Activating Transcription Factor 3

激活转录因子 3
  • 文章类型: Journal Article
    代谢功能障碍相关脂肪性肝炎(MASH)受肝脏中巨噬细胞和周围细胞之间复杂的相互作用调节。这里,我们表明,Atf3调节葡萄糖-脂肪酸循环在巨噬细胞减弱肝细胞脂肪变性,和肝星状细胞(HSC)中的纤维发生。巨噬细胞中Atf3的过表达可以防止西方饮食喂养小鼠中MASH的发展,而Atf3消融具有相反的效果。机械上,Atf3通过叉头盒O1(FoxO1)和Cd36促进葡萄糖诱导的脂肪酸氧化还原。Atf3通过阻断Hdac1介导的FoxO1在K242、K245和K262的脱乙酰化作用来抑制FoxO1活性,并增加Zdhhc4/5介导的CD36在C3、C7、C464和C466的棕榈酰化作用;巨噬细胞Atf3通过视黄醇结合蛋白4(Rbp4)降低肝细胞脂肪生成和HSC活化。抗Rbp4可以防止巨噬细胞中Atf3缺乏诱导的MASH进展。这项研究确定Atf3是葡萄糖-脂肪酸循环的调节剂。靶向巨噬细胞Atf3或Rbp4可能是MASH的合理治疗策略。
    Metabolic dysfunction-associated steatohepatitis (MASH) is regulated by complex interplay between the macrophages and surrounding cells in the liver. Here, we show that Atf3 regulates glucose-fatty acid cycle in macrophages attenuates hepatocyte steatosis, and fibrogenesis in hepatic stellate cells (HSCs). Overexpression of Atf3 in macrophages protects against the development of MASH in Western diet-fed mice, whereas Atf3 ablation has the opposite effect. Mechanistically, Atf3 improves the reduction of fatty acid oxidation induced by glucose via forkhead box O1 (FoxO1) and Cd36. Atf3 inhibits FoxO1 activity via blocking Hdac1-mediated FoxO1 deacetylation at K242, K245, and K262 and increases Zdhhc4/5-mediated CD36 palmitoylation at C3, C7, C464, and C466; furthermore, macrophage Atf3 decreases hepatocytes lipogenesis and HSCs activation via retinol binding protein 4 (Rbp4). Anti-Rbp4 can prevent MASH progression that is induced by Atf3 deficiency in macrophages. This study identifies Atf3 as a regulator of glucose-fatty acid cycle. Targeting macrophage Atf3 or Rbp4 may be a plausible therapeutic strategy for MASH.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    血吸虫病是一种以肝纤维化为特征的寄生虫病,由肝星状细胞(HSC)的激活和随后的胶原蛋白产生驱动的过程。我们实验室的先前研究已经证明了日本血吸虫蛋白P40(SjP40)抑制HSC活化并发挥抗纤维化作用的能力。在这项研究中,我们旨在阐明重组SjP40(rSjP40)抑制HSCs活化的分子机制。使用rSjP40抑制LX-2细胞活化的细胞模型,我们进行了RNA-seq分析,确定ATF3为最显著改变的基因.进一步的研究表明,rSjP40部分通过抑制ATF3活化来抑制HSC活化。在小鼠肝脏中敲除ATF3可显着减轻日本血吸虫诱导的肝纤维化。此外,我们的结果表明ATF3是microRNA-494-3p的直接靶标,与抗肝纤维化作用相关的microRNA。发现rSjP40通过上调LX-2细胞中的microRNA-494-3p下调ATF3表达。这种下调导致肝纤维化蛋白α-SMA和COL1A1的表达受到抑制,最终缓解了日本血吸虫引起的肝纤维化。
    Schistosomiasis is a parasitic disease characterized by liver fibrosis, a process driven by the activation of hepatic stellate cells (HSCs) and subsequent collagen production. Previous studies from our laboratory have demonstrated the ability of Schistosoma japonicum protein P40 (SjP40) to inhibit HSCs activation and exert an antifibrotic effect. In this study, we aimed to elucidate the molecular mechanism underlying the inhibitory effect of recombinant SjP40 (rSjP40) on HSCs activation. Using a cell model in which rSjP40 inhibited LX-2 cell activation, we performed RNA-seq analyses and identified ATF3 as the most significantly altered gene. Further investigation revealed that rSjP40 inhibited HSCs activation partly by suppressing ATF3 activation. Knockdown of ATF3 in mouse liver significantly alleviated S. japonicum-induced liver fibrosis. Moreover, our results indicate that ATF3 is a direct target of microRNA-494-3p, a microRNA associated with anti-liver fibrosis effects. rSjP40 was found to downregulate ATF3 expression by upregulating microRNA-494-3p in LX-2 cells. This downregulation led to the inhibition of the expression of liver fibrosis proteins α-SMA and COL1A1, ultimately alleviating liver fibrosis caused by S. japonicum.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    激活转录因子3(ATF3)已被确定为与成骨细胞分化相关的调节因子。然而,ATF3对牙周炎中人牙周干细胞(hPDLSCs)成骨分化和增殖的影响尚未见报道。为了建立牙周炎的体外模型,用脂多糖(LPS)攻击hPDLSC。细胞计数试剂盒-8测定用于评估细胞活力,同时采用逆转录定量PCR和蛋白质印迹法检测ATF3的表达。使用ELISA评估炎症释放,以及西方印迹。使用C11BODIPY581/591探针探索脂质过氧化,生化试剂盒,硫代巴比妥酸反应性物质(TBARS)测定和DCFH-DA染色。铁和Fe2+水平,使用相应的试剂盒和蛋白质印迹法测定铁凋亡相关蛋白的表达水平。使用碱性磷酸酶染色评估成骨分化能力,茜素红染色和蛋白质印迹。使用蛋白质印迹鉴定与Nrf2/HO-1信号转导相关的蛋白质的表达水平。结果表明,ATF3在LPS诱导的hPDLSCs中表达上调。ATF3的敲减减轻了LPS诱导的hPDLSCs的炎症反应,随着TNF-α水平的增加,IL-6,IL-1β,Cox-2和iNOS,IL-10水平降低。ATF3沉默也导致TBARS生产率降低,降低活性氧的水平,铁,Fe2+,ACSL4和TFR1,而它升高了SLC7A11和GPX4的水平。此外,ATF3沉默促进hPDLSC矿化和细胞分化,并升高OCN2、RUNX2和BMP2的水平。此外,ATF3耗竭上调与Nrf2/HO-1信号相关的蛋白质的表达水平。Nrf2抑制剂ML385部分抵消了ATF3干扰对LPS攻击的炎症反应的影响,脂质过氧化,hPDLSCs的铁细胞凋亡和成骨分化能力。总之,结果表明,ATF3沉默抑制炎症和铁性凋亡,通过调节Nrf2/HO-1信号促进LPS诱导的hPDLSCs成骨分化,这可能为牙周炎的治疗提供有希望的治疗靶点。
    Activating transcription factor 3 (ATF3) has been identified as a regulator associated with osteoblast differentiation. However, the effects of ATF3 on the osteogenic differentiation and proliferation of human periodontal stem cells (hPDLSCs) in periodontitis have not been reported. With the purpose of establishing an in vitro model of periodontitis, hPDLSCs were challenged with lipopolysaccharide (LPS). The Cell Counting Kit-8 assay was applied to assess cell viability, while reverse transcription-quantitative PCR and western blotting were employed to detect ATF3 expression. Inflammatory release was assessed using ELISA, together with western blotting. Lipid peroxidation was explored using the C11 BODIPY 581/591 probe, biochemical kits, thiobarbituric acid reactive substances (TBARS) assay and DCFH-DA staining. Iron and Fe2+ levels, and the expression levels of ferroptosis-related proteins were measured using corresponding kits and western blotting. Osteogenic differentiative capability was evaluated using alkaline phosphatase staining, Alizarin red staining and western blotting. The expression levels of proteins associated with Nrf2/HO-1 signaling were identified using western blotting. The results indicated that ATF3 expression was upregulated in LPS-induced hPDLSCs. The knockdown of ATF3 alleviated the LPS-induced inflammatory response in hPDLSCs, together with increased levels of TNF-α, IL-6, IL-1β, Cox-2 and iNOS, and decreased levels of IL-10. ATF3 silencing also led to lower TBARS production rate, and reduced levels of reactive oxygen species, iron, Fe2+, ACSL4 and TFR1, whereas it elevated the levels of SLC7A11 and GPX4. In addition, ATF3 silencing promoted hPDLSC mineralization and cell differentiation, and elevated the levels of OCN2, RUNX2 and BMP2. Additionally, ATF3 depletion upregulated the expression levels of proteins related with Nrf2/HO-1 signaling. The Nrf2 inhibitor ML385 partially counteracted the effects of ATF3 interference on the LPS-challenged inflammatory response, lipid peroxidation, ferroptosis as well as osteogenic differentiative capability in hPDLSCs. In summary, the results revealed that ATF3 silencing suppressed inflammation and ferroptosis, while it improved osteogenic differentiation in LPS-induced hPDLSCs by regulating Nrf2/HO-1 signaling, which may provide promising therapeutic targets for the treatment of periodontitis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    非肥胖糖尿病(NOD)小鼠是一种既定的,自发的1型糖尿病模型,其中糖尿病通过胰岛炎发展。使用下一代测序,加上途径分析,早期胰岛炎的分子指纹图谱在一组4~12周龄的小鼠中绘制.所得的动态时间线显示增殖能力的初始降低,随后在6至8周之间出现炎性特征,其在10至12周之间增加至调节平台。通过激活中枢免疫原性因子如Infg,Il1b,还有Tnfa,和典型炎症信号的激活。对调控景观的分析揭示了转录因子Atf3作为NOD胰岛中炎症信号传导的潜在新型调节剂。此外,Hedgehog信号通路与Atf3调节相关,这表明两者在调节胰岛炎症中起作用;然而,需要进一步的研究来确定这种联系的性质。
    Non-obese diabetes (NOD) mice are an established, spontaneous model of type 1 diabetes in which diabetes develops through insulitis. Using next-generation sequencing, coupled with pathway analysis, the molecular fingerprint of early insulitis was mapped in a cohort of mice ranging from 4 to 12 weeks of age. The resulting dynamic timeline revealed an initial decrease in proliferative capacity followed by the emergence of an inflammatory signature between 6 and 8 weeks that increased to a regulatory plateau between 10 and 12 weeks. The inflammatory signature is identified by the activation of central immunogenic factors such as Infg, Il1b, and Tnfa, and activation of canonical inflammatory signaling. Analysis of the regulatory landscape revealed the transcription factor Atf3 as a potential novel modulator of inflammatory signaling in the NOD islets. Furthermore, the Hedgehog signaling pathway correlated with Atf3 regulation, suggesting that the two play a role in regulating islet inflammation; however, further studies are needed to establish the nature of this connection.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    椎间盘退变(IVDD)严重影响人们的工作和生活质量。我们先前证明沉默激活转录因子3(ATF3)通过调节髓核细胞(NPC)的铁凋亡来阻断IVDD的病理过程,凋亡,炎症,和细胞外基质(ECM)代谢。然而,miR-874-3p是否通过靶向ATF3介导IVDD病理过程尚不清楚.我们进行了单细胞RNA测序(scRNA-seq)和生物信息学分析,以确定ATF3是IVDD中的关键铁性凋亡基因。然后,西方印迹,流式细胞术,ELISA,和动物实验验证miR-874-3p/ATF3信号轴在IVDD中的作用和调控机制。ATF3在IVDD患者和多种细胞类型的IVDD大鼠中高表达,正如scRNA-seq和生物信息学分析所揭示的那样。GO分析揭示了ATF3参与调节细胞凋亡和ECM代谢。此外,我们验证了miR-874-3p可能通过抑制NPC铁性凋亡来抵抗IVDD,凋亡,ECM降解,和靶向ATF3的炎症反应。体内实验显示miR-874-3p/ATF3轴对IVDD的保护作用。这些发现提出了miR-874-3p和ATF3作为IVDD生物标志物的潜力,并表明靶向miR-874-3p/ATF3轴可能是IVDD的治疗靶标。
    Intervertebral disc degeneration (IVDD) severely affects the work and the quality of life of people. We previously demonstrated that silencing activation transcription factor 3 (ATF3) blocked the IVDD pathological process by regulating nucleus pulposus cell (NPC) ferroptosis, apoptosis, inflammation, and extracellular matrix (ECM) metabolism. Nevertheless, whether miR-874-3p mediated the IVDD pathological process by targeting ATF3 remains unclear. We performed single-cell RNA sequencing (scRNA-seq) and bioinformatics analysis to identify ATF3 as a key ferroptosis gene in IVDD. Then, Western blotting, flow cytometry, ELISA, and animal experiments were performed to validate the roles and regulatory mechanisms of miR-874-3p/ATF3 signalling axis in IVDD. ATF3 was highly expressed in IVDD patients and multiple cell types of IVDD rat, as revealed by scRNA-seq and bioinformatics analysis. GO analysis unveiled the involvement of ATF3 in regulating cell apoptosis and ECM metabolism. Furthermore, we verified that miR-874-3p might protect against IVDD by inhibiting NPC ferroptosis, apoptosis, ECM degradation, and inflammatory response by targeting ATF3. In vivo experiments displayed the protective effect of miR-874-3p/ATF3 axis on IVDD. These findings propose the potential of miR-874-3p and ATF3 as biomarkers of IVDD and suggest that targeting the miR-874-3p/ATF3 axis may be a therapeutic target for IVDD.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    铁凋亡是由铁依赖性脂质过氧化触发的程序性细胞死亡的新形式。Brusatol(BRU),一种天然的核因子红系2相关因子2抑制剂,在各种类型的癌症中表现出有效的抗癌作用。然而,BRU治疗肝细胞癌(HCC)的确切机制尚不清楚。使用细胞计数试剂盒-8和集落形成测定和异种移植模型检测BRU在HCC中的抗癌作用。利用RNA测序(RNA-seq)和HCC细胞的生物信息学分析来阐明BRU在HCC中的作用机制。活性氧(ROS)的水平,谷胱甘肽(GSH),丙二醛(MDA),和Fe2+使用测定试剂盒测量。应用RT-qPCR检测转录激活因子3(ATF3)的表达,西方印迹,和免疫荧光染色。使用siATF3检查ATF3在BRU诱导的铁死亡中的作用。BRU显著抑制HCC细胞增殖,在体外和体内。BRU激活了铁凋亡信号通路并增加了ATF3的表达。此外,ATF3敲低阻碍了BRU诱导的铁凋亡。BRU通过ATF3介导的铁凋亡抑制HCC生长,支持BRU作为肝癌有前途的治疗剂。
    Ferroptosis is a novel form of programmed cell death that is triggered by iron-dependent lipid peroxidation. Brusatol (BRU), a natural nuclear factor erythroid 2-related factor 2 inhibitor, exhibits potent anticancer effects in various types of cancer. However, the exact mechanism of BRU in the treatment of hepatocellular carcinoma (HCC) remains unknown. The anticancer effects of BRU in HCC were detected using cell counting kit-8 and colony formation assays and a xenograft model. RNA sequencing (RNA-seq) and bioinformatics analyses of HCC cells were utilized to elucidate the mechanism underlying the effects of BRU in HCC. The levels of reactive oxygen species (ROS), glutathione (GSH), malondialdehyde (MDA), and Fe2+ were measured using assay kits. The expression of activating transcription factor 3 (ATF3) was tested using RT-qPCR, western blotting, and immunofluorescence staining. The role of ATF3 in BRU-induced ferroptosis was examined using siATF3. BRU significantly inhibited HCC cell proliferation, both in vitro and in vivo. BRU activated the ferroptosis signaling pathway and increased ATF3 expression. Furthermore, ATF3 knockdown impeded BRU-induced ferroptosis. BRU suppressed HCC growth through ATF3-mediated ferroptosis, supporting BRU as a promising therapeutic agent for HCC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    LINC00894可能与突触功能有关,但是它在神经细胞中的生物学功能仍然未知。在这项研究中,在HEK-293T和BE(2)-M17(M17)神经母细胞瘤细胞的MTT或CCK-8测定中,LINC00894敲低降低了掺入新合成DNA的Edu和细胞活力。和LINC00894敲低增加细胞凋亡在膜联蛋白V-FITC染色,HEK-293T和M17细胞中活化的Caspase3的表达和活性氧(ROS)的水平。此外,LINC00894还在体外模型中保护细胞免受过氧化氢诱导的细胞凋亡。利用与定量逆转录聚合酶链反应(RT-qPCR)和免疫印迹整合的RNA测序(RNA-seq),我们确定LINC00894影响HEK-293T中激活转录因子3(ATF3)的表达,M17和SH-SY5Y神经母细胞瘤细胞。最后,我们发现ATF3的异位表达可恢复LINC00894下调M17细胞的增殖并抑制细胞凋亡。同时ATF3的敲低也显著增加了LINC00894敲低诱导的M17细胞的细胞活力抑制和凋亡促进。我们的体外模型结果表明,LINC00894可以通过影响ATF3表达来促进神经元细胞增殖并抑制细胞凋亡。
    LINC00894 may be associated with synaptic function, but its biology function in neural cells is still unknown. In this study, LINC00894 knockdown decreased the EdU incorporated into newly synthesized DNA and cell viability in MTT or CCK-8 assay in HEK-293T and BE(2)-M17 (M17) neuroblastoma cells. And LINC00894 knockdown increased cellular apoptosis in Annexin V-FITC staining, the expression of activated Caspase3 and the level of reactive oxygen species (ROS) both in HEK-293T and M17 cells. Moreover, LINC00894 also protected cells from hydrogen peroxide induced apoptosis in in vitro models. Utilizing RNA sequencing (RNA-seq) integrated with quantitative reverse transcription polymerase chain reaction (RT-qPCR) and immunoblot, we identified that LINC00894 affected activating transcription factor 3 (ATF3) expression in HEK-293T, M17, and SH-SY5Y neuroblastoma cells. Finally, we found that ectopic expression of ATF3 restored cell proliferation and inhibited cell apoptosis in LINC00894 downregulated M17 cells. While knockdown of ATF3 also significantly increased the cell viability inhibition and apoptosis promotion induced by LINC00894 knockdown in M17 cells. Our results from in vitro models revealed that LINC00894 could promote neuronal cell proliferation and inhibit cellular apoptosis by affecting ATF3 expression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    心理压力增加胃肠道疾病的风险。然而,应激引起的胃肠道损伤背后的机制尚不清楚。我们研究的目的是阐明应激性胃肠道损伤的推定机制并制定干预策略。为了实现这一点,我们采用束缚应激小鼠模型,研究与小鼠心理应激相关的病理生理变化的方法。通过口服肠道不可吸收的伊文思蓝染料并监测其血浆水平,我们能够追踪活体小鼠胃肠道损伤的进展。此外,流式细胞术用于评估活力,死亡,和脾白细胞的炎症状态,提供对应激诱导的对与应激诱导的胃肠道损伤相关的先天免疫系统的影响的见解。我们的发现表明,中性粒细胞代表负责应激诱导的炎症的主要先天免疫白细胞谱系。脾中性粒细胞显示促炎细胞因子IL-1,细胞活性氧的表达水平升高,线粒体负荷,与其他先天免疫细胞如巨噬细胞相比,应激挑战后的细胞死亡,单核细胞,和树突状细胞。调节的细胞死亡分析表明,NETosis是其他分析的调节的细胞死亡途径中主要的应激诱导的细胞死亡反应。NETosis最终形成和释放中性粒细胞胞外陷阱,通过与病原体结合在调节炎症中起关键作用。用NETosis抑制剂GSK484治疗可挽救应激诱导的中性粒细胞胞外阱释放和胃肠道损伤,强调中性粒细胞胞外陷阱参与应激诱导的胃肠道炎症。我们的结果表明,中性粒细胞NETosis可以作为管理心理压力引起的胃肠道损伤的有希望的药物靶标。
    Psychological stress increases risk of gastrointestinal tract diseases. However, the mechanism behind stress-induced gastrointestinal injury is not well understood. The objective of our study is to elucidate the putative mechanism of stress-induced gastrointestinal injury and develop an intervention strategy. To achieve this, we employed the restraint stress mouse model, a well-established method to study the pathophysiological changes associated with psychological stress in mice. By orally administering gut-nonabsorbable Evans blue dye and monitoring its plasma levels, we were able to track the progression of gastrointestinal injury in live mice. Additionally, flow cytometry was utilized to assess the viability, death, and inflammatory status of splenic leukocytes, providing insights into the stress-induced impact on the innate immune system associated with stress-induced gastrointestinal injury. Our findings reveal that neutrophils represent the primary innate immune leukocyte lineage responsible for stress-induced inflammation. Splenic neutrophils exhibited elevated expression levels of the pro-inflammatory cytokine IL-1, cellular reactive oxygen species, mitochondrial burden, and cell death following stress challenge compared to other innate immune cells such as macrophages, monocytes, and dendritic cells. Regulated cell death analysis indicated that NETosis is the predominant stress-induced cell death response among other analyzed regulated cell death pathways. NETosis culminates in the formation and release of neutrophil extracellular traps, which play a crucial role in modulating inflammation by binding to pathogens. Treatment with the NETosis inhibitor GSK484 rescued stress-induced neutrophil extracellular trap release and gastrointestinal injury, highlighting the involvement of neutrophil extracellular traps in stress-induced gastrointestinal inflammation. Our results suggest that neutrophil NETosis could serve as a promising drug target for managing psychological stress-induced gastrointestinal injuries.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:马兜铃酸肾病(AAN)是由马兜铃酸(AA)引起的快速进行性间质性肾病。AAN与肾病和尿路上皮癌的发展有关。据估计,全球有超过1亿人面临发展AAN的风险。然而,驱动AAN肾脏恶化的潜在机制仍然知之甚少,治疗选择有限。
    方法:我们从与AAN相关的基因表达综合(GEO)获得了GSE27168和GSE136276系列矩阵数据。使用RStudio环境,我们应用limma包和WGCNA包来鉴定共差异表达的基因(co-DEGs)。通过GO/KEGG/GSVA分析,我们揭示了共同的生物学途径。随后,将co-DEG置于String数据库中以构建蛋白质-蛋白质相互作用(PPI)网络。在Cytohubba插件中实现的MCC算法用于识别hub基因。将hub基因与转录因子(TF)数据库交叉引用以鉴定hubTF。利用CIBERSORT进行免疫浸润分析以鉴定关键免疫细胞群。在体内和体外验证了AAN相关hubTF的表达。最后,对两种TFs进行siRNA干预以验证其在AAN中的调节作用。
    结果:我们的分析通过\"limma\"和\"WGCNA\"R包确定了88个共同DEG。构建了一个包含53个节点和34条边的PPI网络,其置信水平>0.4。ATF3和c-JUN被确定为可能与AAN相关的枢纽TFs。此外,ATF3和c-JUN的表达与单核细胞呈正相关,嗜碱性粒细胞,和船只,与嗜酸性粒细胞和内皮细胞呈负相关。我们观察到这两种hubTF的蛋白质和mRNA水平显着增加。此外,发现靶向ATF3而非c-JUN的siRNA干预,减轻AA诱导的细胞损伤。在AAN细胞模型中,ATF3的敲低保护免受氧化应激和炎症。
    结论:这项研究为ATF3在AAN中的作用提供了新的见解。综合分析揭示了分子机制,并确定了AAN治疗的潜在生物标志物和药物靶标。
    BACKGROUND: Aristolochic acid nephropathy (AAN) is a rapidly progressive interstitial nephropathy caused by Aristolochic acid (AA). AAN is associated with the development of nephropathy and urothelial carcinoma. It is estimated that more than 100 million people worldwide are at risk of developing AAN. However, the underlying mechanisms driving renal deterioration in AAN remain poorly understood, and the treatment options are limited.
    METHODS: We obtained GSE27168 and GSE136276 series matrix data from the Gene Expression Omnibus (GEO) related to AAN. Using the R Studio environment, we applied the limma package and WGCNA package to identify co-differently expressed genes (co-DEGs). By GO/KEGG/GSVA analysis, we revealed common biological pathways. Subsequently, co-DEGs were subjected to the String database to construct a protein-protein interaction (PPI) network. The MCC algorithms implemented in the Cytohubba plugin were employed to identify hub genes. The hub genes were cross-referenced with the transcription factor (TF) database to identify hub TFs. Immune infiltration analysis was performed to identify key immune cell groups by utilizing CIBERSORT. The expressions of AAN-associated hub TFs were verified in vivo and in vitro. Finally, siRNA intervention was performed on the two TFs to verify their regulatory effect in AAN.
    RESULTS: Our analysis identified 88 co-DEGs through the \"limma\" and \"WGCNA\" R packages. A PPI network comprising 53 nodes and 34 edges was constructed with a confidence level >0.4. ATF3 and c-JUN were identified as hub TFs potentially linked to AAN. Additionally, expressions of ATF3 and c-JUN positively correlated with monocytes, basophils, and vessels, and negatively correlated with eosinophils and endothelial cells. We observed a significant increase in protein and mRNA levels of these two hub TFs. Furthermore, it was found that siRNA intervention targeting ATF3, but not c-JUN, alleviated cell damage induced by AA. The knockdown of ATF3 protects against oxidative stress and inflammation in the AAN cell model.
    CONCLUSIONS: This study provides novel insights into the role of ATF3 in AAN. The comprehensive analysis sheds light on the molecular mechanisms and identifies potential biomarkers and drug targets for AAN treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    使癌细胞在葡萄糖剥夺下存活的压力适应性机制仍然难以捉摸。N6-甲基腺苷(m6A)修饰在决定癌细胞命运和细胞对营养缺乏的应激反应中起着重要作用。然而,在葡萄糖剥夺下,m6A修饰是否在调节癌细胞存活中起作用尚不清楚。这里,我们发现葡萄糖剥夺降低了m6A修饰水平.增加m6A修饰导致葡萄糖剥夺下肝癌细胞坏死增加,而减少m6A修饰具有相反的效果。整合的m6A-seq和RNA-seq揭示了葡萄糖剥夺下m6A修饰的潜在靶标,包括转录因子FOSL1;进一步,葡萄糖剥夺通过减少外显子1和5'-UTR区域的m6A修饰以m6A-YTHDF2依赖性方式抑制FOSL1mRNA衰减来上调FOSL1。功能上,FOSL1在体外和体内保护肝癌细胞免受葡萄糖剥夺诱导的坏死。机械上,FOSL1通过与其启动子结合而转录抑制ATF3。同时,ATF3和MAFF通过它们的亮氨酸拉链结构域相互作用形成异二聚体,与NRF2竞争结合NRF2靶基因启动子中的抗氧化反应元件,从而抑制它们的转录。因此,FOSL1减少了ATF3-MAFF异二聚体的形成,从而增强NRF2转录活性和葡萄糖剥夺肝癌细胞的抗氧化能力。因此,FOSL1减轻了葡萄糖剥夺诱导的活性氧积累的坏死诱导作用。总的来说,我们的研究揭示了m6A-FOSL1-ATF3轴在葡萄糖剥夺下肝癌细胞坏死中的保护作用,并可能为癌症治疗提供新的靶点。
    Stress-adaptive mechanisms enabling cancer cells to survive under glucose deprivation remain elusive. N6-methyladenosine (m6A) modification plays important roles in determining cancer cell fate and cellular stress response to nutrient deficiency. However, whether m6A modification functions in the regulation of cancer cell survival under glucose deprivation is unknown. Here, we found that glucose deprivation reduced m6A modification levels. Increasing m6A modification resulted in increased hepatoma cell necrosis under glucose deprivation, whereas decreasing m6A modification had an opposite effect. Integrated m6A-seq and RNA-seq revealed potential targets of m6A modification under glucose deprivation, including the transcription factor FOSL1; further, glucose deprivation upregulated FOSL1 by inhibiting FOSL1 mRNA decay in an m6A-YTHDF2-dependent manner through reducing m6A modification in its exon1 and 5\'-UTR regions. Functionally, FOSL1 protected hepatoma cells against glucose deprivation-induced necrosis in vitro and in vivo. Mechanistically, FOSL1 transcriptionally repressed ATF3 by binding to its promoter. Meanwhile, ATF3 and MAFF interacted via their leucine zipper domains to form a heterodimer, which competed with NRF2 for binding to antioxidant response elements in the promoters of NRF2 target genes, thereby inhibiting their transcription. Consequently, FOSL1 reduced the formation of the ATF3-MAFF heterodimer, thereby enhancing NRF2 transcriptional activity and the antioxidant capacity of glucose-deprived-hepatoma cells. Thus, FOSL1 alleviated the necrosis-inducing effect of glucose deprivation-induced reactive oxygen species accumulation. Collectively, our study uncovers the protective role of m6A-FOSL1-ATF3 axis in hepatoma cell necrosis under glucose deprivation, and may provide new targets for cancer therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号