ALKBH5

ALKBH5
  • 文章类型: Journal Article
    背景:细胞外基质(ECM)积累升高是纤维化疾病发病机理的主要促成因素。最近的研究表明,N6-甲基腺苷(m6A)RNA修饰在调节RNA稳定性中起关键作用,并有助于各种病理状况的引发。Howbeit,m6A影响ECM沉积的确切机制尚不清楚。
    方法:在本研究中,我们使用肥厚性瘢痕(HTSs)作为研究ECM相关疾病的范例。我们专注于ALKBH5介导的m6A去甲基化在HTSs病理进展中的作用,并检查了其与临床分期的相关性。在体内和体外研究了ALKBH5消融对ECM成分的影响。ALKBH5的下游靶标及其潜在机制,使用集成高通量分析确定,RNA结合蛋白免疫沉淀和RNA下拉测定。此外,在纤维化瘢痕模型中评估外源性ALKBH5过表达的治疗潜力.
    结果:ALKBH5在来自HTS病变的成纤维细胞中降低,并且与其临床分期呈负相关。重要的是,ALKBH5的消融促进COL3A1、COL1A1和ELN的表达,导致体内和体外ECM的病理沉积和重建。从治疗的角度来看,ALKBH5的外源性过表达可显著抑制纤维化瘢痕模型中胶原的异常沉积。根据集成的高通量分析确定,包括COL3A1、COL1A1和ELN在内的关键ECM组件是ALKBH5的直接下游目标。通过其机制,ALKBH5通过从mRNA中去除m6A抑制COL3A1、COL1A1和ELN的表达,从而以依赖YTHDF1的方式降低它们的稳定性。
    结论:我们的研究确定ALKBH5是病理性ECM沉积的内源性抑制因子,有助于开发针对HTS的重新编程的M6A靶向治疗。
    BACKGROUND: Elevated extracellular matrix (ECM) accumulation is a major contributing factor to the pathogenesis of fibrotic diseases. Recent studies have indicated that N6-methyladenosine (m6A) RNA modification plays a pivotal role in modulating RNA stability and contribute to the initiation of various pathological conditions. Howbeit, the precise mechanism by which m6A influences ECM deposition remains unclear.
    METHODS: In this study, we used hypertrophic scars (HTSs) as a paradigm to investigate ECM-related diseases. We focused on the role of ALKBH5-mediated m6A demethylation within the pathological progression of HTSs and examined its correlation with clinical stages. The effects of ALKBH5 ablation on ECM components were studied both in vivo and in vitro. Downstream targets of ALKBH5, along with their underlying mechanisms, were identified using integrated high-throughput analysis, RNA-binding protein immunoprecipitation and RNA pull-down assays. Furthermore, the therapeutic potential of exogenous ALKBH5 overexpression was evaluated in fibrotic scar models.
    RESULTS: ALKBH5 was decreased in fibroblasts derived from HTS lesions and was negatively correlated with their clinical stages. Importantly, ablation of ALKBH5 promoted the expression of COL3A1, COL1A1, and ELN, leading to pathological deposition and reconstruction of the ECM both in vivo and in vitro. From a therapeutic perspective, the exogenous overexpression of ALKBH5 significantly inhibited abnormal collagen deposition in fibrotic scar models. As determined by integrated high-throughput analysis, key ECM components including COL3A1, COL1A1, and ELN are direct downstream targets of ALKBH5. By means of its mechanism, ALKBH5 inhibits the expression of COL3A1, COL1A1, and ELN by removing m6A from mRNAs, thereby decreasing their stability in a YTHDF1-dependent manner.
    CONCLUSIONS: Our study identified ALKBH5 as an endogenous suppressor of pathological ECM deposition, contributing to the development of a reprogrammed m6A-targeted therapy for HTSs.
    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
    暂无摘要。
    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
    ALKBH5是参与RNAm6A修饰调控的去甲基酶之一。除了其在RNAm6A修饰的动态调控中的作用外,近年来发现ALKBH5在各种组织纤维化过程中发挥重要作用。然而,ALKBH5在纤维化中的机制和作用的报道不一致.多种细胞类型,包括实质细胞,免疫细胞(中性粒细胞和T细胞),巨噬细胞,内皮细胞,和成纤维细胞,在纤维化的各个阶段发挥作用。因此,这篇综述分析了ALKBH5调节这些细胞的机制,它对他们功能的影响,和纤维化的结果。此外,本文综述了ALKBH5在肺纤维化等纤维化疾病中的作用,肝纤维化,心脏纤维化,和肾脏纤维化,并讨论了迄今为止发现的各种ALKBH5抑制剂,探讨ALKBH5作为纤维化临床靶点的潜力。
    ALKBH5 is one of the demethylases involved in the regulation of RNA m6A modification. In addition to its role in the dynamic regulation of RNA m6A modification, ALKBH5 has been found to play important roles in various tissues fibrosis processes in recent years. However, the mechanisms and effects of ALKBH5 in fibrosis have been reported inconsistently. Multiple cell types, including parenchymal cells, immune cells (neutrophils and T cells), macrophages, endothelial cells, and fibroblasts, play roles in various stages of fibrosis. Therefore, this review analyzes the mechanisms by which ALKBH5 regulates these cells, its impact on their functions, and the outcomes of fibrosis. Furthermore, this review summarizes the role of ALKBH5 in fibrotic diseases such as pulmonary fibrosis, liver fibrosis, cardiac fibrosis, and renal fibrosis, and discusses various ALKBH5 inhibitors that have been discovered to date, exploring the potential of ALKBH5 as a clinical target for fibrosis.
    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
    翻译是细胞应激靶向的主要基因表达步骤之一,通常称为平移应力,其中包括用抗癌药物治疗。虽然翻译应激阻断大量mRNA的翻译起始,尽管如此,它激活被称为短上游开放阅读框(uORFs)-mRNA的特定mRNA的翻译。其中,ATF4mRNA编码一种转录因子,该转录因子可重新编程响应各种应激的细胞中的基因表达。尽管ATF4mRNA的应激诱导翻译依赖于uORFs的存在(主要ATF4ORF的上游),介导这种效应的机制,特别是在化学抗性期间,仍然难以捉摸。这里,我们报告ALKBH5(AlkB同系物5)和FTO(FTO:脂肪量和肥胖相关蛋白),两种RNA去甲基化酶,在用化疗药物索拉非尼处理的转化肝细胞系(Hep3B)中促进ATF4mRNA的翻译。使用体外荧光素酶报告基因翻译测定法,我们发现两种酶的消耗减少了药物治疗后报告分子ATF4mRNA的翻译。始终如一,如通过与RT-qPCR偶联的多核糖体测定所评估的,任一蛋白质的消耗消除了ATF3mRNA加载到翻译核糖体中。总的来说,这些结果表明,ALKBH5和FTO介导的ATF4mRNA的翻译在其起始步骤受到调节。使用体外甲基化测定,我们发现,ALKBH5是抑制报告基因ATF4mRNA在uORF2保守腺苷(A235)位点甲基化所必需的,这表明ALKBH5介导的ATF4mRNA翻译涉及其A235的去甲基化.通过将A/G突变引入ATF4mRNA报告基因来防止A235甲基化,使其翻译对ALKBH5缺失不敏感,支持ALKBH5去甲基化活性在翻译中的作用。最后,靶向ALKBH5或FTO使Hep3B对索拉非尼诱导的细胞死亡敏感,有助于他们的抵抗。总之,我们的数据显示,ALKBH5和FTO是促进索拉非尼治疗耐药的新因素,部分通过介导ATF4mRNA的翻译。
    Translation is one of the main gene expression steps targeted by cellular stress, commonly referred to as translational stress, which includes treatment with anticancer drugs. While translational stress blocks the translation initiation of bulk mRNAs, it nonetheless activates the translation of specific mRNAs known as short upstream open reading frames (uORFs)-mRNAs. Among these, the ATF4 mRNA encodes a transcription factor that reprograms gene expression in cells responding to various stresses. Although the stress-induced translation of the ATF4 mRNA relies on the presence of uORFs (upstream to the main ATF4 ORF), the mechanisms mediating this effect, particularly during chemoresistance, remain elusive. Here, we report that ALKBH5 (AlkB Homolog 5) and FTO (FTO: Fat mass and obesity-associated protein), the two RNA demethylating enzymes, promote the translation of ATF4 mRNA in a transformed liver cell line (Hep3B) treated with the chemotherapeutic drug sorafenib. Using the in vitro luciferase reporter translational assay, we found that depletion of both enzymes reduced the translation of the reporter ATF4 mRNA upon drug treatment. Consistently, depletion of either protein abrogates the loading of the ATF3 mRNA into translating ribosomes as assessed by polyribosome assays coupled to RT-qPCR. Collectively, these results indicate that the ALKBH5 and FTO-mediated translation of the ATF4 mRNA is regulated at its initiation step. Using in vitro methylation assays, we found that ALKBH5 is required for the inhibition of the methylation of a reporter ATF4 mRNA at a conserved adenosine (A235) site located at its uORF2, suggesting that ALKBH5-mediated translation of ATF4 mRNA involves demethylation of its A235. Preventing methylation of A235 by introducing an A/G mutation into an ATF4 mRNA reporter renders its translation insensitive to ALKBH5 depletion, supporting the role of ALKBH5 demethylation activity in translation. Finally, targeting either ALKBH5 or FTO sensitizes Hep3B to sorafenib-induced cell death, contributing to their resistance. In summary, our data show that ALKBH5 and FTO are novel factors that promote resistance to sorafenib treatment, in part by mediating the translation of ATF4 mRNA.
    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
    N6-甲基腺苷(m6A)由甲基转移酶(称为“作家”)和去甲基酶(称为“橡皮擦”)动态调节,促进可逆调制。m6A水平的变化显著影响细胞功能,比如从细胞核输出RNA,mRNA代谢,蛋白质合成,和RNA剪接。它们与一系列病理密切相关。此外,m6A调节失调已成为许多疾病的有希望的治疗靶标。M6A在控制重要的下游分子和关键的生物途径中起着关键作用,有助于许多条件的发病机理和进化。这篇综述概述了m6A去甲基酶,明确详述FTO和ALKBH5的结构和功能特征。此外,我们探索他们在各种疾病中的独特参与,检查调节其表达的因素,并讨论了抑制剂开发的进展。
    N6-methyladenosine (m6A) is dynamically regulated by methyltransferases (termed \"writers\") and demethylases (referred to as \"erasers\"), facilitating a reversible modulation. Changes in m6A levels significantly influence cellular functions, such as RNA export from the nucleus, mRNA metabolism, protein synthesis, and RNA splicing. They are intricately associated with a spectrum of pathologies. Moreover, dysregulation of m6A modulation has emerged as a promising therapeutic target across many diseases. m6A plays a pivotal role in controlling vital downstream molecules and critical biological pathways, contributing to the pathogenesis and evolution of numerous conditions. This review provides an overview of m6A demethylases, explicitly detailing the structural and functional characteristics of FTO and ALKBH5. Additionally, we explore their distinct involvement in various diseases, examine factors regulating their expression, and discuss the progress in inhibitor development.
    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
    胃肠道肿瘤是人类最常见的恶性肿瘤,常伴有预后不良。N6-甲基腺苷(m6A)修饰作为最丰富的RNA修饰广泛存在于真核细胞中。它在RNA剪接和加工中起着至关重要的作用,核出口,翻译,和稳定性。人AlkB同源物5(ALKBH5)是一种在各种胃肠道癌症中表现出异常表达的RNA脱甲基酶。它与肿瘤发生密切相关,扩散,迁移,和胃肠道癌症的其他生物学功能。然而,近年来研究表明,ALKBH5在胃肠道肿瘤中的作用和机制是复杂的,甚至存在争议。因此,本文综述了ALKBH5作为肿瘤抑制因子或启动子在胃肠道肿瘤中的作用。它检查了ALKBH5的生物学功能及其作为治疗靶标的潜力,为胃肠道肿瘤研究提供新的视角和见解。
    Gastrointestinal cancer is the most common malignancy in humans, often accompanied by poor prognosis. N6-methyladenosine (m6A) modification is widely present in eukaryotic cells as the most abundant RNA modification. It plays a crucial role in RNA splicing and processing, nuclear export, translation, and stability. Human AlkB homolog 5 (ALKBH5) is a type of RNA demethylase exhibiting abnormal expression in various gastrointestinal cancers.It is closely related to the tumorigenesis, proliferation, migration, and other biological functions of gastrointestinal cancer. However, recent studies indicated that the role and mechanism of ALKBH5 in gastrointestinal cancer are complicated and even controversial. Thus, this review summarizes recent advances in elucidating the role of ALKBH5 as a tumor suppressor or promoter in gastrointestinal cancer. It examines the biological functions of ALKBH5 and its potential as a therapeutic target, providing new perspectives and insights for gastrointestinal cancer research.
    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
    肾小管上皮细胞(RTEC)凋亡失调是肾结石发生发展的关键步骤之一。尽管N6-甲基腺苷(m6A)修饰已被广泛研究并与各种病理过程相关,关于其在RTEC损伤和凋亡中的具体作用的研究仍然有限。在这项研究中,我们发现ALKBH5的过表达降低了RTEC细胞中m6A修饰的水平,并显着促进了RTEC细胞凋亡。进一步的机制研究表明,ALKBH5主要降低RTEC中粘蛋白1(MUC1)基因mRNA的m6A水平。此外,ALKBH5损害了MUC1mRNA在RTECs中的稳定性,导致MUC1的表达减弱。最后,我们确定ALKBH5-MUC1轴主要通过调节PI3K/Akt信号通路促进RTEC细胞凋亡。这项研究揭示了ALKBH5-MUC1-PI3K/Akt调节系统在RTEC细胞凋亡中的关键作用,并为治疗肾结石提供了新的治疗靶点。
    Dysregulation of renal tubular epithelial cell (RTEC) apoptosis is one of the critical steps underlying the occurrence and development of nephrolithiasis. Although N6-methyladenosine (m6A) modification has been extensively studied and associated with various pathologic processes, research on its specific role in RTEC injury and apoptosis remains limited. In this study, we found that overexpression of ALKBH5 reduced the level of m6A modification in RTEC cells and notably promoted RTEC apoptosis. Further mechanism studies revealed that ALKBH5 mainly  decreased the m6A level on the mRNA of  Mucin 1 (MUC1) gene in RTECs. Moreover, ALKBH5  impaired the stability of MUC1 mRNA in RTECs, leading to  attenuated expression of MUC1. Finally, we determined that the ALKBH5-MUC1 axis primarily facilitated RTEC apoptosis by regulating the PI3K/Akt signaling pathway. This study revealed the critical role of the ALKBH5-MUC1-PI3K/Akt regulatory system in RTEC apoptosis and provided new therapeutic targets for treating nephrolithiasis.
    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
    SMARCA5,SWI/SNF家族中的一种蛋白质,先前已通过甲基化与溃疡性结肠炎(UC)的发展有关。然而,SMARCA5促进结肠炎症和Th17和Treg细胞失衡的具体分子机制尚不清楚.本研究旨在探索这些分子机制。采用葡聚糖硫酸钠诱导建立UC小鼠模型,然后测量老鼠的体重,疾病活动指数(DAI)评分,结肠长度,结肠的病理变化,和FITC-葡聚糖浓度。IL-17a的水平,IFN-γ,IL-6,TNF-α,TGF-β,和IL-10被测量,随着ZO-1和Occludin蛋白的表达。流式细胞术用于评估UC小鼠脾脏和肠系膜淋巴结中IL-17+CD4+(Th17+)细胞和FOXP3+CD25+CD4+(Treg+)细胞的存在。我们观察到SMARCA5和RNF180增加,而ALKBH5在UC小鼠结肠组织中下调。SMARCA5或RNF180敲低或ALKBH5过表达改善UC小鼠结肠炎症和Th17/Treg细胞失衡,体重增加,结肠长度,FOXP3+CD25+CD4+T细胞,ZO-1,Occludin的水平,TGF-β,IL-10和FOXP3。它降低了DAI分数,IL-17+CD4+T细胞,和IL-17a的水平,IFN-γ,IL-6,TNF-α,和ROR-γt。ALKBH5通过m6A修饰抑制SMARCA5表达,而RNF180通过泛素化降低ALKBH5表达。我们的发现表明,RNF180通过调节ALKBH5/SMARCA5轴加重UC小鼠的结肠炎症和Th17/Treg细胞失衡。
    SMARCA5, a protein in the SWI/SNF family, has been previously implicated in the development of ulcerative colitis (UC) through methylation. However, the specific molecular mechanisms by which SMARCA5 contributes to colonic inflammation and the imbalance between Th17 and Treg cells remain unclear. This study was designed to explore these molecular mechanisms. A UC mouse model was established using dextran sulfate sodium induction, followed by measurements of mouse weight, disease activity index (DAI) score, colon length, pathological changes in the colon, and FITC-dextran concentration. The levels of IL-17a, IFN-γ, IL-6, TNF-α, TGF-β, and IL-10 were measured, along with the protein expression of ZO-1 and Occludin. Flow cytometry was used to assess the presence of IL-17 + CD4 + (Th17 +) cells and FOXP3 + CD25 + CD4 + (Treg +) cells in the spleen and mesenteric lymph nodes of UC mice. We observed that SMARCA5 and RNF180 were increased, while ALKBH5 was downregulated in UC mouse colon tissue. SMARCA5 or RNF180 knockdown or ALKBH5 overexpression ameliorated the colon inflammation and Th17/Treg cell imbalance in UC mice, shown by increased body weight, colon length, FOXP3 + CD25 + CD4 + T cells, and the levels of ZO-1, Occludin, TGF-β, IL-10, and FOXP3. It decreased DAI scores, IL-17 + CD4 + T cells, and levels of IL-17a, IFN-γ, IL-6, TNF-α, and ROR-γt. ALKBH5 inhibited SMARCA5 expression via m6A modification, while RNF180 reduced ALKBH5 expression via ubiquitination. Our findings indicate that RNF180 aggravated the colon inflammation and Th17/Treg cell imbalance in UC mice by regulating the ALKBH5/SMARCA5 axis.
    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
    N-甲基腺苷(m6A)代表在结直肠癌中观察到的普遍RNA修饰。尽管它丰富,m6A甲基化对lncRNACARMN的生物学意义在结直肠癌中仍然难以捉摸,尤其是突变型p53的功能获得。这里,我们阐明了CARMN在突变型p53的结直肠癌患者中表现出降低的表达水平,这归因于其丰富的m6A甲基化,促进癌症增殖,在体外和体内的侵袭和转移。进一步的研究表明,ALKBH5作为CARMN的直接脱甲基酶,靶向477个甲基化位点,从而保持CARMN表达。然而,突变型p53与ALKBH5启动子的相互作用阻碍了其转录,增强CARMN上的m6A甲基化水平。随后,YTHDF2/YTHDF3识别并降解m6A修饰的CARMN。同时,过表达CARMN在体外和体内显着抑制结直肠癌的进展。此外,miR-5683被鉴定为lncRNACARMN的直接下游靶标,通过协同下调FGF2表达发挥抗肿瘤作用。我们的发现揭示了CARMN在突变型p53结直肠癌中的调节因子和功能机制,可能为癌症诊断和治疗的基于去甲基化的策略提供了见解。CARMN的m6A甲基化是突变型p53功能获得诱导的结直肠癌恶性进展的主要因素,确定一种有希望的癌症治疗方法。
    N-methyladenosine (m6A) represents a prevalent RNA modification observed in colorectal cancer. Despite its abundance, the biological implications of m6A methylation on the lncRNA CARMN remain elusive in colorectal cancer, especially for mutant p53 gain-of-function. Here, we elucidate that CARMN exhibits diminished expression levels in colorectal cancer patients with mutant p53, attributed to its rich m6A methylation, which promotes cancer proliferation, invasion and metastasis in vitro and in vivo. Further investigation illustrates that ALKBH5 acts as a direct demethylase of CARMN, targeting 477 methylation sites, thereby preserving CARMN expression. However, the interaction of mutant p53 with the ALKBH5 promoter impedes its transcription, enhancing m6A methylation levels on CARMN. Subsequently, YTHDF2/YTHDF3 recognise and degrade m6A-modified CARMN. Concurrently, overexpressing CARMN significantly suppressed colorectal cancer progression in vitro and in vivo. Additionally, miR-5683 was identified as a direct downstream target of lncRNA CARMN, exerting an antitumour effect by cooperatively downregulating FGF2 expression. Our findings revealed the regulator and functional mechanism of CARMN in colorectal cancer with mutant p53, potentially offering insights into demethylation-based strategies for cancer diagnosis and therapy. The m6A methylation of CARMN that is prime for mutant p53 gain-of-function-induced malignant progression of colorectal cancer, identifying a promising approach for cancer therapy.
    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
    血管内皮细胞稳态失衡是许多血管疾病进展的关键机制。RNA修饰,特别是N6-甲基腺苷(M6A),在许多生物过程中起着重要的作用。然而,m6ARNA甲基化在内皮功能障碍中的调节功能仍未得到充分表征。在这项研究中,我们确定m6A甲基转移酶METTL3对于调节内皮功能至关重要.功能上,METTL3耗竭导致内皮细胞增殖减少,生存和炎症反应。相反,METTL3的过表达引起相反的效果。机械上,MeRIP-seq确定METTL3催化TRAF1mRNA的m6A修饰并增强TRAF1翻译,从而上调TRAF1蛋白。TRAF1的过表达成功地挽救了由于METTL3敲低引起的内皮细胞增殖和粘附的抑制。此外,m6A甲基化介导的TRAF1表达可以被去甲基酶ALKBH5逆转。ALKBH5的敲除上调了m6A的水平和TRAF1的蛋白水平,也增加了内皮细胞的粘附和炎症反应。总的来说,我们的研究结果表明,METTL3通过TRAF1m6A修饰调节血管内皮稳态,提示靶向METTL3-m6A-TRAF1轴可能对血管疾病患者具有治疗潜力.
    The imbalance of vascular endothelial cell homeostasis is the key mechanism for the progression of many vascular diseases. RNA modification, particularly N6-Methyladenosine (m6A), plays important function in numerous biological processes. Nevertheless, the regulatory function of m6A RNA methylation in endothelial dysfunction remains insufficiently characterized. In this study, we established that the m6A methyltransferase METTL3 is critical for regulating endothelial function. Functionally, depletion of METTL3 results in decreased endothelial cells proliferation, survival and inflammatory response. Conversely, overexpression of METTL3 elicited the opposite effects. Mechanistically, MeRIP-seq identified that METTL3 catalyzed m6A modification of TRAF1 mRNA and enhanced TRAF1 translation, thereby up-regulation of TRAF1 protein. Over-expression of TRAF1 successfully rescued the inhibition of proliferation and adhesion of endothelial cells due to METTL3 knockdown. Additionally, m6A methylation-mediated TRAF1 expression can be reversed by the demethylase ALKBH5. Knockdown of ALKBH5 upregulated the level of m6A and protein level of TRAF1, and also increased endothelial cells adhesion and inflammatory response. Collectively, our findings suggest that METTL3 regulates vascular endothelium homeostasis through TRAF1 m6A modification, suggesting that targeting the METTL3-m6A-TRAF1 axis may hold therapeutic potential for patients with vascular diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号