KCTD6

  • 文章类型: Journal Article
    最近的研究表明,特应性皮炎(AD)中激活的焦凋亡会切换炎症过程,并导致异常的角化和表皮屏障功能障碍。很少有研究关注焦亡相关基因与人角质形成细胞分化之间的相互作用机制。
    来自基因表达综合(GEO)的AD数据集用于鉴定不同表达的焦亡相关基因(DEPRG)。鉴定Hub基因并进行富集分析以选择上皮发育相关基因。通过免疫组织化学(IHC)检测AD患者的病变以验证hub基因。人角质形成细胞系,gasderminD(GSDMD)过表达,Caspase1siRNA,组蛋白去乙酰化酶1(HDAC1)siRNA,和HDAC1过表达载体用于功能获得和丧失实验。通过qPCR确定角质化蛋白的调节,蛋白质印迹(WB),免疫荧光(IF),双荧光素酶报告分析,免疫共沉淀(Co-IP),和染色质免疫沉淀(ChIP)。
    在特应性皮炎非病灶性皮肤(ANL)和健康对照(HC)或特应性皮炎病灶性皮肤(AL)和HC之间共鉴定出27个DEPRG。富集分析显示这些DEPRG主要富集在炎症反应和角质形成细胞分化中。在通过蛋白质-蛋白质相互作用网络鉴定的10个hub基因中,只有GSDMD与上皮紧密连接核心基因的表达呈统计学负相关。此外,GSDMD在AD病变中上调,并通过减少聚丝团蛋白(FLG)的表达来抑制人角质形成细胞的分化。机械上,由Caspase1激活的GSDMD通过HDAC1降低FLG表达。HDAC1通过减少FLG启动子处的组蛋白乙酰化来降低FLG表达。此外,GSDMD阻断了钾通道四聚化域6(KCTD6)与HDAC1的相互作用,以抑制HDAC1的降解。
    这项研究表明,GSDMD在AD病变中上调,并且GSDMD通过表观遗传修饰调节角质形成细胞,这可能为AD提供潜在的治疗靶点。
    UNASSIGNED: Recent studies have shown that activated pyroptosis in atopic dermatitis (AD) switches inflammatory processes and causes abnormal cornification and epidermal barrier dysfunction. Little research has focused on the interaction mechanism between pyroptosis-related genes and human keratinocyte differentiation.
    UNASSIGNED: The AD dataset from the Gene Expression Omnibus (GEO) was used to identify differently expressed pyroptosis-related genes (DEPRGs). Hub genes were identified and an enrichment analysis was performed to select epithelial development-related genes. Lesions of AD patients were detected via immunohistochemistry (IHC) to verify the hub gene. Human keratinocytes cell lines, gasdermin D (GSDMD) overexpression, Caspase1 siRNA, Histone Deacetylase1 (HDAC1) siRNA, and HDAC1 overexpression vectors were used for gain-and-loss-of-function experiments. Regulation of cornification protein was determined by qPCR, western blot (WB), immunofluorescence (IF), dual-luciferase reporter assay, co-immunoprecipitation (Co-IP), and chromatin immunoprecipitation (ChIP).
    UNASSIGNED: A total of 27 DEPRGs were identified between either atopic dermatitis non-lesional skin (ANL) and healthy control (HC) or atopic dermatitis lesional skin (AL) and HC. The enrichment analysis showed that these DEPRGs were primarily enriched in the inflammatory response and keratinocytes differentiation. Of the 10 hub genes identified via the protein-protein interaction network, only GSDMD was statistically and negatively associated with the expression of epithelial tight junction core genes. Furthermore, GSDMD was upregulated in AD lesions and inhibited human keratinocyte differentiation by reducing filaggrin (FLG) expression. Mechanistically, GSDMD activated by Caspase1 reduced FLG expression via HDAC1. HDAC1 decreased FLG expression by reducing histone acetylation at the FLG promoter. In addition, GSDMD blocked the interaction of Potassium Channel Tetramerization Domain Containing 6 (KCTD6) and HDAC1 to prohibit HDAC1 degradation.
    UNASSIGNED: This study revealed that GSDMD was upregulated in AD lesions and that GSDMD regulated keratinocytes via epigenetic modification, which might provide potential therapeutic targets for AD.
    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
    SonicHedgehog(Hh)信号转导通路在许多发育过程中起着关键作用,当放松管制时,可能会导致几种癌症,包括基底细胞癌,髓母细胞瘤,结直肠,前列腺,还有胰腺癌.近年来,已经开发了几种Hh抑制剂,主要作用于Smo受体。然而,由于Smo突变或非规范的Hh途径激活引起的耐药性突出了确定Hh途径调节的进一步机制的需要。其中,组蛋白去乙酰化酶HDAC1对Hh转录因子Gli1的去乙酰化增加了Hh活性。在另一端,KCASH家族的抑癌因子结合HDAC1,导致其泛素化和随后的蛋白酶体降解,离开Gli1乙酰化和无活性。最近证明,钾通道含有蛋白KCTD15能够与KCASH2蛋白相互作用并使其稳定,增强其对HDAC1和Hh通路的作用。KCTD15和KCTD1蛋白具有高度同源性,并聚集在特定的KCTD亚家族中。我们在这里描述了KCTD1在Hh途径上的作用。因此,我们证明了KCTD1与KCASH1和KCASH2蛋白的相互作用,以及它通过减少它们的泛素化和蛋白酶体介导的降解在它们稳定中的作用。因此,KCTD1表达降低HDAC1蛋白水平和Hh/Gli1活性,抑制Hh肿瘤细胞中的Hh依赖性细胞增殖。此外,对公开数据库的表达数据的分析表明,在Hh依赖性MB样品中KCTD1表达降低,与正常的小脑相比,这表明KCTD1可能是针对Hh依赖性肿瘤治疗方法的新的推定靶标。
    The Sonic Hedgehog (Hh) signal transduction pathway plays a critical role in many developmental processes and, when deregulated, may contribute to several cancers, including basal cell carcinoma, medulloblastoma, colorectal, prostate, and pancreatic cancer. In recent years, several Hh inhibitors have been developed, mainly acting on the Smo receptor. However, drug resistance due to Smo mutations or non-canonical Hh pathway activation highlights the need to identify further mechanisms of Hh pathway modulation. Among these, deacetylation of the Hh transcription factor Gli1 by the histone deacetylase HDAC1 increases Hh activity. On the other end, the KCASH family of oncosuppressors binds HDAC1, leading to its ubiquitination and subsequent proteasomal degradation, leaving Gli1 acetylated and not active. It was recently demonstrated that the potassium channel containing protein KCTD15 is able to interact with KCASH2 protein and stabilize it, enhancing its effect on HDAC1 and Hh pathway. KCTD15 and KCTD1 proteins share a high homology and are clustered in a specific KCTD subfamily. We characterize here KCTD1 role on the Hh pathway. Therefore, we demonstrated KCTD1 interaction with KCASH1 and KCASH2 proteins, and its role in their stabilization by reducing their ubiquitination and proteasome-mediated degradation. Consequently, KCTD1 expression reduces HDAC1 protein levels and Hh/Gli1 activity, inhibiting Hh dependent cell proliferation in Hh tumour cells. Furthermore, analysis of expression data on publicly available databases indicates that KCTD1 expression is reduced in Hh dependent MB samples, compared to normal cerebella, suggesting that KCTD1 may represent a new putative target for therapeutic approaches against Hh-dependent tumour.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号