Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Publication year range
1.
Prostaglandins Other Lipid Mediat ; 172: 106832, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38460759

ABSTRACT

Atherosclerosis (AS) represents a prevalent initiating factor for cardiovascular events. Insulin-like growth factor 2 mRNA binding protein 3 (IGF2BP3) is an oncofetal RNA-binding protein that participates in cardiovascular diseases. This work aimed to elaborate the effects of IGF2BP3 on AS and the probable mechanism by using an oxidized low-density lipoprotein (ox-LDL)-induced human umbilical vein endothelial cells (HUVECs) model. Results indicated that IGF2BP3 expression was declined in the blood of AS patients and ox-LDL-induced HUVECs. IGF2BP3 elevation alleviated ox-LDL-provoked viability loss, apoptosis, oxidative DNA damage and endothelial dysfunction in HUVECs. Moreover, IGF2BP3 bound SESN1 and stabilized SESN1 mRNA. Furthermore, SESN1 interference reversed the impacts of IGF2BP3 overexpression on the apoptosis, oxidative DNA damage and endothelial dysfunction of ox-LDL-challenged HUVECs. Additionally, the activation of Nrf2 signaling mediated by IGF2BP3 up-regulation in ox-LDL-treated HUVECs was blocked by SESN1 absence. Collectively, SESN1 stabilized by IGF2BP3 might protect against AS by activating Nrf2 signaling.


Subject(s)
Human Umbilical Vein Endothelial Cells , Lipoproteins, LDL , NF-E2-Related Factor 2 , Oxidative Stress , RNA, Messenger , RNA-Binding Proteins , Signal Transduction , Humans , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Lipoproteins, LDL/pharmacology , Lipoproteins, LDL/metabolism , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Oxidative Stress/drug effects , Signal Transduction/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Apoptosis/drug effects , Atherosclerosis/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , RNA Stability/drug effects , DNA Damage , Sestrins
2.
J Mol Histol ; 55(1): 109-120, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38165567

ABSTRACT

Endothelial cells are a crucial component of the vessel-tissue wall and exert an important role in atherosclerosis (AS). To explore the role of Orientin in AS, human vascular endothelial cells (HUVECs) were induced by oxidized low-density lipoprotein (ox-LDL) to simulate the vascular endothelial injury during AS. Cell viability was detected by CCK-8 assay. Oxidative stress and inflammation related markers were measured using kits, RT-qPCR or western blot. Besides, cell apoptosis was assessed with TUNEL staining and cell autophagy was evaluated by LC3 immunofluorescent staining. Additionally, western blot was utilized to evaluate the expression of Sestrin 1 (SESN1) and proteins in AMPK/mTOR signaling. Afterwards, SESN1 was silenced to determine the expression of autophagy-related proteins. The further application of autophagy inhibitor 3-methyladenine (3-MA) was used to clarify the regulatory mechanism of Orientin on autophagy. Results showed that the decreased viability of HUVECs caused by ox-LDL induction was elevated by Orientin. Oxidative stress and inflammation were also attenuated after Orientin addition in HUVECs under ox-LDL condition. Moreover, Orientin suppressed apoptosis and induced autophagy of HUVECs stimulated by ox-LDL, accompanied by enhanced level of phospho (p)-AMPK and declined level of p-mTOR. Interestingly, SESN1 level was elevated by Orientin, and SESN1 depletion alleviated autophagy and reduced p-AMPK expression but enhanced p-mTOR expression. The further experiments indicated that SESN1 silencing or 3-MA addition reversed the inhibitory effects of Orientin on the oxidative stress, inflammation and apoptosis of HUVECs. Collectively, Orientin could induce autophagy by activating SESN1 expression, thereby regulating AMPK/mTOR signaling in ox-LDL-induced HUVECs.


Subject(s)
AMP-Activated Protein Kinases , Flavonoids , Glucosides , Sestrins , Humans , Sestrins/metabolism , Human Umbilical Vein Endothelial Cells , AMP-Activated Protein Kinases/metabolism , Oxidative Stress , Lipoproteins, LDL/pharmacology , TOR Serine-Threonine Kinases/metabolism , Apoptosis , Autophagy , Inflammation/metabolism
3.
Aging (Albany NY) ; 15(11): 5052-5065, 2023 06 08.
Article in English | MEDLINE | ID: mdl-37294547

ABSTRACT

BACKGROUND: Atherosclerosis (AS) is a disease characterized by the disorder of lipid metabolism and the formation of atherosclerotic plaques in the arterial wall, leading to arterial stenosis. Sestrins 1 (SESN1) plays an important regulatory role in AS, but the specific regulatory mechanism is still unclear. METHODS: ApoE-/- mouse models of AS were constructed. After overexpressing SESN1, oil red O staining was used to detect the degree of aortic plaque. HE staining detected the endothelial damage of the surrounding tissues. ELISA was used to detect the levels of vascular inflammation and oxidative stress. The iron metabolism in vascular tissues was detected by immunofluorescence. The expressions of SESN1 and ferroptosis-related proteins were detected by western blot. In the oxidized low-density lipoprotein (ox-LDL)-induced injury model in human umbilical vein endothelial cells (HUVECs), CCK8, ELISA, immunofluorescence and western blot were respectively used to detect cell viability, inflammatory response, oxidative stress and ferroptosis. The regulatory mechanism of SESN1 on endothelial ferroptosis in AS was further explored following the addition of P21 inhibitor UC2288. RESULTS: Overexpression of SESN1 could inhibit the extent of the plaque and reduce the endothelial injury of plaque tissues in AS mice. In both mouse and cell models of AS, SESN1 overexpression inhibited inflammatory response, oxidative stress response, and endothelial ferroptosis. The inhibitory effect of SESN1 on endothelial ferroptosis might be achieved through activation of P21. CONCLUSION: SESN1 overexpression plays an inhibitory role in vascular endothelial ferroptosis through the activation of P21 in AS.


Subject(s)
Atherosclerosis , Ferroptosis , Plaque, Atherosclerotic , Humans , Animals , Mice , Sestrins/metabolism , Atherosclerosis/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Plaque, Atherosclerotic/metabolism , Oxidative Stress , Apoptosis
4.
Bioengineered ; 13(4): 10786-10802, 2022 04.
Article in English | MEDLINE | ID: mdl-35485136

ABSTRACT

Myocardial infarction (MI) is the leading cause of sudden death. Long non-doing RNAs (lncRNAs) were demonstrated to play crucial roles in multiple diseases, including cancer and cardiovascular diseases. Nevertheless, the molecular mechanism of lncNRAs in MI is unclear. In this study, we integrated bioinformatics and molecular biological experiments to identify the novel lncRNA transcripts and elucidated its regulatory mechanism in MI. First, we identified 10 dysregualted lncRNAs and found that lncRNA Gm47283 was the top risk factor in MI. Bioinformatics analysis predicted that lncRNA Gm47283 exerted function via targeting miR-706 and Ptgs2. Ptgs2 was also the known regulator of ferroptosis. Inhibition or overexpression of lncRNA Gm47283 could regulate Ptgs2 expression and downstream ferroptosis activity. Overexpression of miR-706 could inhibit the expression of Ptgs2 and the activity of ferroptosis, thereby attenuated cellular injury. Mechanically, co-transfection experiments showed that overexpression of miR-706 could reverse the damage effect that was caused by lncRNA Gm47283 overexpression, via inhibiting Ptgs2 and ferroptosis. Additionally, inhibition of lncRNA Gm47283 by stem cell membrane coated siRNA could attenuate MI in vivo. Our study elucidated a novel mechanism containing lncRNA Gm47283/miR-706/Ptgs2/ferroptosis in MI, which provided a potential therapeutic for MI.Graphical Abstract. Stem cell membrane coated siRNA of lncRNA Gm47283 inhibits cardiomyocyte ferroptosis in myocardial infarction rat. Stem cell membrane-coated siRNA of lncRNA Gm47283 increases miR-706, and then miR-706 suppresses the expression of Ptgs2 to reduce lipid peroxidation toxicity, and then inhibits cardiomyocyte ferroptosis. PUFA: polyunsaturated fatty acid.


Subject(s)
Ferroptosis , MicroRNAs , Myocardial Infarction , RNA, Long Noncoding , Animals , Cyclooxygenase 2/genetics , Ferroptosis/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Small Interfering , Rats
5.
J Biomater Appl ; 37(2): 303-314, 2022 08.
Article in English | MEDLINE | ID: mdl-35403475

ABSTRACT

Myocardial ischemia-reperfusion injury (MI/RI) refers to the clinical state of decreased coronary blood flow caused by various causes. The main pathogenesis of MI/RI is mitochondrial oxidative damage. In this study, we designed a novel mitochondrial targeted astaxanthin (AST) liposome, namely, STPP-AST-LIP, targeting mitochondria of H9c2 myocardial cells. STPP-AST-LIP not only reduced the production of mitochondrial reactive oxygen species (ROS), but also increased the survival rate of MI/RI H9c2 cells. In addition, rat experiments further confirmed that STPP-AST-LIP could improve myocardial cardiac function in MI/RI rats, significantly inhibited apoptosis of myocardial cells, and had a protective effect on the heart of rats after MI/RI.


Subject(s)
Myocardial Reperfusion Injury , Animals , Apoptosis , Liposomes/metabolism , Mitochondria/metabolism , Mitochondria/pathology , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidative Stress , Rats , Reactive Oxygen Species/metabolism , Xanthophylls
6.
Mol Med Rep ; 25(5)2022 May.
Article in English | MEDLINE | ID: mdl-35293601

ABSTRACT

Endothelial cells are an important component of the heart and vasculature and form a crucial link between the cardiovascular system and the immune system. Sestrin 1 (SESN1) has an important role in atherosclerosis by inhibiting NOD­like receptor family pyrin domain containing 3 inflammasome activation. However, whether SESN1 is involved in human umbilical vein endothelial cell (HUVEC) injury caused by atherosclerosis has remained to be elucidated. The present study aimed to investigate the functions of SESN1 in the inflammatory response, apoptosis and endothelial­mesenchymal transition (EndMT) of HUVECs following stimulation with oxidized low­density lipoprotein (Ox­LDL). SESN1 expression at the mRNA and protein levels was detected using reverse transcription­quantitative PCR (RT­qPCR) and western blot analysis. Following SESN1 overexpression in Ox­LDL­stimulated HUVECs, cell viability was determined using a Cell Counting Kit­8 assay. Terminal deoxynucleotidyl transferase­mediated nick­end labeling staining was employed to detect cell apoptosis and western blot analysis was used to determine the levels of apoptosis­related proteins. RT­qPCR, ELISA and western blot were utilized to determine the levels of inflammatory factors. Immunofluorescence staining, RT­qPCR and western blot analysis were employed to assess the EndMT of Ox­LDL­stimulated HUVECs. The results revealed that SESN1 exhibited a low expression in HUVECs following Ox­LDL stimulation. SESN1 overexpression suppressed inflammation, apoptosis and EndMT in Ox­LDL­induced HUVECs. In addition, SESN1 stimulated adenosine monophosphate­activated protein kinase catalytic subunit α1/sirtuin 1 signaling to suppress Ox­LDL receptor­1 expression. An AMPK and SIRT1 inhibitor reversed the effects of SESN1 overexpression on the inflammatory response, apoptosis and EndMT of HUVECs exposed to Ox­LDL. Taken together, the present study demonstrated that SENS1 exerts a suppressive effect on Ox­LDL­induced inflammation, apoptosis and EndMT of HUVECs, suggesting that SENS1 may be used as a novel biomarker for endothelial injury­related disorders.


Subject(s)
AMP-Activated Protein Kinases , Human Umbilical Vein Endothelial Cells , Sestrins , Sirtuin 1 , AMP-Activated Protein Kinases/metabolism , Apoptosis , Heat-Shock Proteins/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/metabolism , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Scavenger Receptors, Class E/metabolism , Sestrins/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism
7.
Bioengineered ; 13(2): 2917-2926, 2022 02.
Article in English | MEDLINE | ID: mdl-35043753

ABSTRACT

Transcription factor forkhead box protein 1 (FOXP1) has been shown cardiovascular protection. We aimed to analyze the role of FOXP1 in oxidized low-density lipoprotein (ox-LDL)-induced macrophages and its possible regulatory effect on sestrin1 (SESN1) expression. After stimulation with ox-LDL, FOXP1 expression in RAW264.7 cells was evaluated with RT-qPCR and Western blotting. Then, FOXP1 was overexpressed, followed by detection of inflammatory mediator levels using ELISA kits and RT-qPCR. Lipid accumulation was detected with oil red O staining. Additionally, the JASPAR database was used to predict the potential genes that could be transcriptionally regulated by FOXP1. ChIP and luciferase reporter assays were used to verify this combination. To further clarify the regulatory effects of FOXP1 on SESN1 in damage of macrophages triggered by ox-LDL, SESN1 was silenced to determine the inflammation and lipid accumulation under the condition of FOXP1 overexpression. Results indicated that ox-LDL stimulation led to a significant decrease in FOXP1 expression. FOXP1 overexpression notably reduced the levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1ß and IL-6, accompanied by a decreased in phosphorylated NF-κB p65 expression. Besides, FOXP1-upregulation inhibited lipid accumulation and reduced CD36 expression level in RAW264.7 cells upon ox-LDL stimulation. Moreover, results of ChIP and luciferase reporter assays suggested that FOXP1 could transcriptionally regulate SESN1 expression. Further experiments supported that SESN1 silencing restored the inhibitory effects of FOXP1 overexpression on the inflammation and lipid accumulation in RAW264.7 cells exposed to ox-LDL. Collectively, FOXP1 transcriptionally activates SESN1 for the alleviation of ox-LDL-induced inflammation and lipid accumulation in macrophages.


Subject(s)
Cell Cycle Proteins/genetics , Forkhead Transcription Factors/physiology , Inflammation/genetics , Lipid Metabolism/genetics , Macrophages/metabolism , Repressor Proteins/physiology , Animals , Cell Cycle Proteins/metabolism , Foam Cells/metabolism , Foam Cells/pathology , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Lipoproteins, LDL , Mice , RAW 264.7 Cells , Transcriptional Activation/genetics
8.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-735314

ABSTRACT

@#Objective    To explore the relationship between obstructive sleep apnea-hypopnea syndrome (OSAHS) and aortic dissection (AD). Methods    Fifty three patients with AD diagnosed by CTA in our hospital from January 2016 to January 2018 were selected. All the patients with AD were scored by the STOP-BANG questionnaire. The patients who scored more than or equal to 3 received polysomnography (PSG) after surgical or conservative treatment, and according to whether the sleep apnea-hypopnea index was higher than or equal to 5. Fifty-three patients were divided into an OSAHS group and a non OSAHS group. Results    There were 18 patients with 17 males and 1 female at average age of 43.3±8.4 years in the OSAHS group, and 35 patients with 23 males and 12 females at average age of 56.6±12.9 years in the non OSAHS group. There was no statistical difference between the two groups in the Stanford classification of aortic dissection, the time of onset, personal history, the history of diabetes, coronary heart disease and hyperlipidemia, or post-treatment systolic/diastolic blood pressure before sleep (P>0.05). The age of patients in the OSAHS group was significantly less than that in the non OSAHS group (P<0.01), the proportion of men/women (P=0.021), weight (P<0.01), height (P=0.028), body mass index (P<0.01), and post-treatment systolic/diastolic blood pressure after waking up (P=0.028,P=0.044) in the OSAHS group were significantly higher than those in the non OSAHS group. In the OSAHS group, the proportion of previous hypertension was significantly higher than that in the non OSAHS group (P=0.042).   Conclusion    AD patients combined with OSAHS are mostly male patients. The number of young and high-fat people is significantly more than that in the non OSAHS group. OSAHS may be one of the risk factors for young, high-fat men with AD.

SELECTION OF CITATIONS
SEARCH DETAIL
...