Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Acta Biochim Biophys Sin (Shanghai) ; 55(8): 1184-1192, 2023 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-37021975

RESUMEN

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.


Asunto(s)
Autofagia , Metilación de ADN , Hepatocitos , Homocisteína , Autofagia/genética , ADN , Homocisteína/metabolismo , Homocisteína/farmacología , Humanos , ADN Metiltransferasa 3B
2.
Acta Biochim Biophys Sin (Shanghai) ; 55(2): 1-13, 2023 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-36815373

RESUMEN

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.


Asunto(s)
Preeclampsia , Trofoblastos , Humanos , Embarazo , Femenino , Trofoblastos/metabolismo , Metilación de ADN , Preeclampsia/genética , Preeclampsia/metabolismo , Regiones Promotoras Genéticas , Proteínas del Grupo Polycomb/metabolismo , Apoptosis , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo
3.
Lab Invest ; 102(1): 25-37, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34725437

RESUMEN

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.


Asunto(s)
Apolipoproteínas E/genética , Aterosclerosis/genética , Homocisteína/farmacología , Mediadores de Inflamación/metabolismo , Macrófagos/efectos de los fármacos , Proteínas/genética , Transducción de Señal/genética , Animales , Apolipoproteínas E/metabolismo , Aterosclerosis/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Perfilación de la Expresión Génica/métodos , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Macrófagos/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas/metabolismo , Factor de Transcripción STAT2/genética , Factor de Transcripción STAT2/metabolismo , Células THP-1 , Proteínas de Unión al GTP rap1/genética , Proteínas de Unión al GTP rap1/metabolismo
4.
Cell Biol Int ; 46(8): 1236-1248, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35347798

RESUMEN

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.


Asunto(s)
Apoptosis , ADN (Citosina-5-)-Metiltransferasa 1 , Estrés del Retículo Endoplásmico , Hepatocitos , N-Metiltransferasa de Histona-Lisina , Homocisteína , Animales , Apoptosis/genética , Apoptosis/fisiología , ADN (Citosina-5-)-Metiltransferasa 1/genética , Estrés del Retículo Endoplásmico/genética , Hepatocitos/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Homocisteína/genética , Homocisteína/metabolismo , Metionina/metabolismo , Ratones , Oxidorreductasas/genética
5.
Acta Biochim Biophys Sin (Shanghai) ; 55(2): 274-284, 2022 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-36082934

RESUMEN

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.


Asunto(s)
Cicatriz Hipertrófica , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Cicatriz Hipertrófica/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/genética , Proliferación Celular/genética , Fibroblastos/metabolismo
6.
Acta Biochim Biophys Sin (Shanghai) ; 54(12): 1-9, 2022 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-36514215

RESUMEN

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.


Asunto(s)
Cicatriz Hipertrófica , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Cicatriz Hipertrófica/metabolismo , Metilación de ADN , Proliferación Celular/genética , Fibroblastos/metabolismo
7.
J Mol Cell Cardiol ; 138: 34-48, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31733201

RESUMEN

Homocysteine (Hcy) is an independent risk factor for atherosclerosis, which is characterized by lipid accumulation in the atherosclerotic plaque. Increasing evidence supports that as the main receptor of high-density lipoprotein, scavenger receptor class B member 1 (SCARB1) is protective against atherosclerosis. However, the underlying mechanism regarding it in Hcy-mediated atherosclerosis remains unclear. Here, we found the remarkable inhibition of SCARB1 expression in atherosclerotic plaque and Hcy-treated foam cells, whereas overexpression of SCARB1 can suppress lipid accumulation in foam cells following Hcy treatment. Analysis of SCARB1 promoter showed that no significant change of methylation level was observed both in vivo and in vitro under Hcy treatment. Moreover, it was found that the negative regulation of DNMT3b on SCARB1 was due to the decreased recruitment of SP1 to SCARB1 promoter. Thus, we concluded that inhibition of SCARB1 expression induced by DNMT3b at least partly accelerated Hcy-mediated atherosclerosis through promoting lipid accumulation in foam cells, which was attributed to the decreased binding of SP1 to SCARB1 promoter. In our point, these findings will provide novel insight into an epigenetic mechanism for atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , Aterosclerosis/patología , Antígenos CD36/metabolismo , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Homocisteína/efectos adversos , Transducción de Señal , Factor de Transcripción Sp1/metabolismo , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/metabolismo , Aterosclerosis/complicaciones , Metilación de ADN/genética , Dieta , Progresión de la Enfermedad , Regulación hacia Abajo/genética , Células Espumosas/metabolismo , Células HEK293 , Humanos , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/patología , Masculino , Metionina , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Placa Aterosclerótica/complicaciones , Placa Aterosclerótica/patología , Regiones Promotoras Genéticas , Unión Proteica , Células THP-1 , ADN Metiltransferasa 3B
8.
Circ J ; 84(4): 616-625, 2020 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-32115441

RESUMEN

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.


Asunto(s)
Autofagia , Senescencia Celular , Metilación de ADN , Epigénesis Genética , MicroARNs/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Animales , Beclina-1/genética , Beclina-1/metabolismo , Hipoxia de la Célula , Línea Celular , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , MicroARNs/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Regiones Promotoras Genéticas , Ratas , Proteína Sequestosoma-1/metabolismo , Transducción de Señal , ADN Metiltransferasa 3B
9.
Exp Cell Res ; 362(1): 217-226, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29155363

RESUMEN

Endothelial progenitor cells (EPCs) contribute to neovasculogenesis and reendothelialization of damaged blood vessels to maintain the endothelium. Dysfunction of EPCs is implicated in the pathogenesis of vascular injury induced by homocysteine (Hcy). We aimed to investigate the role of Cyclin A in Hcy-induced EPCs dysfunction and explore its molecular mechanism. In this study, by treatment of EPCs with Hcy, we found that the expression of Cyclin A mRNA and protein were significantly downregulated in a dose-dependent manner. Knockdown of Cyclin A prominently reduced proliferation of EPCs, while over-expression of Cyclin A significantly promoted the cell proliferation, suggesting that Hcy inhibits EPCs proliferation through downregulation of Cyclin A expression. In addition, epigenetic study also demonstrated that Hcy induces DNA hypomethylation of the Cyclin A promoter in EPCs through downregulated expression of DNMT1. Moreover, we found that Hcy treatment of EPCs leads to increased SAM, SAH and MeCP2, while the ratio of SAM/SAH and MBD expression decrease. In summary, our results indicate that Hcy inhibits Cyclin A expression through hypomethylation of Cyclin A and thereby suppress EPCs proliferation. These findings demonstrate a novel mechanism of DNA methylation mediated by DNMT1 in prevention of Hcy associated cardiovascular disease.


Asunto(s)
Proliferación Celular/fisiología , Ciclina A/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Metilación de ADN/fisiología , Células Progenitoras Endoteliales/metabolismo , Homocisteína/metabolismo , Animales , Células Cultivadas , Regulación hacia Abajo/fisiología , Epigénesis Genética/fisiología , Humanos , Regiones Promotoras Genéticas/fisiología , Ratas
10.
J Cell Biochem ; 118(9): 2921-2932, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28230279

RESUMEN

Folate deficiency is a known risk factor for liver injury; however, the underlying mechanism remains unclear. In this study, we employed a high homocysteine-induced liver injury model of Apolipoprotein E-deficient (ApoE-/- ) mice fed high-methionine diet and found that high homocysteine induced endoplasmic reticulum (ER) stress and liver cell apoptosis by downregulation of cystic fibrosis transmembrane conductance regulator (CFTR) expression; observations that were attenuated with supplementation of dietary folate. The regulation on CFTR expression was mediated by CFTR promoter methylation and trimethylation of lysine 27 on histone H3 (H3K27me3). Mechanistically, folate inhibited homocysteine-induced CFTR promoter methylation and H3K27me3, which resulted in upregulation of CFTR expression, and reduced ER stress and liver cell apoptosis. Further study showed that folate inhibited the expression of DNA methyltransferase 1 and enhancer of zeste homolog 2, downregulated the cellular concentrations of S-adenosylmethionine (SAM) and S-adenosylhomocysteine (SAH) and upregulated the SAM/SAH ratio, leading to the inhibition of Hcy-induced DNA hypermethylation and H3K27me3 in CFTR promoter. In conclusion, our results provide insight into the protective role of folate in homocysteine-induced ER stress and liver cell apoptosis through the regulation of CFTR expression. J. Cell. Biochem. 118: 2921-2932, 2017. © 2017 Wiley Periodicals, Inc. HIGHLIGHTS: Folate protects hepatocytes of hyperhomocysteinemia mice from apoptosis. Folate alleviates Hcy-induced hepatocyte apoptosis. Folate inhibits Hcy-induced ER stress via upregulation of CFTR expression in hepatocytes. Folate inhibits Hcy-induced methylation of CFTR promotor and H3K27me3.


Asunto(s)
Apoptosis/efectos de los fármacos , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Metilación de ADN/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Ácido Fólico/farmacología , Hepatocitos/metabolismo , Hiperhomocisteinemia/metabolismo , Regiones Promotoras Genéticas , Animales , Apoptosis/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Metilación de ADN/genética , Estrés del Retículo Endoplásmico/genética , Hepatocitos/patología , Hiperhomocisteinemia/genética , Hiperhomocisteinemia/patología , Ratones , Ratones Noqueados
11.
J Cell Biochem ; 118(12): 4617-4627, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28485501

RESUMEN

Hyperhomocysteinemia (HHcy) promotes atherogenesis by modification of histone acetylation patterns and regulation of miRNA expression while the underlying molecular mechanisms are not well known. In this study, we investigated the effects of homocysteine (Hcy) on the expression of histone deacetylase 1 (HDAC1) and tested our hypothesis that Hcy-induced atherosclerosis is mediated by increased HDAC1 expression, which is regulated by miR-34a. The expression of HDAC1 increased and acetylation of histone H3 at lysine 9 (H3K9ac) decreased in the aorta of ApoE-/- mice fed with high methionine diet, whereas miR-34a expression was inhibited. Over-expression of HDAC1 inhibited H3K9ac level and promoted the accumulation of total cholesterol, free cholesterol, and triglycerides in the foam cells. Furthermore, up-regulation of miR-34a reduced HDAC1 expression and inhibited the accumulation of total cholesterol (TC), free cholesterol (FC), and triglycerides (TG) in the foam cells. These data suggest that HDAC1-related H3K9ac plays a key role in Hcy-mediated lipid metabolism disorders, and that miR-34a may be a novel therapeutic target in Hcy-related atherosclerosis. J. Cell. Biochem. 118: 4617-4627, 2017. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Aterosclerosis/metabolismo , Colesterol/metabolismo , Células Espumosas/metabolismo , Histona Desacetilasa 1/metabolismo , Homocisteína/metabolismo , MicroARNs/metabolismo , Triglicéridos/metabolismo , Acetilación , Animales , Apolipoproteínas E/deficiencia , Aterosclerosis/genética , Aterosclerosis/patología , Colesterol/genética , Células Espumosas/patología , Histona Desacetilasa 1/genética , Homocisteína/genética , Masculino , Ratones , Ratones Noqueados , MicroARNs/genética , Triglicéridos/genética
12.
Sheng Li Xue Bao ; 67(2): 207-13, 2015 Apr 25.
Artículo en Zh | MEDLINE | ID: mdl-25896052

RESUMEN

The aim of the present study is to explore the role of miR-124 and its promoter region DNA methylation in homocysteine (Hcy)-induced atherosclerosis. ApoE(-/-) mice were fed with hypermethionine diet for 16 weeks to duplicate hyperhomocysteinemia model. Meanwhile, a normal control group (C57BL/6J mice fed with normal diet, N-control) and a model control group (ApoE(-/-) mice fed with normal diet, A-control) were set. The degree of atherosclerosis was observed by HE and oil red O staining. Automatic biochemical analyzer was used to detect the serum levels of Hcy. Foam cell model was duplicated and oil red O staining was used to confirm whether the model was successfully established. And foam cells were stimulated with 0, 50, 100, 200, 500 µmol/L Hcy and 50 µmol/L Hcy + 10 µmol/L AZC respectively. Real-time quantitative PCR (RT-qPCR) was used to detect the expressions of miR-124 in mice aorta and foam cells; Nested landing methylation specific PCR (nMS-PCR) was used to detect the levels of miR-124 promoter DNA methylation in mice aorta and foam cells. Meanwhile, the effects of DNA methylation inhibitor AZC on miR-124 expression were observed at the cellular level. The effect of miR-124 promoter DNA methylation status on lipid accumulation in foam cells was observed by oil red O staining. The results showed that compared with model control group, the serum levels of Hcy in high methionine group were significantly increased (P < 0.01) and developed aortic atherosclerotic plaque, the expression of miR-124 was markedly decreased (P < 0.01), while the levels of miR-124 promoter DNA methylation were significantly increased (P < 0.01). Given different levels of Hcy, the expression of miR-124 in foam cells was decreased, while the levels of miR-124 promoter DNA methylation were increased in a dose-dependent manner (P < 0.05, P < 0.01). AZC reversed the results of mentioned indices as above markedly (P < 0.05). Downregulation of miR-124 may play a role in Hcy-induced atherosclerosis and its promoter DNA methylation status may be an important mechanism in this process.


Asunto(s)
Aterosclerosis/genética , Metilación de ADN , MicroARNs/genética , Animales , Aorta/metabolismo , Apolipoproteínas E , Aterosclerosis/inducido químicamente , Dieta , Células Espumosas/metabolismo , Homocisteína/efectos adversos , Hiperhomocisteinemia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas
13.
Front Psychol ; 15: 1291353, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38495417

RESUMEN

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.

14.
Front Oncol ; 13: 1174530, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37534258

RESUMEN

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.

15.
J Inflamm Res ; 16: 505-521, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36798871

RESUMEN

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.

16.
Biomed Pharmacother ; 161: 114537, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36933378

RESUMEN

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.


Asunto(s)
Ferroptosis , Silicosis , Ratones , Animales , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento de Hepatocito , Factor de Crecimiento Transformador beta1/metabolismo , Fibrosis , Silicosis/tratamiento farmacológico , Silicosis/metabolismo
17.
Front Oncol ; 12: 861857, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35371991

RESUMEN

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.

18.
J Control Release ; 341: 828-843, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34942304

RESUMEN

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.


Asunto(s)
Artemisininas , Aterosclerosis , Placa Aterosclerótica , Proantocianidinas , Animales , Artemisininas/uso terapéutico , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Colesterol/metabolismo , Ratones , Placa Aterosclerótica/tratamiento farmacológico , Proantocianidinas/uso terapéutico
19.
Oxid Med Cell Longev ; 2022: 9635674, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35656020

RESUMEN

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.


Asunto(s)
Poscondicionamiento Isquémico , MicroARNs , Daño por Reperfusión Miocárdica , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Epigénesis Genética , Humanos , MicroARNs/metabolismo , Daño por Reperfusión Miocárdica/genética , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/metabolismo
20.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 37(3): 240-245, 2021 Mar.
Artículo en Zh | MEDLINE | ID: mdl-33766232

RESUMEN

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.


Asunto(s)
ARN Largo no Codificante , Autofagia/genética , Hepatocitos , Homocisteína/farmacología , Humanos , ARN Largo no Codificante/genética , ARN Nucleolar Pequeño
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA