Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Front Psychol ; 15: 1291353, 2024.
Article in English | MEDLINE | ID: mdl-38495417

ABSTRACT

Hot Executive Function (hot EF) refers to cognitive process involved in high emotion or motivation, and the operation of this function is related to the activities of the ventromedial prefrontal lobe and orbitofrontal lobe. Meanwhile, rhythmic-movement activity is a musical activity in which one expresses and feels music with one's own body movements which involves cognitive abilities such as adjusting and understanding emotions among children. To explore how rhythmic-movement activity with rewards influences the development of hot EF in children of 5-6 years old, the organization principles of rhythmic-movement activity with rewards intervention on hot EF were designed, and 62 children of 5-6 years old in a kindergarten in Yantai of China were selected as research participants (M = 5.80 years old, SD = 0.37 years old) for pre-test and post-test experimental design. The experimental group received rhythmic-movement activity with rewards three times a week for 6 weeks, while the control group did not. The gift delay task and the children's gambling task were used to measure two sub-components of hot EF before and after the intervention, and the results show that rhythmic-movement activity with rewards has a significant effect on gratification delay and affective decision-making ability of children. Finally, the effects and enlightenment of rhythmic-movement activity with rewards on hot EF are discussed.

2.
Front Oncol ; 13: 1174530, 2023.
Article in English | MEDLINE | ID: mdl-37534258

ABSTRACT

Purpose: To introduce a model for automatic segmentation of thoracic organs at risk (OARs), especially the esophagus, in non-small cell lung cancer radiotherapy, using a novel two-step deep learning network. Materials and methods: A total of 59 lung cancer patients' CT images were enrolled, of which 39 patients were randomly selected as the training set, 8 patients as the validation set, and 12 patients as the testing set. The automatic segmentations of the six OARs including the esophagus were carried out. In addition, two sets of treatment plans were made on the basis of the manually delineated tumor and OARs (Plan1) as well as the manually delineated tumor and the automatically delineated OARs (Plan2). The Dice similarity coefficient (DSC), 95% Hausdorff distance (HD95), and average surface distance (ASD) of the proposed model were compared with those of U-Net as a benchmark. Next, two groups of plans were also compared according to the dose-volume histogram parameters. Results: The DSC, HD95, and ASD of the proposed model were better than those of U-Net, while the two groups of plans were almost the same. The highest mean DSC of the proposed method was 0.94 for the left lung, and the lowest HD95 and ASD were 3.78 and 1.16 mm for the trachea, respectively. Moreover, the DSC reached 0.73 for the esophagus. Conclusions: The two-step segmentation method can accurately segment the OARs of lung cancer. The mean DSC of the esophagus realized preliminary clinical significance (>0.70). Choosing different deep learning networks based on different characteristics of organs offers a new option for automatic segmentation in radiotherapy.

3.
Acta Biochim Biophys Sin (Shanghai) ; 55(8): 1184-1192, 2023 Apr 06.
Article in English | MEDLINE | ID: mdl-37021975

ABSTRACT

Autophagy plays a critical role in the physiology and pathophysiology of hepatocytes. High level of homocysteine (Hcy) promotes autophagy in hepatocytes, but the underlying mechanism is still unknown. Here, we investigate the relationship between Hcy-induced autophagy level and the expression of nuclear transcription factor EB (TFEB). The results show that Hcy-induced autophagy level is mediated by upregulation of TFEB. Silencing of TFEB decreases the level of autophagy-related protein LC3BII/I and increases p62 expression level in hepatocytes after exposure to Hcy. Moreover, the effect of Hcy on the expression of TFEB is regulated by hypomethylation of the TFEB promoter catalyzed by DNA methyltransferase 3b (DNMT3b). In summary, this study shows that Hcy can activate autophagy by inhibiting DNMT3b-mediated DNA methylation and upregulating TFEB expression. These findings provide another new mechanism for Hcy-induced autophagy in hepatocytes.


Subject(s)
Autophagy , DNA Methylation , Hepatocytes , Homocysteine , Autophagy/genetics , DNA , Homocysteine/metabolism , Homocysteine/pharmacology , Humans , DNA Methyltransferase 3B
4.
Biomed Pharmacother ; 161: 114537, 2023 May.
Article in English | MEDLINE | ID: mdl-36933378

ABSTRACT

Silicosis is a devastating interstitial lung disease characterized by silicon nodules and diffuse pulmonary fibrosis. To date, inefficient therapy is still a challenge of this disease due to its complicated pathogenesis. Hepatocyte growth factor (HGF) which is highly expressed in hepatocyte with anti-fibrotic and anti-apoptotic function was downregulated in silicosis. In addition, the upregulation of transforming growth factor-beta (TGF-ß), another pathological molecular was observed to aggravate the severity and accelerate the progression of silicosis. Here AAV expressed HGF with targeting pulmonary capillaries and SB431542, the inhibitor of TGF-ß signal pathway, were simultaneously adopted to synergistically reduce silicosis fibrosis. In vivo result demonstrated that the cooperation of HGF with SB431542 showed strong anti-fibrosis effects on the silicosis mice via tracheal administration of silica, compared to the separate treatment. The high efficacy was mainly achieved by remarkably by reducing ferroptosis of lung tissue. In our point, the combination of AAV9-HGF with SB431542 provide an alternative to relieve silicosis fibrosis from the perspective of targeting pulmonary capillaries.


Subject(s)
Ferroptosis , Silicosis , Mice , Animals , Transforming Growth Factor beta/metabolism , Hepatocyte Growth Factor , Transforming Growth Factor beta1/metabolism , Fibrosis , Silicosis/drug therapy , Silicosis/metabolism
5.
J Inflamm Res ; 16: 505-521, 2023.
Article in English | MEDLINE | ID: mdl-36798871

ABSTRACT

Background: Atherosclerosis and type 2 diabetes mellitus contribute to a large part of cardiovascular events, but the underlying mechanism remains unclear. In this study, we focused on identifying the linking genes of the diagnostic biomarkers and effective therapeutic targets associated with these two diseases. Methods: The transcriptomic datasets of atherosclerosis and type 2 diabetes mellitus were obtained from the GEO database. Differentially expressed genes analysis was performed by R studio software, and differential analysis including functional enrichment, therapeutic small molecular agents prediction, and protein-protein interaction analysis were applied to the common shared differentially expressed genes. Hub genes were identified and further validated using an independent dataset and clinical samples. Furthermore, we measured the expression correlations, immune cell infiltration, and diagnostic capability of the three key genes. Results: We screened out 28 up-regulated and six down-regulated common shared differentially expressed genes. Functional enrichment analysis showed that cytokines and immune activation were involved in the development of these two diseases. Six small molecules with the highest absolute enrichment value were identified. Three critical genes (CD4, PLEK, and THY1) were further validated both in validation sets and clinical samples. The gene correlation analysis showed that CD4 was strongly positively correlated with PLEK, and ROC curves confirmed the good discriminatory capacity of CD4 and PLEK in two diseases.We have established the co-expression network between atherosclerosis lesions progressions and type 2 diabetes mellitus, and identified CD4 and PLEK as key genes in the two diseases, which may facilitate both development of diagnosis and therapeutic strategies.

6.
Acta Biochim Biophys Sin (Shanghai) ; 55(2): 1-13, 2023 02 25.
Article in English | MEDLINE | ID: mdl-36815373

ABSTRACT

Accumulating evidence has shown that the apoptosis of trophoblast cells plays an important role in the pathogenesis of preeclampsia, and an intricate interplay between DNA methylation and polycomb group (PcG) protein-mediated gene silencing has been highlighted recently. Here, we provide evidence that the expression of nervous system polycomb 1 (NSPc1), a BMI1 homologous polycomb protein, is significantly elevated in trophoblast cells during preeclampsia, which accelerates trophoblast cell apoptosis. Since NSPc1 acts predominantly as a transcriptional inactivator that specifically represses HOXA11 expression in trophoblast cells during preeclampsia, we further show that NSPc1 is required for DNMT3a recruitment and maintenance of the DNA methylation in the HOXA11 promoter in trophoblast cells during preeclampsia. In addition, we find that the interplay of DNMT3a and NSPc1 represses the expression of HOXA11 and promotes trophoblast cell apoptosis. Taken together, these results indicate that the cooperation between NSPc1 and DNMT3a reduces HOXA11 expression in preeclampsia pathophysiology, which provides novel therapeutic approaches for targeted inhibition of trophoblast cell apoptosis during preeclampsia pathogenesis.


Subject(s)
Pre-Eclampsia , Trophoblasts , Humans , Pregnancy , Female , Trophoblasts/metabolism , DNA Methylation , Pre-Eclampsia/genetics , Pre-Eclampsia/metabolism , Promoter Regions, Genetic , Polycomb-Group Proteins/metabolism , Apoptosis , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism
7.
Acta Biochim Biophys Sin (Shanghai) ; 54(12): 1-9, 2022 Dec 25.
Article in English | MEDLINE | ID: mdl-36514215

ABSTRACT

Long noncoding RNAs (lncRNAs) are increasingly being implicated as key regulators of cell proliferation, apoptosis, and differentiation. However, the molecular mechanisms of specific lncRNAs in the context of hypertrophic scar remain largely unclear. Here, we find that the lncRNA FPASL (fibroblast proliferation-associated LncRNA) is downregulated in HS, and FPASL reduces fibroblast proliferation and colony formation and blocks cell cycle progression. Using GO annotation enrichment analysis along with AZC (a specific inhibitor of DNA methylation), we identify that DNA methylation is responsible for downregulating FPASL in hypertrophic scar. Subsequent studies demonstrate that high expression of DNMT3b inhibits FPASL expression in HS. Mechanistic study reveals a significant increase in fibroblast proliferation after transfection with LNA-FPASL, which is further inhibited by knockdown of DNMT3b. Thus, our study reveals that DNMT3b mediates hypermethylation of the lncRNA FPASL promoter and the downregulation of lncRNA FPASL promotes fibroblast proliferation in hypertrophic scar.


Subject(s)
Cicatrix, Hypertrophic , RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Cicatrix, Hypertrophic/metabolism , DNA Methylation , Cell Proliferation/genetics , Fibroblasts/metabolism
8.
Acta Biochim Biophys Sin (Shanghai) ; 55(2): 274-284, 2022 Sep 25.
Article in English | MEDLINE | ID: mdl-36082934

ABSTRACT

Hypertrophic scar is a problem for numerous patients, especially after burns, and is characterized by increased fibroblast proliferation and collagen deposition. Increasing evidence demonstrates that lncRNAs contribute to the development and progression of various diseases. However, the function of lncRNAs in hypertrophic scar formation remains poorly characterized. In this study, a novel fibroblast proliferation-associated lncRNA, named lncRNA FPASL (MSTRG.389905.1), which is mainly localized in the cytoplasm, is found to be downregulated in hypertrophic scar, as detected by lncRNA microarray and qRT-PCR. The full-length FPASL is characterized and further investigation confirms that it has no protein-coding potential. FPASL knockdown in fibroblasts triggers fibroblast proliferation, whereas overexpression of FPASL directly attenuates the proliferation of fibroblasts. Furthermore, target genes of the differentially expressed lncRNAs in hypertrophic scars and the matched adjacent normal tissues are enriched in fibroblast proliferation signaling pathways, including the PI3K/AKT and MAPK signaling pathways, as determined by GO annotation and KEGG enrichment analysis. We also demonstrate that knockdown of FPASL activates the PI3K/AKT and MAPK signaling pathways, and specific inhibitors of the PI3K/AKT and MAPK signaling pathways can reverse the proliferation of fibroblasts promoted by FPASL knockdown. Our findings contribute to a better understanding of the role of lncRNAs in hypertrophic scar and suggest that FPASL may act as a potential novel therapeutic target for hypertrophic scar.


Subject(s)
Cicatrix, Hypertrophic , RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Cicatrix, Hypertrophic/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/genetics , Cell Proliferation/genetics , Fibroblasts/metabolism
9.
Oxid Med Cell Longev ; 2022: 9635674, 2022.
Article in English | MEDLINE | ID: mdl-35656020

ABSTRACT

Ischemic postconditioning (IPostC) has been proposed as a strategy to mitigate the risk of ischemia/reperfusion (I/R) injury, and autophagy is involved in I/R-induced aged myocardial injury, while the underlying mechanism of IPostC-regulated autophagy is unknown. Here, we implemented miRNA sequencing analysis in aged cardiomyocytes to identify a novel miR-181a-2-3p after HPostC, which inhibits autophagy by targeting AMBRA1 in aged myocardium to protect I/R-induced aged myocardial injury. Mechanistically, we identified that IPostC can induce DNA hypomethylation and H3K14 hyperacetylation of miR-181a-2-3p promoter due to the decreased binding of DNMT3b and HDAC2 at its promoter, which contributes to enhancing the expression of miR-181a-2-3p. More importantly, cooperation of DNMT3b and HDAC2 inhibits the binding of c-Myc at the miR-181a-2-3p promoter in aged cardiomyocytes. In summary, IPostC attenuates I/R-induced aged myocardial injury through upregulating miR-181a-2-3p expression, which is an attribute to transcriptional and epigenetic regulation of its promoter. Our data indicate that miR-181a-2-3p may be a potential therapeutic target against I/R injury in aged myocardium.


Subject(s)
Ischemic Postconditioning , MicroRNAs , Myocardial Reperfusion Injury , Adaptor Proteins, Signal Transducing/metabolism , Epigenesis, Genetic , Humans , MicroRNAs/metabolism , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/metabolism
10.
Front Oncol ; 12: 861857, 2022.
Article in English | MEDLINE | ID: mdl-35371991

ABSTRACT

Purpose: To introduce an end-to-end automatic segmentation model for organs at risk (OARs) in thoracic CT images based on modified DenseNet, and reduce the workload of radiation oncologists. Materials and Methods: The computed tomography (CT) images of 36 lung cancer patients were included in this study, of which 27 patients' images were randomly selected as the training set, 9 patients' as the testing set. The validation set was generated by cross validation and 6 patients' images were randomly selected from the training set during each epoch as the validation set. The autosegmentation task of the left and right lungs, spinal cord, heart, trachea and esophagus was implemented, and the whole training time was approximately 5 hours. Geometric evaluation metrics including the Dice similarity coefficient (DSC), 95% Hausdorff distance (HD95) and average surface distance (ASD), were used to assess the autosegmentation performance of OARs based on the proposed model and were compared with those based on U-Net as benchmarks. Then, two sets of treatment plans were optimized based on the manually contoured targets and OARs (Plan1), as well as the manually contours targets and the automatically contoured OARs (Plan2). Dosimetric parameters, including Dmax, Dmean and Vx, of OARs were obtained and compared. Results: The DSC, HD95 and ASD of the proposed model were better than those of U-Net. The differences in the DSC of the spinal cord and esophagus, differences in the HD95 of the spinal cord, heart, trachea and esophagus, as well as differences in the ASD of the spinal cord were statistically significant between the two models (P<0.05). The differences in the dose-volume parameters of the two sets of plans were not statistically significant (P>0.05). Moreover, compared with manual segmentation, autosegmentation significantly reduced the contouring time by nearly 40.7% (P<0.05). Conclusions: The bilateral lungs, spinal cord, heart and trachea could be accurately delineated using the proposed model in this study; however, the automatic segmentation effect of the esophagus must still be further improved. The concept of feature map reuse provides a new idea for automatic medical image segmentation.

11.
Cell Biol Int ; 46(8): 1236-1248, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35347798

ABSTRACT

Endoplasmic reticulum (ER) stress and apoptosis play a critical role in liver injury. Endoplasmic reticulum oxidoreductase 1α (ERO1α) is an oxidase that exists in the luminal side of the ER membrane, participating in protein folding and secretion and inhibiting apoptosis, but the underlying mechanism on liver injury induced by homocysteine (Hcy) remains obscure. In this study, hyperhomocysteinemia (HHcy) mice model was established in cbs+/- mice by feeding a high-methionine diet for 12 weeks; and cbs+/- mice fed with high-methionine diet exhibited more severe liver injury compared to cbs+/+ mice. Mechanistically, we found that Hcy promoted ER stress and apoptosis of hepatocytes and thereby aggravated liver injury through inhibiting ERO1α expression; accordingly, overexpression of ERO1α remarkably alleviated ER stress and apoptosis of hepatocytes induced by Hcy. Epigenetic modification analysis revealed that Hcy significantly increased levels of DNA methylation and H3 lysine 9 dimethylation (H3K9me2) on ERO1α promoter, which attributed to upregulated DNA methyltransferase 1 (DNMT1) and G9a, respectively. Further study showed that DNMT1 and G9a cooperatively regulated ERO1α expression in hepatocytes exposed to Hcy. Taken together, our work demonstrates that Hcy activates ER stress and apoptosis of hepatocytes by downregulating ERO1α expression via cooperation between DNMT1 and G9a, which provides new insight into the mechanism of Hcy-induced ER stress and apoptosis of hepatocytes in liver injury.


Subject(s)
Apoptosis , DNA (Cytosine-5-)-Methyltransferase 1 , Endoplasmic Reticulum Stress , Hepatocytes , Histone-Lysine N-Methyltransferase , Homocysteine , Animals , Apoptosis/genetics , Apoptosis/physiology , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Endoplasmic Reticulum Stress/genetics , Hepatocytes/metabolism , Histone-Lysine N-Methyltransferase/genetics , Homocysteine/genetics , Homocysteine/metabolism , Methionine/metabolism , Mice , Oxidoreductases/genetics
12.
Lab Invest ; 102(1): 25-37, 2022 01.
Article in English | MEDLINE | ID: mdl-34725437

ABSTRACT

Atherosclerosis is a chronic inflammatory vascular disease, and inflammation plays a critical role in its formation and progression. Elevated serum homocysteine (Hcy) is an independent risk factor for atherosclerosis. Previous studies have shown that fatty acid binding protein 4 (FABP4) plays an important role in macrophage inflammation and lipid metabolism in atherosclerosis induced by Hcy. However, the underlying molecular mechanism of FABP4 in Hcy-induced macrophage inflammation remains unknown. In this study, we found that FABP4 activated the Janus kinase 2/signal transducer and activator of transcription 2 (JAK2/STAT2) pathway in macrophage inflammation induced by Hcy. Of note, we further observed that ras-related protein Rap-1a (Rap1a) induced the Tyr416 phosphorylation and membrane translocation of non-receptor tyrosine kinase (c-Src) to activate the JAK2/STAT2 pathway. In addition, the suppressor of cytokine signaling 1 (SOCS1)-a transcriptional target of signal transducer and activator of transcription (STATs) inhibited the JAK2/STAT2 pathway and Rap1a expression via a negative feedback loop. In summary, these results demonstrated that FABP4 promotes c-Src phosphorylation and membrane translocation via Rap1a to activate the JAK2/STAT2 pathway, contributing to Hcy-accelerated macrophage inflammation in ApoE-/- mice.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/genetics , Homocysteine/pharmacology , Inflammation Mediators/metabolism , Macrophages/drug effects , Proteins/genetics , Signal Transduction/genetics , Animals , Apolipoproteins E/metabolism , Atherosclerosis/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Gene Expression Profiling/methods , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Proteins/metabolism , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , THP-1 Cells , rap1 GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/metabolism
13.
J Control Release ; 341: 828-843, 2022 01.
Article in English | MEDLINE | ID: mdl-34942304

ABSTRACT

The development of new reagents combining with nanotechnology has become an efficient strategy for improving the immune escaping ability and increasing local drug concentration for natural compounds with low therapy efficiency. In this study, we prepared biomimetic membrane-coated Prussian blue nanoparticles (PB NPs) for the treatment of atherosclerosis, using the function of Artemisinin (ART) and Procyanidins (PC) on the lipid influx and cholesterol efflux of macrophages, two logical steps involved in the plaque progression. In vitro results indicated that the prepared nanocomplexes have significant scavenging effect on ROS and NO, followed by inhibiting NF-κB/NLRP3 pathway, leading to the suppression of lipid influx. Meanwhile, they can notably reduce the uptake and internalization of oxLDL through significantly enhancing AMPK/mTOR/autophagy pathway, accompanied by promoting cholesterol efflux. In vivo study showed that the improved biocompatibility and immune-escape ability of nanocomplexes allowed less drug clearance during the circulation and high drug accumulation in the atherosclerotic plaque of ApoE-/- mice model. More importantly, the ART and PC co-loaded nanocomplexes showed the high efficacy against atherosclerosis of ApoE-/- mice model with both 8-week low dosage treatment or 1-week high dosage treatment. These findings indicated that ART and PC co-loaded nanocomplexes was promising for the targeted treatment of atherosclerosis.


Subject(s)
Artemisinins , Atherosclerosis , Plaque, Atherosclerotic , Proanthocyanidins , Animals , Artemisinins/therapeutic use , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Cholesterol/metabolism , Mice , Plaque, Atherosclerotic/drug therapy , Proanthocyanidins/therapeutic use
14.
Mol Ther Nucleic Acids ; 26: 1318-1335, 2021 Dec 03.
Article in English | MEDLINE | ID: mdl-34853730

ABSTRACT

It has been demonstrated that homocysteine (Hcy) can cause inflammatory diseases. Long noncoding RNAs (lncRNA) and microRNAs (miRNAs) are involved in this biological process, but the mechanism underlying Hcy-induced inflammation remains poorly understood. Here, we found that lncRNA TGFB3-AS1 was highly expressed in macrophages treated with Hcy and the peripheral blood monocytes from cystathionine beta-synthase heterozygous knockout (CBS +/-) mice with a high-methionine diet using lncRNA microarray. In vivo and in vitro experiments further confirmed that TGFB3-AS1 accelerated Hcy-induced inflammation of macrophages through the Rap1a/wnt signaling pathway. Meanwhile, TGFB3-AS1 interacted with Rap1a and reduced degradation of Rap1a through inhibiting its ubiquitination in macrophages treated with Hcy. Rap1a mediated inflammation induced by Hcy and serves as a direct target of miR-144. Moreover, TGFB3-AS1 regulated miR-144 by binding to pri-miR-144 and inhibiting its maturation, which further regulated Rap1a expression. More importantly, we found that high expression of TGFB3-AS1 was positively correlated with the levels of Hcy and proinflammatory cytokines in serum of healthy individuals and patients with HHcy. Our study revealed a novel mechanism by which TGFB3-AS1 promoted inflammation of macrophages through inhibiting miR-144 maturation to stay miR-144 regulated inhibition of functional Rap1a expression.

15.
Aging Cell ; 20(10): e13485, 2021 10.
Article in English | MEDLINE | ID: mdl-34592792

ABSTRACT

Atherosclerosis is a serious age-related disease, which has a tremendous impact on health care globally. Macrophage inflammation is crucial for the initiation and progression of atherosclerosis, and microRNAs (miRNAs) recently have emerged as potent modulators of inflammation, while the underlying mechanisms of its involvement in homocysteine (Hcy)-mediated macrophage inflammation of atherosclerosis remain largely unknown. Here, we demonstrated that elevated Hcy inhibits the expression of miR-195-3p, which in turn enhances IL-31 expression and thereby causes the secretion of macrophages pro-inflammatory factors IL-1ß, IL-6 and TNF-α and accelerate atherosclerosis. Furthermore, we identified that Hcy can induce DNA hypermethylation and H3K9 deacetylation of miR-195-3p promoter due to the increased the binding of DNMT3a and HDAC11 at its promoter. More importantly, Sp1 interacts with DNMT3a suppressed the binding of HDAC11 at miR-195-3p promoter and promoted its transcription. In summary, our results revealed a novel mechanism that transcriptional and epigenetic regulation of miR-195-3p inhibits macrophage inflammation through targeting IL-31, which provides a candidate diagnostic marker and novel therapeutic target in cardiovascular diseases induced by Hcy.


Subject(s)
Atherosclerosis/chemically induced , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Homocysteine/adverse effects , Interleukins/metabolism , Animals , Apoptosis , Humans , Mice
16.
Mol Oncol ; 15(11): 3203-3221, 2021 11.
Article in English | MEDLINE | ID: mdl-34057794

ABSTRACT

Chronic kidney disease (CKD) is a common and complex disease in kidneys which has been associated with an increased risk of renal cell carcinoma. Elevated homocysteine (Hcy) levels are known to influence the development and progression of CKD by regulating podocyte injury and apoptosis. To investigate the molecular mechanisms triggered in podocytes by Hcy, we used cbs+/- mice and observed that higher Hcy levels increased the apoptosis rate of podocytes with accompanying glomerular damage. Hcy-induced podocyte injury and apoptosis in cbs+/- mice was regulated by inhibition of microRNA (miR)-1929-5p expression. Overexpression of miR-1929-5p in podocytes inhibited apoptosis by upregulating Bcl-2. Furthermore, the expression of miR-1929-5p was regulated by epigenetic modifications of its promoter. Hcy upregulated DNA methyltransferase 1 (DNMT1) and enhancer of zeste homolog 2 (EZH2) levels, resulting in increased DNA methylation and H3K27me3 levels on the miR-1929-5p promoter. Additionally, we observed that c-Myc recruited DNMT1 and EZH2 to the miR-1929-5p promoter and suppressed the expression of miR-1929-5p. In summary, we demonstrated that Hcy promotes podocyte apoptosis through the regulation of the epigenetic modifiers DNMT1 and EZH2, which are recruited by c-Myc to the promoter of miR-1929-5p to silence miR-1929-5p expression.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1 , Enhancer of Zeste Homolog 2 Protein , Homocysteine , MicroRNAs , Podocytes , Proto-Oncogene Proteins c-myc , Animals , Apoptosis/genetics , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Homocysteine/pharmacology , Humans , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Podocytes/metabolism , Podocytes/pathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism
17.
Mol Med Rep ; 23(6)2021 06.
Article in English | MEDLINE | ID: mdl-33880587

ABSTRACT

Our previous study reported that microRNA (miR)­30a­5p upregulation under hypoxia postconditioning (HPostC) exert a protective effect on aged H9C2 cells against hypoxia/reoxygenation injury via DNA methyltransferase 3B­induced DNA hypomethylation at the miR­30a­5p gene promoter. This suggests that miR­30a­5p may be a potential preventative and therapeutic target for ischemic heart disease in aged myocardium. The present study aimed to investigate the underlying mechanisms of miR­30a­5p transcription in aged myocardium in ischemic heart disease. Cardiomyocytes were treated with 8 mg/ml D­galactose for 9 days, and then exposed to hypoxic conditions. Cell viability was determined using a cell viability assay. Expression levels of histone deacetylase 2 (HDAC2), LC3B­II/I, beclin­1 and p62 were detected via reverse transcription­quantitative PCR and western blotting. Chromatin immunoprecipitation­PCR and luciferase reporter assays were performed to evaluate the effect of c­Myc binding and activity on the miR­30a­5p promoter in senescent cardiomyocytes following HPostC. It was found that HPostC enhanced the acetylation levels of H3K14 at the miR­30a­5p gene promoter in senescent cardiomyocytes, which attributed to the decreased expression of HDAC2. In addition, c­Myc could positively regulate miR­30a­5p transcription to inhibit senescent cardiomyocyte autophagy. Mechanically, it was observed that increased H3K14 acetylation level exposed to romidepsin facilitated c­Myc binding to the miR­30a­5p gene promoter region, which led to the increased transcription of miR­30a­5p. Taken together, these results demonstrated that HDAC2­mediated H3K14 hyperacetylation promoted c­Myc binding to the miR­30a­5p gene promoter, which contributed to HPostC senescent cardioprotection.


Subject(s)
Histones/metabolism , Hypoxia/metabolism , MicroRNAs/genetics , Myocytes, Cardiac/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/metabolism , Animals , Autophagy , Beclin-1 , Cell Survival/drug effects , DNA (Cytosine-5-)-Methyltransferases , DNA Methylation , MicroRNAs/metabolism , Protective Agents/pharmacology , Rats , Up-Regulation , DNA Methyltransferase 3B
18.
Int J Biochem Cell Biol ; 135: 105974, 2021 06.
Article in English | MEDLINE | ID: mdl-33831591

ABSTRACT

Homocysteine (Hcy) is a strong and independent risk factor of atherosclerosis. It can accelerate atherosclerosis through increased production of inflammatory factors, especially interleukin-1 ß (IL-1ß), while the precise mechanisms remain to be well elucidated. In this study, we investigated the role of the tumor suppressor gene SNF5 related to switch/sucrose non-fermentable complex (SWI/SNF) in the occurrence and development of atherosclerosis induced by Hcy. Using Hyperhomocysteinemia (HHcy) atherosclerotic model with apolipoprotein E knockout (ApoE-/-) mice fed with high-methionine diet, we showed that Hcy aggravates inflammation in macrophages during the atherosclerotic plaque formation. Further analysis showed that SNF5 promotes IL-1ß expression and secretion. In addition, due to the existence of H3K4 methylation signals in the vicinity of IL-1ß, we found that Hcy significantly promotes the expression of H3K4me1, and lysine-specific histone demethylase 1A (KDM1A) acts as a transcriptional repressor to regulate the expression of H3K4me1 by demethylating H3K4me1. In summary, our results demonstrated that Hcy up-regulates the expression of SNF5 through KDM1A, resulting in an increased level of H3K4me1 modification and IL-1ß in macrophages, which in turn promotes the formation of atherosclerosis. Our study will provide more evidence for further revealing the specific mechanism of Hcy-induced inflammation and the diagnosis, prevention, and treatment of atherosclerosis.


Subject(s)
Atherosclerosis/pathology , Gene Expression Regulation/drug effects , Histones/metabolism , Homocysteine/toxicity , Inflammation/complications , Interleukin-1beta/metabolism , SMARCB1 Protein/metabolism , Animals , Atherosclerosis/chemically induced , Atherosclerosis/metabolism , Histones/genetics , Interleukin-1beta/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , SMARCB1 Protein/genetics
19.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 37(3): 240-245, 2021 Mar.
Article in Chinese | MEDLINE | ID: mdl-33766232

ABSTRACT

Objective To study the role of long non-coding RNA growth arrest specific transcript 5 (lncGAS5) in the autophagy of hepatocytes induced by homocysteine (Hcy). Methods HL7702 human hepatocyte cells were cultured in vitro and divided into control group and Hcy group. Western blotting was used to detect the expression levels of microtubule-associated protein 1 light chain 3B (LC3B) and P62. The cells were transfected with mRFP-GFP-LC3 adenovirus to observe the autophagy flow with laser scanning confocal microscope. Real-time quantitative PCR was performed to detect the expression level of lncGAS5. lncGAS5 small interfering RNA (si-lncGAS5) and negative control small interfering RNA (si-NC) were transfected into the cells. After the transfected cells were treated with Hcy, the changes of LC3B, P62 and autophagy flow were analyzed with the above methods. Results Compared with the control group, the LC3BII/LC3BI ratio increased and the expression of P62 protein decreased in the Hcy group. When the lever of Hcy lifted, the number of autophagosomes and autolysosomes and the expression of lncGAS5 increased in the cells. After knock-down of lncGAS5, the ratio of LC3BII/LC3BI decreased and the expression of P62 increased. Moreover, the number of autophagosomes and autolysosomes were reduced in the cells. Conclusion lncGAS5 can promote the autophagy of hepatocytes induced by Hcy.


Subject(s)
RNA, Long Noncoding , Autophagy/genetics , Hepatocytes , Homocysteine/pharmacology , Humans , RNA, Long Noncoding/genetics , RNA, Small Nucleolar
20.
Circ J ; 84(4): 616-625, 2020 03 25.
Article in English | MEDLINE | ID: mdl-32115441

ABSTRACT

BACKGROUND: Ischemic postconditioning (IPostC) is an endogenous protective mechanism to reduce ischemia-reperfusion (I/R) injury. However, whether IPostC protects aged cardiomyocytes against I/R injury is not fully understood. Considering the protective function of microRNA 30a (miR-30a) against ischemia-induced injury in H9C2 cells, its role in the protective effects of IPostC on I/R injury of aged cardiomyocytes was investigated further.Methods and Results:To mimic I/R and IPostC in vitro, the aged cardiomyocyte model for hypoxia postconditioning (HPostC) treatment was established by 9 days of incubation with 8 mg/mL D-galactose and then followed by exposure to hypoxic environment. HPostC significantly alleviated hypoxia/reoxygenation (H/R) injury and reduced autophagy of aged cardiomyocytes, as evidenced by decreased LC3B-II expression and increased p62 by Western blot. Quantified by quantitative real-time polymerase chain reaction (qRT-PCR), miR-30a was increased in aged cardiomyocytes treated with HPostC compared with I/R injury group. Overexpression of miR-30a by LV3-rno-miR-30a mimic promoted cardioprotective effect of HPostC in aged cardiomyocytes by suppressing BECN1-mediated autophagy, all of which was abrogated by knockdown of miR-30a expression. Epigenetic analyses demonstrated that HPostC reduced DNA methyltransferase 3b-mediated DNA hypomethylation levels at miR-30a promoter, leading to upregulation of miR-30a. CONCLUSIONS: HPostC protected aged cardiomyocytes survival against H/R injury via DNMT3b-dependent activation of miR-30a. miR-30a could be a potential therapeutic target for ischemic myocardial infarction.


Subject(s)
Autophagy , Cellular Senescence , DNA Methylation , Epigenesis, Genetic , MicroRNAs/metabolism , Myocardial Reperfusion Injury/prevention & control , Animals , Beclin-1/genetics , Beclin-1/metabolism , Cell Hypoxia , Cell Line , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , MicroRNAs/genetics , Microtubule-Associated Proteins/metabolism , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Promoter Regions, Genetic , Rats , Sequestosome-1 Protein/metabolism , Signal Transduction , DNA Methyltransferase 3B
SELECTION OF CITATIONS
SEARCH DETAIL
...