrial dysfunction in HUVECs, while AP39 inhibits ox-HDL-induced endothelial cell damage and mitochondrial dysfunction by upregulating SIRT1.

3.
Cardiovasc Drugs Ther ; 34(1): 113-121, 2020 02.
Article in English | MEDLINE | ID: mdl-32090295

ABSTRACT

Hydrogen sulfide (H2S), a novel gaseous signaling molecule, is a vital physiological signal in mammals. H2S protects the cardiovascular system via modulation of vasodilation, vascular remodeling, and inhibition of vascular calcification, and also has anti-atherosclerosis properties. Autophagy is a lysosomal-mediated intracellular degradation mechanism for excessive or abnormal proteins and lipids. The contribution of autophagy to normal and disease-state cell physiology is extremely complicated. Autophagy acts as a double-edged sword in the cardiovascular system. It can defend against damage to cells caused by environmental changes and it can also induce active cell death under certain conditions. In recent years, accumulating evidence indicates that H2S can up- or downregulate autophagy in many pathological processes, thereby switching from a harmful to a beneficial role. In this review, we summarize progress on understanding the mechanism by which H2S regulates autophagy in cardiovascular disease. We also discuss a H2S switch phenomenon that regulates autophagy and provides protection in cardiovascular diseases.


Subject(s)
Autophagy , Cardiovascular Diseases/metabolism , Cardiovascular System/metabolism , Hydrogen Sulfide/metabolism , Animals , Apoptosis , Autophagy/drug effects , Cardiovascular Agents/therapeutic use , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/pathology , Cardiovascular Diseases/physiopathology , Cardiovascular System/drug effects , Cardiovascular System/pathology , Cardiovascular System/physiopathology , Humans , Hydrogen Sulfide/therapeutic use , Signal Transduction
4.
Cell Biol Int ; 42(3): 313-323, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29064597

ABSTRACT

High concentrations of plasma lipoprotein(a) [Lp(a)] have been inferred to be an independent risk factor for cardiovascular and cerebrovascular diseases, such as coronary artery diseases, restenosis, and stroke. Apolipoprotein(a) [apo(a)] is one of the most important components of Lp(a) and contributes greatly to the increased concentration of plasma Lp(a). As a critical positive transacting factor of apo(a) gene, Ets1 has been proven as a target gene of several miRNAs, such as miR-193b, miR-125b-5p, miR-200b, miR-1, and miR-499. In this study, a series of experiments on miRNAs and relative miRNAs inhibitor delivered HepG2 cells were conducted, and two miRNAs that downregulate the apo(a) by targeting the 3'-UTR of Ets1 were identified. Results showed that apo(a) and Ets1 were differentially expressed in SMMC7721 and HepG2 cell lines. Meanwhile, apo(a) and Ets1 were inversely correlated with several hepatic endogenous miRNAs, such as miR-125b-5p, miR-23b-3p, miR-26a-5p, and miR-423-5p, which were predicted to bind to Ets1. Results show that miR-125b-5p and miR-23b-3p mimics could inhibit the synthesis of apo(a) by directly targeting Ets1 in HepG2, thereby reducing the plasma Lp (a) concentration.


Subject(s)
Apolipoproteins A/biosynthesis , MicroRNAs/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , 3' Untranslated Regions , Apolipoproteins A/genetics , Apolipoproteins A/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Down-Regulation , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , MicroRNAs/genetics , Proto-Oncogene Protein c-ets-1/genetics
5.
Cell Biol Int ; 40(8): 906-16, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27298021

ABSTRACT

Lipoprotein(a) [Lp(a)] is a strong genetic risk factor for coronary heart diseases. However, the metabolism of this protein remains poorly understood. Efficient and specific drugs that can decrease high plasma levels of Lp(a) have not been developed yet. Hydrogen sulfide (H2 S), a member of the gas transmitter family, performs important biological actions, including protection against cardiovascular diseases and maintenance of the lipid metabolism equilibrium in hepatocytes and adipocytes. In this study, we investigated the possible molecular mechanism of H2 S that influences apolipoprotein(a) [apo(a)] biosynthesis. We also determined the effects of H2 S on apo(a) expression and secretion in HepG2 cells as well as the underlying mechanisms. Results showed that H2 S significantly inhibited the expression and secretion levels of apo(a). These effects were attenuated by the PKCα inhibitor and FXR siRNA. H2 S also reduced HNF4α expression and enhanced FXR expression. The Akt inhibitor partially reversed H2 S-induced inhibition of apo(a) and HNF4α expression and apo(a) secretion. This study reveals that H2 S suppressed apo(a) expression and secretion via the PKCα-FXR and PI3K/Akt-HNF4α pathways.


Subject(s)
Apolipoproteins A/antagonists & inhibitors , Hepatocytes/drug effects , Hydrogen Sulfide/pharmacology , Protein Kinase C-alpha/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Apolipoproteins A/biosynthesis , Bodily Secretions/drug effects , Hep G2 Cells , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/metabolism , Humans , Lipid Metabolism , Lipoprotein(a)/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism
6.
Front Cardiovasc Med ; 9: 909178, 2022.
Article in English | MEDLINE | ID: mdl-36035922

ABSTRACT

Hydrogen sulfide (H2S), a gas transmitter found in eukaryotic organisms, plays an essential role in several physiological processes. H2S is one of the three primary biological gas transmission signaling mediators, along with nitric oxide and carbon monoxide. Several animal and in vitro experiments have indicated that H2S can prevent coronary endothelial mesenchymal transition, reduce the expression of endothelial cell adhesion molecules, and stabilize intravascular plaques, suggesting its potential role in the treatment of atherosclerosis (AS). H2S donors are compounds that can release H2S under certain circumstances. Development of highly targeted H2S donors is a key imperative as these can allow for in-depth evaluation of the anti-atherosclerotic effects of exogenous H2S. More importantly, identification of an optimal H2S donor is critical for the creation of H2S anti-atherosclerotic prodrugs. In this review, we discuss a wide range of H2S donors with anti-AS potential along with their respective transport pathways and design-related limitations. We also discuss the utilization of nano-synthetic technologies to manufacture H2S donors. This innovative and effective design example sheds new light on the production of highly targeted H2S donors.

7.
Curr Med Chem ; 28(1): 152-168, 2021.
Article in English | MEDLINE | ID: mdl-32141415

ABSTRACT

Atherosclerosis is a chronic inflammatory vascular disease. Atherosclerotic cardiovascular disease is the main cause of death in both developed and developing countries. Many pathophysiological factors, including abnormal cholesterol metabolism, vascular inflammatory response, endothelial dysfunction and vascular smooth muscle cell proliferation and apoptosis, contribute to the development of atherosclerosis and the molecular mechanisms underlying the development of atherosclerosis are not fully understood. Ubiquitination is a multistep post-translational protein modification that participates in many important cellular processes. Emerging evidence suggests that ubiquitination plays important roles in the pathogenesis of atherosclerosis in many ways, including regulation of vascular inflammation, endothelial cell and vascular smooth muscle cell function, lipid metabolism and atherosclerotic plaque stability. This review summarizes important contributions of various E3 ligases to the development of atherosclerosis. Targeting ubiquitin E3 ligases may provide a novel strategy for the prevention of the progression of atherosclerosis.


Subject(s)
Atherosclerosis/enzymology , Ubiquitin-Protein Ligases , Ubiquitination , Endothelial Cells/metabolism , Humans , Inflammation , Lipid Metabolism , Myocytes, Smooth Muscle/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
8.
Curr Med Chem ; 28(18): 3666-3680, 2021.
Article in English | MEDLINE | ID: mdl-33200693

ABSTRACT

Atherosclerosis is a chronic arterial wall illness that forms atherosclerotic plaques within the arteries. Plaque formation and endothelial dysfunction are atherosclerosis' characteristics. It is believed that the occurrence and development of atherosclerosis mainly include endothelial cell damage, lipoprotein deposition, inflammation and fibrous cap formation, but its molecular mechanism has not been elucidated. Therefore, protecting the vascular endothelium from damage is one of the key factors against atherosclerosis. The factors and processes involved in vascular endothelial injury are complex. Finding out the key factors and mechanisms of atherosclerosis caused by vascular endothelial injury is an important target for reversing and preventing atherosclerosis. Changes in cell adhesion are the early characteristics of EndMT, and cell adhesion is related to vascular endothelial injury and atherosclerosis. Recent researches have exhibited that endothelial-mesenchymal transition (EndMT) can urge atherosclerosis' progress, and it is expected that inhibition of EndMT will be an object for anti-atherosclerosis. We speculate whether inhibition of EndMT can become an effective target for reversing atherosclerosis by improving cell adhesion changes and vascular endothelial injury. Studies have shown that H2S has a strong cardiovascular protective effect. As H2S has anti- inflammatory, anti-oxidant, inhibiting foam cell formation, regulating ion channels and enhancing cell adhesion and endothelial functions, the current research on H2S in cardiovascular aspects is increasing, but anti-atherosclerosis's molecular mechanism and the function of H2S in EndMT have not been explicit. In order to explore the mechanism of H2S against atherosclerosis, to find an effective target to reverse atherosclerosis, we sum up the progress of EndMT promoting atherosclerosis, and Hydrogen sulfide's potential anti- EndMT effect is discussed in this review.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Atherosclerosis/drug therapy , Endothelial Cells , Epithelial-Mesenchymal Transition , Humans , Signal Transduction
9.
Cell Cycle ; 20(15): 1477-1486, 2021 08.
Article in English | MEDLINE | ID: mdl-34266366

ABSTRACT

Endothelial-mesenchymal transition (EndMT) is widely involved in the occurrence and development of cardiovascular diseases. Although there is no direct evidence, it is very promising as an effective target for the treatment of these diseases. Endothelial cells need to respond to the complex cardiovascular environment through EndMT, but sustained stimuli will cause the imbalance of EndMT. Blocking the signal transduction promoting EndMT is an effective method to control the imbalance of EndMT. In particular, we also discussed the potential role of endothelial cell apoptosis and autophagy in regulating the imbalance of EndMT. In addition, promoting mesenchymal-endothelial transformation (MEndT) is also a method to control the imbalance of EndMT. However, targeting EndMT to treat cardiovascular disease still faces many challenges. By reviewing the research progress of EndMT, we have put forward some insights and translated them into challenges and opportunities for new treatment strategies for cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/pathology , Endothelial Cells/pathology , Epithelial-Mesenchymal Transition , Animals , Apoptosis , Autophagy , Cardiovascular Agents/therapeutic use , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/metabolism , Cell Plasticity , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Epithelial-Mesenchymal Transition/drug effects , Humans , Phenotype , Signal Transduction
10.
DNA Cell Biol ; 39(4): 661-670, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32101022

ABSTRACT

Fibroblast growth factor 21 (FGF21) is a hormone-like member of the FGF family that is associated with cell death in atherosclerosis. However, its underlying mechanisms remain unclear. In this study, the effect of FGF21 on endothelial cell pyroptosis and its potential mechanisms were investigated. Results showed that FGF21 inhibits oxidized low-density lipoprotein (ox-LDL)-induced pyroptosis and related molecular expression in human umbilical vein endothelial cells (HUVECs). Mitochondrial function was damaged by ox-LDL and restored by FGF21. A mechanism proved that ubiquinol cytochrome c reductase core protein I (UQCRC1) was downregulated by ox-LDL and upregulated by FGF21. Further, the silencing of UQCRC1 aggravated HUVEC pyroptosis and impaired mitochondrial function and reactive oxygen species (ROS) production. Moreover, Tet methylcytosine dioxygenase (TET2) was involved in the regulation of UQCRC1 expression and pyroptosis. In summary, FGF21 inhibited ox-LDL-induced HUVEC pyroptosis through the TET2-UQCRC1-ROS pathway.


Subject(s)
Electron Transport Complex III/metabolism , Fibroblast Growth Factors/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Lipoproteins, LDL/metabolism , Pyroptosis/physiology , Atherosclerosis/pathology , Cell Survival , Cells, Cultured , DNA-Binding Proteins/metabolism , Dioxygenases , Electron Transport Complex III/genetics , Fibroblast Growth Factors/genetics , Humans , Membrane Potential, Mitochondrial/physiology , Mitochondria/metabolism , Oxidative Stress/physiology , Proto-Oncogene Proteins/metabolism , RNA Interference , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Signal Transduction
11.
Clin Chim Acta ; 501: 142-146, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31730809

ABSTRACT

Atherosclerosis is a chronic inflammatory response that increases the risk of cardiovascular diseases. An in-depth study of the pathogenesis of atherosclerosis is critical for the treatment of atherosclerotic cardiovascular disease. The development of atherosclerosis involves many cells, such as endothelial cells, vascular smooth muscle cells, macrophages, and others. The considerable effects of macrophages in atherosclerosis are inextricably linked to macrophage polarization and the resulting phenotype. Moreover, the significant impact of macrophages on atherosclerosis depend not only on the function of the different macrophage phenotypes but also on the relative ratio of different phenotypes in the plaque. Research on atherosclerosis therapy indicates that the reduced plaque size and enhanced stability are partly due to modulating macrophage polarization. Therefore, regulating macrophage polarization and changing the proportion of macrophage phenotypes in plaques is a new therapeutic approach for atherosclerosis. This review provides a new perspective for atherosclerosis therapy by summarizing the relationship between macrophage polarization and atherosclerosis, as well as treatment targeting macrophage polarization.


Subject(s)
Atherosclerosis/metabolism , Macrophages/metabolism , Animals , Humans , Macrophage Activation
12.
Atherosclerosis ; 268: 170-176, 2018 01.
Article in English | MEDLINE | ID: mdl-29232562

ABSTRACT

Transmembrane 6 superfamily 2 (TM6SF2), a gene identified at the locus 19p12, has been recognized to regulate plasma lipids. Here, we provide an overview of the roles of TM6SF2 as a novel target for plasma lipid regulation. We first review the association of TM6SF2 variant with plasma lipid traits, cardiovascular disease (CVD) and non-alcoholic fatty liver disease (NAFLD). Then, we present an overview about the in vivo validation of TM6SF2 as a regulator of plasma lipid levels using mice, with overexpression or knockdown/knockout of TM6SF2. Thereafter, we discuss the mechanisms underlying TM6SF2 regulation of lipid metabolism involving intestinal cholesterol absorption and hepatic cholesterol biosynthesis and transport. In conclusion, increasing evidence suggests that TM6SF2 may be a major regulator of plasma lipid levels and become a therapeutic target in cardiovascular disease.


Subject(s)
Cholesterol/blood , Dyslipidemias/blood , Intestinal Absorption , Liver/metabolism , Membrane Proteins/metabolism , Animals , Anticholesteremic Agents/therapeutic use , Biomarkers/blood , Dyslipidemias/diagnosis , Dyslipidemias/drug therapy , Dyslipidemias/genetics , Humans , Intestinal Absorption/drug effects , Liver/drug effects , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Mice, Transgenic
13.
Sheng Li Xue Bao ; 59(6): 831-9, 2007 Dec 25.
Article in Zh | MEDLINE | ID: mdl-18157478

ABSTRACT

To test the hypothesis that concentration polarization of atherogenic lipids may occur in the arterial system and play an important role in localization of atherosclerosis, we simulated and measured in vitro the luminal surface concentration of low density lipoprotein (LDL) in local stenosis at the distal end of carotid artery by number simulation and laser scanning confocal microscopy, then we designed carotid stenosis model to test the role of LDL concentration polarization in atherogenesis. The in vitro experiment showed that the luminal surface LDL concentration was higher than the bulk concentration as predicted by the concentration polarization theory. The relative luminal surface LDL concentration changed with the flow velocity and ratio of stenosis. The wall concentration of LDL was highest in the round tube with 40% stenosis at the same velocity, while the wall concentration of LDL was higher when Re was 250 than Re was 500 at the same extent of narrowness. The animal experiment also revealed that general atherogenic plaques obviously occurred at the distal end of local stenosis where concentration polarized. The results strongly support our hypothesis that concentration polarization of lipoproteins occurs in local stenosis at the distal end of carotid artery, and this in turn promotes the localization of atherosclerosis which develops in the arterial system.


Subject(s)
Atherosclerosis/physiopathology , Carotid Stenosis/physiopathology , Lipoproteins, LDL/metabolism , Animals , Disease Models, Animal
14.
Int J Biol Macromol ; 98: 637-645, 2017 May.
Article in English | MEDLINE | ID: mdl-28192139

ABSTRACT

Lipoprotein(a)[Lp(a)] is a risk factor for coronary heart diseases. However, the metabolism of this protein remains poorly understood. Efficient and specific drugs that can decrease high plasma levels of Lp(a) have not been developed yet. Vitamin C is responsible for maintaining the catalytic activity of a group of iron and 2-oxoglutarate (2OG)-dependent dioxygenases and induces the generation of 5-hydroxymethylcytosine (5hmC) via Ten-eleven translocation (Tet) dioxygenases. In addition, It has been reported vitamin C deficiency induces atherosclerosis and increases Lp(a) and apo(a) plasma levels in Lp(a)+ mice. However, the mechanism is still unclear. In this study, we investigated the effects of vitamin C on apo(a) expression and the possible molecular mechanism of vitamin C that influences apolipoprotein(a) [apo(a)] biosynthesis in HepG2 cells. Results showed that vitamin C significantly inhibited the expression and secretion levels of apo(a). Vitamin C can also increase ELK1 expression and hydroxymethylation of ELK1 promoter and the globle DNA in HepG2 cells. In addition, the effects of vitamin C inhibiting the apo(a) expression were attenuated by ELK1siRNA and Tet2siRNA. These results suggested vitamin C down-regulate apo(a) expression via Tet2-dependent DNA demethylation in HepG2 cells.


Subject(s)
Apoprotein(a)/genetics , Ascorbic Acid/administration & dosage , DNA-Binding Proteins/genetics , Proto-Oncogene Proteins/genetics , ets-Domain Protein Elk-1/genetics , Animals , Apoprotein(a)/biosynthesis , DNA Methylation/drug effects , DNA-Binding Proteins/biosynthesis , Dioxygenases , Gene Expression Regulation/drug effects , Hep G2 Cells , Humans , Iron/blood , Mice , Promoter Regions, Genetic/drug effects , Proto-Oncogene Proteins/biosynthesis , ets-Domain Protein Elk-1/biosynthesis
15.
Front Pharmacol ; 8: 486, 2017.
Article in English | MEDLINE | ID: mdl-28798687

ABSTRACT

Ten-eleven translocation-2 (TET2) protein is a DNA demethylase that regulates gene expression through DNA demethylation and also plays important roles in various diseases including atherosclerosis. Endothelial dysfunction represents an early key event in atherosclerotic disease. The cystathionine-γ-lyase (CSE)/hydrogen sulfide (H2S) is a key endogenous system with protective effects on endothelial functions. In this study, we examined how TET2 regulates oxidized low-density lipoprotein (oxLDL)-induced dysfunction of human umbilical vein endothelial cells (HUVECs) and determined the role of the CSE/H2S system. Treatment with oxLDL resulted in downregulation of both TET2 expression and CSE/H2S system in HUVECs. TET2 was found to have protective effects on oxLDL-induced HUVEC dysfunction, which was confirmed with TET2 overexpression plasmid or TET2 shRNA plasmid. Moreover, TET2 was found to upregulate the CSE/H2S system and inhibit NF-κB activation, leading to decreased expression of ICAM-1 and VCAM-1 and attenuated adhesion of THP-1 cells to oxLDL-activated HUVECs. The protective effect of TET2 was reduced by treatment with CSE siRNA. Further studies revealed that CSE promoter region contains a well-defined CpG island. We also showed that TET2 enhanced 5-hydroxymethylcytosine (5hmC) level and promoted DNA demethylation of CSE gene promoter, leading to an increase in CSE expression. In conclusion, TET2 has protective effects on oxLDL-induced HUVEC dysfunction, likely through upregulating the CSE/H2S system by DNA demethylation of CSE gene promoter. TET2 may become a novel therapeutic target for endothelial dysfunction-associated vascular diseases.

16.
Atherosclerosis ; 262: 113-122, 2017 07.
Article in English | MEDLINE | ID: mdl-28535426

ABSTRACT

BACKGROUND AND AIMS: Proprotein convertase subtilisin/kexin 9 (PCSK9) has emerged as a popular target in the development of new cholesterol-lowering drugs and therapeutic interventions for atherosclerosis. PCSK9 could accelerate atherosclerosis through mechanisms beyond the degradation of the hepatic low-density lipoprotein receptor. Several clinical studies suggested that PCSK9 is involved in atherosclerotic inflammation. Accordingly, this study aimed to explore the role of PCSK9 in vascular inflammation that promotes atherosclerotic progression. METHODS: We examined whether PCSK9 silencing via transduction with the lentivirus-mediated PCSK9 shRNA (LV-PCSK9 shRNA) vector affects the formation of vascular lesions in hyperlipidemia-induced atherosclerosis in apolipoprotein E knockout (apoE KO) mice. In vitro, the effects of PCSK9 on oxLDL-induced macrophages inflammation were investigate using LV-PCSK9 and LV-PCSK9 shRNA for PCSK9 overexpression and PCSK9 silencing. RESULTS: Immunohistochemical analysis showed that PCSK9 expression increased within atherosclerotic plaques in apoE KO mice. These in vivo results showed that the LV-PCSK9 shRNA group of mice developed less aortic atherosclerotic plaques compared with the control group. These lesions also had the reduced number of macrophages and decreased expression of vascular inflammation regulators, such as tumor necrosis factor-α, interleukin 1 beta, monocyte chemoattractant protein-1, toll-like receptor 4 and nuclear factor kappa B (NF-κB). We further showed that PCSK9 overexpression in macrophages in vitro increased the secretion of oxLDL-induced proinflammatory cytokines. PCSK9 overexpression upregulated TLR4 expression and increased p-IκBα levels, IkBα degradation, and NF-κB nuclear translocation in macrophages, but PCSK9 knockdown had the opposite effects in oxLDL-treated macrophages. CONCLUSIONS: PCSK9 gene interference could suppress atherosclerosis directly through decreasing vascular inflammation and inhibiting the TLR4/NF-κB signaling pathway without affecting plasma cholesterol level in high-fat diet-fed apoE KO mice. PCSK9 may be an inflammatory mediator in the pathogenesis of atherosclerosis.


Subject(s)
Aorta/enzymology , Aortic Diseases/enzymology , Atherosclerosis/enzymology , Inflammation Mediators/metabolism , Inflammation/enzymology , NF-kappa B/metabolism , Proprotein Convertase 9/metabolism , Toll-Like Receptor 4/metabolism , Animals , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Disease Models, Animal , Genetic Predisposition to Disease , Inflammation/genetics , Inflammation/pathology , Lipoproteins, LDL/pharmacology , Macrophages/drug effects , Macrophages/enzymology , Male , Mice , Mice, Knockout, ApoE , Phenotype , Plaque, Atherosclerotic , Proprotein Convertase 9/genetics , RAW 264.7 Cells , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
17.
Oncotarget ; 7(47): 76423-76436, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27821816

ABSTRACT

Tet methylcytosine dioxygenase 2 (TET2) mediates the conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC). The loss of TET2 is associated with advanced atherosclerotic lesions. Our previous study showed that TET2 improves endothelial cell function by enhancing endothelial cell autophagy. Accordingly, this study determined the role of TET2 in atherosclerosis and potential mechanisms. In ApoE-/- mice fed high-fat diet, TET2 overexpression markedly decreased atherosclerotic lesions with uniformly increased level of 5hmC and decreased level of 5mC in genomic DNA. TET2 overexpression also promoted autophagy and downregulated inflammation factors, such as vascular cell adhesion molecule 1, intercellular adhesion molecule 1, monocyte chemotactic protein 1, and interleukin-1. Consistently, TET2 knockdown with small hairpin RNA (shRNA) in ApoE-/- mice decreased 5hmC and increased 5mC levels in atherosclerotic lesions. Meanwhile, autophagy was inhibited and atherosclerotic lesions progressed with an unstable lesion phenotype characterized by large lipid core, macrophage accumulation, and upregulated inflammation factor expression. Experiments with the cultured endothelial cells revealed that oxidized low-density lipoprotein (ox-LDL) inhibited endothelial cell autophagy. TET2 shRNA strengthened impaired autophagy and autophagic flux in the ox-LDL-treated endothelial cells. TET2 overexpression reversed these effects by decreasing the methylation level of the Beclin 1 promoter, which contributed to the downregulation of inflammation factors. Overall, we identified that TET2 was downregulated during the pathogenesis of atherosclerosis. The downregulation of TET2 promotes the methylation of the Beclin 1 promoter, leading to endothelial cell autophagy, impaired autophagic flux, and inflammatory factor upregulation. Upregulation of TET2 may be a novel therapeutic strategy for treating atherosclerosis.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Autophagy/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , 5-Methylcytosine/analogs & derivatives , Animals , Apolipoproteins E/deficiency , Atherosclerosis/pathology , Cytokines/metabolism , DNA Methylation , Dioxygenases , Disease Models, Animal , Gene Expression , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunohistochemistry , Inflammation Mediators/metabolism , Lipoproteins, LDL/metabolism , Male , Mice , Mice, Knockout , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology
18.
Zhongguo Yi Liao Qi Xie Za Zhi ; 29(6): 439-42, 2005 Nov.
Article in Zh | MEDLINE | ID: mdl-16494058

ABSTRACT

This review is a summary of some useful methods and advances about improving clinical applications to small-diameter vascular grafts in recent years, and it points out the developing orientation of small-diameter vascular grafts in the future.


Subject(s)
Bioartificial Organs , Vascular Grafting , Endothelial Cells , Endothelium, Vascular , Tissue Engineering
19.
Atherosclerosis ; 243(1): 223-35, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26407666

ABSTRACT

Oxidised lipoprotein(a) [oxLp(a)] is considered as a more potent arteriosclerotic factor than native Lp(a). However, the molecular mechanisms underlying this potency remain unclear. Reactive oxygen species (ROS) possibly act as intracellular second messengers that participate in autophagy stimulation. In this study, the effect of oxLp(a) on endothelial cell autophagy was determined. The mechanism and effect of autophagy on endothelial cells were also investigated. Results showed that oxLp(a) could induce autophagy depending on the generation of cellular ROS. Superoxide dismutase, an antioxidant, could inhibit oxLp(a)-induced autophagy in human umbilical vascular endothelial cells. Furthermore, poly(adenosine diphosphate-ribose) polymerase-1 (PARP-1)-liver kinase B1 (LKB1)-adenosine monophosphate-activated protein kinase (AMPK)-mammalian target of rapamycin (mTOR) and LKB1-AMPK-mTOR pathways are involved in oxLp(a)-induced autophagy. These pathways are also dependent on ROS. Thus, oxLp(a) induced autophagy via LKB1-AMPK-mTOR and PAPR-1-LKB1-AMPK-mTOR pathways, which are dependent on ROS generation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Lipoprotein(a)/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Protein Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Antioxidants/metabolism , Apoptosis , Arteriosclerosis/physiopathology , Autophagy , Green Fluorescent Proteins/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Microscopy, Electron, Transmission , Poly (ADP-Ribose) Polymerase-1 , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Superoxide Dismutase/metabolism
20.
Mol Med Rep ; 7(6): 1865-70, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23588928

ABSTRACT

The aim of the present study was to investigate the attenuation of endothelial cell senescence by H2S and to explore the mechanisms underlying the anti-aging effects of H2S. Senescence was induced in human umbilical vein endothelial cells (HUVECs) by incubation in 25 µmol/l H2O2 for 1 h. Senescence-associated ß-galactosidase (SA-ß-gal) activity was examined to determine the effects of H2S on senescent HUVECs. The results indicated that SA-ß-gal activity in the H2O2-treated HUVECs was 11.2 ± 1.06%, which was attenuated in the NaHS group. Pretreatment with nicotinamide (NAM), a sirtuin 1 (SIRT1) inhibitor, inhibited the reduction in senescence associated with H2S. Immunoblot analyses revealed that SIRT1 levels in senescent HUVECs treated with NaHS (60 µM) were indistinguishable from controls; however, analyses of SIRT1 activity indicated that SIRT1 enzyme activity was enhanced. In addition, we found that H2S improves the function of senescent HUVECs. The present study demonstrated that H2S protects against HUVEC senescence, potentially through modulation of SIRT1 activity. Furthermore, this study establishes a novel endothelial protective effect of H2S.


Subject(s)
Cellular Senescence/drug effects , Hydrogen Peroxide/toxicity , Hydrogen Sulfide/pharmacology , Sirtuin 1/metabolism , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Human Umbilical Vein Endothelial Cells , Humans , Niacinamide/pharmacology , Sirtuin 1/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL