Interferon Regulatory Factor-7

干扰素调节因子 - 7
  • 文章类型: Journal Article
    破骨细胞是多核骨吸收细胞,它们的形成受到严格的调节,以防止过度的骨质流失。然而,破骨细胞形成受限的机制仍未完全确定.这里,我们发现甾醇调节元件结合蛋白2(SREBP2)作为破骨细胞形成和炎性骨丢失的负调节因子。胆固醇和SREBP2,胆固醇生物合成的关键转录因子,在破骨细胞形成的晚期增加。SREBP2在骨髓细胞中的消融导致体内和体外破骨细胞生成增加,导致骨量低。此外,在小鼠炎性骨溶解和关节炎模型中,SREBP2的缺失加速了炎性骨破坏。SREBP2介导的破骨细胞生成的调节独立于其在胆固醇生物合成中的典型功能,但是介导的,在某种程度上,通过它的下游目标,干扰素调节因子7(IRF7)。一起来看,我们的研究强调了SREBP2-IRF7调节回路在破骨细胞分化中作为负反馈回路的以前未描述的作用,并代表了抑制病理性骨破坏的新机制.
    Osteoclasts are multinucleated bone-resorbing cells, and their formation is tightly regulated to prevent excessive bone loss. However, the mechanisms by which osteoclast formation is restricted remain incompletely determined. Here, we found that sterol regulatory element binding protein 2 (SREBP2) functions as a negative regulator of osteoclast formation and inflammatory bone loss. Cholesterols and SREBP2, a key transcription factor for cholesterol biosynthesis, increased in the late phase of osteoclastogenesis. The ablation of SREBP2 in myeloid cells resulted in increased in vivo and in vitro osteoclastogenesis, leading to low bone mass. Moreover, deletion of SREBP2 accelerated inflammatory bone destruction in murine inflammatory osteolysis and arthritis models. SREBP2-mediated regulation of osteoclastogenesis is independent of its canonical function in cholesterol biosynthesis but is mediated, in part, by its downstream target, interferon regulatory factor 7 (IRF7). Taken together, our study highlights a previously undescribed role of the SREBP2-IRF7 regulatory circuit as a negative feedback loop in osteoclast differentiation and represents a novel mechanism to restrain pathological bone destruction.
    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
    分析不表现出显著认知缺陷的2型糖尿病(T2DM)个体的特定脑区域内的基因表达变化可以产生对朝向更严重表型进展的潜在机制的有价值的见解。在这项研究中,长期T2DM小鼠的皮质和海马的转录组学分析显示,大脑皮质中28个基因和海马中15个基因的表达发生了变化。在这些基因中,六个在皮质和海马中显示出一致的变化:干扰素调节因子7(Irf7),缺氧诱导因子3α(Hif-3α),周期昼夜节律时钟2(Per2),黄嘌呤脱氢酶(Xdh),和转化生长因子β刺激的克隆22/TSC22(Tsc22d3)上调,而Claudin-5(Cldn5)下调。通过RT-qPCR实现这些变化的确认。在蛋白质水平,CLDN5和IRF7表现出相似的改变,CLDN5下调,IRF7上调。此外,2型糖尿病小鼠海马和皮质组织IκBα水平降低,这也暗示了NF-κB通路的参与。一起来看,这些结果表明,血脑屏障的减弱和通过干扰素1和NF-κB通路的异常炎症反应是长期T2DM患者认知损害的基础.
    Analyzing changes in gene expression within specific brain regions of individuals with Type 2 Diabetes (T2DM) who do not exhibit significant cognitive deficits can yield valuable insights into the mechanisms underlying the progression towards a more severe phenotype. In this study, transcriptomic analysis of the cortex and hippocampus of mice with long-term T2DM revealed alterations in the expression of 28 genes in the cerebral cortex and 15 genes in the hippocampus. Among these genes, six displayed consistent changes in both the cortex and hippocampus: Interferon regulatory factor 7 (Irf7), Hypoxia-inducible factor 3 alpha (Hif-3α), period circadian clock 2 (Per2), xanthine dehydrogenase (Xdh), and Transforming growth factor β-stimulated clone 22/TSC22 (Tsc22d3) were upregulated, while Claudin-5 (Cldn5) was downregulated. Confirmation of these changes was achieved through RT-qPCR. At the protein level, CLDN5 and IRF7 exhibited similar alterations, with CLDN5 being downregulated and IRF7 being upregulated. In addition, the hippocampus and cortex of the T2DM mice showed decreased levels of IκBα, implying the involvement of NF-κB pathways as well. Taken together, these results suggest that the weakening of the blood-brain barrier and an abnormal inflammatory response via the Interferon 1 and NF-κB pathways underlie cognitive impairment in individuals with long-standing T2DM.
    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
    目的:本研究的主要重点是探讨IRF7调控胰腺腺癌(PAAD)M1型巨噬细胞RPS18转录的分子机制,以及IRF7通过外泌体将RPS18转移到PAAD细胞和调节ILF3表达。
    方法:通过利用来自基因表达综合数据库的单细胞RNA测序(scRNA-seq)数据和空间转录组学(ST)数据,我们鉴定了PAAD组织中具有显著表达差异的不同细胞类型.在这些细胞类型中,我们确定了与脂质代谢密切相关的那些。分析了这些细胞类型中差异表达的基因,并确定了与预后相关的靶基因。流式细胞术用于评估M1和M2巨噬细胞中靶基因的表达水平。利用CRISPR/Cas9编辑技术构建靶基因敲除细胞系,使用慢病毒载体建立具有靶基因敲低和过表达的细胞系。此外,建立了M1巨噬细胞与PAAD细胞来源的外泌体共培养模型。通过代谢组学分析评估了M1巨噬细胞来源的外泌体对模型中PAAD细胞脂质代谢的影响。M1巨噬细胞来源的外泌体对生存力的影响,扩散,司,使用MTT法评估PAAD细胞的迁移和凋亡,流式细胞术,EdU分析,伤口愈合试验,Transwell测定和TUNEL染色。此外,建立小鼠PAAD原位植入模型,和生物发光成像被用来评估M1巨噬细胞来源的外泌体对PAAD细胞的肿瘤内形成能力的影响,以及测量肿瘤的重量和体积。使用免疫组织化学检查肿瘤组织中增殖相关蛋白的表达。
    结果:通过对scRNA-seq和ST技术的联合分析,我们发现PAAD样本中的M1巨噬细胞与脂质代谢信号密切相关,以及M1巨噬细胞和癌细胞之间的负相关。预后风险评分模型的构建将RPS18和IRF7确定为M1巨噬细胞中的两个预后相关基因,呈现负相关和正相关,分别。机械上,研究发现,M1巨噬细胞中的IRF7可抑制RPS18的转录,减少RPS18通过外泌体向PAAD细胞的转移,从而影响PAAD细胞中ILF3的表达。M1巨噬细胞中的IRF7/RPS18还可以抑制脂质代谢,细胞活力,扩散,迁移,PAAD细胞的侵袭和肿瘤内形成能力,同时促进细胞凋亡。
    结论:在M1巨噬细胞中过表达IRF7可能抑制RPS18转录,减少RPS18从M1巨噬细胞来源的外泌体转移到PAAD细胞,从而抑制PAAD细胞中ILF3的表达,抑制脂质代谢途径,限制生存能力,扩散,迁移,PAAD细胞的侵袭,以及增强细胞凋亡,最终抑制体内PAAD细胞中的肿瘤形成。在M1巨噬细胞中靶向IRF7/RPS18可能代表未来PAAD的有希望的免疫治疗方法。
    OBJECTIVE: The main focus of this study is to explore the molecular mechanism of IRF7 regulation on RPS18 transcription in M1-type macrophages in pancreatic adenocarcinoma (PAAD) tissue, as well as the transfer of RPS18 by IRF7 via exosomes to PAAD cells and the regulation of ILF3 expression.
    METHODS: By utilising single-cell RNA sequencing (scRNA-seq) data and spatial transcriptomics (ST) data from the Gene Expression Omnibus database, we identified distinct cell types with significant expression differences in PAAD tissue. Among these cell types, we identified those closely associated with lipid metabolism. The differentially expressed genes within these cell types were analysed, and target genes relevant to prognosis were identified. Flow cytometry was employed to assess the expression levels of target genes in M1 and M2 macrophages. Cell lines with target gene knockout were constructed using CRISPR/Cas9 editing technology, and cell lines with target gene knockdown and overexpression were established using lentiviral vectors. Additionally, a co-culture model of exosomes derived from M1 macrophages with PAAD cells was developed. The impact of M1 macrophage-derived exosomes on the lipid metabolism of PAAD cells in the model was evaluated through metabolomics analysis. The effects of M1 macrophage-derived exosomes on the viability, proliferation, division, migration and apoptosis of PAAD cells were assessed using MTT assay, flow cytometry, EdU assay, wound healing assay, Transwell assay and TUNEL staining. Furthermore, a mouse PAAD orthotopic implantation model was established, and bioluminescence imaging was utilised to assess the influence of M1 macrophage-derived exosomes on the intratumoural formation capacity of PAAD cells, as well as measuring tumour weight and volume. The expression of proliferation-associated proteins in tumour tissues was examined using immunohistochemistry.
    RESULTS: Through combined analysis of scRNA-seq and ST technologies, we discovered a close association between M1 macrophages in PAAD samples and lipid metabolism signals, as well as a negative correlation between M1 macrophages and cancer cells. The construction of a prognostic risk score model identified RPS18 and IRF7 as two prognostically relevant genes in M1 macrophages, exhibiting negative and positive correlations, respectively. Mechanistically, it was found that IRF7 in M1 macrophages can inhibit the transcription of RPS18, reducing the transfer of RPS18 to PAAD cells via exosomes, consequently affecting the expression of ILF3 in PAAD cells. IRF7/RPS18 in M1 macrophages can also suppress lipid metabolism, cell viability, proliferation, migration, invasion and intratumoural formation capacity of PAAD cells, while promoting cell apoptosis.
    CONCLUSIONS: Overexpression of IRF7 in M1 macrophages may inhibit RPS18 transcription, reduce the transfer of RPS18 from M1 macrophage-derived exosomes to PAAD cells, thereby suppressing ILF3 expression in PAAD cells, inhibiting the lipid metabolism pathway, and curtailing the viability, proliferation, migration, invasion of PAAD cells, as well as enhancing cell apoptosis, ultimately inhibiting tumour formation in PAAD cells in vivo. Targeting IRF7/RPS18 in M1 macrophages could represent a promising immunotherapeutic approach for PAAD in the future.
    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:视网膜色素上皮(RPE)细胞在保持视网膜平衡和调节脉络膜与视网膜之间的免疫相互作用方面具有关键作用。这项研究主要集中在描述山奈酚(Kae)对RPE细胞损伤的保护作用。
    方法:对GSE30719数据集进行生物信息学分析,以鉴定与RPE相关的hub基因。随后,我们分析了Kae对RPE细胞凋亡的影响,细胞活力,通过细胞实验和炎症反应,并探索了hub基因与Kae之间的相互作用。
    结果:基于GSE30719数据集,九个hub基因(ISG15、IFIT1、IFIT3、STAT1、OASL、RSAD2、IRF7、MX2和MX1)被鉴定,所有这些在GSE30719病例组中高表达。Kae可以促进脂多糖(LPS)引起的RPE细胞的增殖活性,以及减少细胞凋亡和炎症因子(肿瘤坏死因子受体(TNFR)的产生,白细胞介素-1β(IL-1β)和细胞因子(IL-1、IL-6、IL-12)。STAT1被证明可以抑制细胞增殖,促进细胞凋亡,并在LPS诱导的RPE细胞中分泌IL-1/IL-6/IL-12。此外,在LPS诱导的RPE细胞中发现IRF7与STAT1相互作用,STAT1可以通过去泛素化来维持IRF7水平。此外,我们还发现Kae对LPS诱导的RPE细胞损伤的保护作用是通过STAT1/IRF7轴介导的。
    结论:本研究提供了Kae通过调节STAT1/IRF7信号通路保护RPE细胞的证据,表明其在与RPE细胞损伤相关的视网膜疾病的诊断和管理中的潜在治疗相关性。
    BACKGROUND: Retinal pigment epithelial (RPE) cells have a pivotal function in preserving the equilibrium of the retina and moderating the immunological interaction between the choroid and the retina. This study primarily focuses on delineating the protective effect offered by Kaempferol (Kae) against RPE cell damage.
    METHODS: Bioinformatics analysis was performed on the GSE30719 dataset to identify hub genes associated with RPE. Subsequently, we analyzed the impact of Kae on RPE apoptosis, cell viability, and inflammatory response through cell experiments, and explored the interaction between hub genes and Kae.
    RESULTS: Based on the GSE30719 dataset, nine hub genes (ISG15, IFIT1, IFIT3, STAT1, OASL, RSAD2, IRF7, MX2, and MX1) were identified, all of which were highly expressed in the GSE30719 case group. Kae could boost the proliferative activity of RPE cells caused by lipopolysaccharide (LPS), as well as reduce apoptosis and the generation of inflammatory factors (tumor necrosis factor receptor (TNFR), interleukin-1beta (IL-1β)) and cytokines (IL-1, IL-6, IL-12). STAT1 was shown to inhibit cell proliferation, promote apoptosis, and secrete IL-1/IL-6/IL-12 in LPS-induced RPE cells. Moreover, IRF7 was found to interact with STAT1 in LPS-induced RPE cells, and STAT1 could maintain IRF7 levels through deubiquitination. In addition, we also found that the protective effect of Kae on LPS-induced RPE cell injury was mediated through STAT1/IRF7 axis.
    CONCLUSIONS: This study provided evidence that Kae protects RPE cells via regulating the STAT1/IRF7 signaling pathways, indicating its potential therapeutic relevance in the diagnosis and management of retinal disorders linked with RPE cell damage.
    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
    短嘴和侏儒症综合征(SBDS)的爆发,由新型鹅细小病毒(NGPV)引起,自2015年以来在中国发生。NGPV,单链DNA病毒,被认为是垂直传播的。然而,NGPV免疫逃避的机制尚不清楚。在这项研究中,我们研究了NGPV感染对鸭胚胎成纤维细胞(DEF)细胞环GMP-AMP合酶(cGAS)-干扰素基因刺激因子(STING)信号通路的影响。我们的发现表明,NGPV感染刺激cGAS的mRNA表达,但导致IFN-β诱导较弱。NGPV阻碍IFN-β和下游干扰素刺激基因的表达,从而减少由干扰素刺激DNA(ISD)和聚(I:C)诱导的IFN-β的分泌。RNA-seq结果显示,NGPV感染下调干扰素mRNA表达,同时增强炎症因子的mRNA表达。此外,病毒蛋白过表达的结果表明,与其它病毒蛋白相比,VP1表现出显著的抑制IFN-β表达的能力。结果表明,只有完整的VP1蛋白能抑制IFN-β的表达,而截短的蛋白VP1U和VP2不具有这样的特征。免疫沉淀实验表明,VP1和VP2均可与IRF7蛋白相互作用,而VP1U没有。总之,我们的发现表明,NGPV感染通过IRF7分子潜在地调节干扰素和干扰素刺激因子的表达和分泌,从而损害宿主的先天免疫应答。受VP1蛋白调控。
    Outbreaks of short beak and dwarfism syndrome (SBDS), caused by a novel goose parvovirus (NGPV), have occurred in China since 2015. The NGPV, a single-stranded DNA virus, is thought to be vertically transmitted. However, the mechanism of NGPV immune evasion remains unclear. In this study, we investigated the impact of NGPV infection on the Cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway in duck embryonic fibroblast (DEF) cells. Our findings demonstrate that NGPV infection stimulates the mRNA expression of cGAS but results in weak IFN-β induction. NGPV impedes the expression of IFN-β and downstream interferon-stimulated genes, thereby reducing the secretion of IFN-β induced by interferon-stimulating DNA (ISD) and poly (I: C). RNA-seq results show that NGPV infection downregulates interferon mRNA expression while enhancing the mRNA expression of inflammatory factors. Additionally, the results of viral protein over-expression indicate that VP1 exhibits a remarkable ability to inhibit IFN-β expression compared to other viral proteins. Results indicated that only the intact VP1 protein could inhibit the expression of IFN-β, while the truncated proteins VP1U and VP2 do not possess such characteristics. The immunoprecipitation experiment showed that both VP1 and VP2 could interact with IRF7 protein, while VP1U does not. In summary, our findings indicate that NGPV infection impairs the host\'s innate immune response by potentially modulating the expression and secretion of interferons and interferon-stimulating factors via IRF7 molecules, which are regulated by the VP1 protein.
    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
    TLR是最彻底研究的一组模式识别受体,在先天免疫中起着核心作用。其中,TLR10(CD290)仍然是唯一的TLR家族成员,没有已知的配体和明确定义的功能。研究TLR10的一个主要障碍是其在小鼠中的缺失。最近的一项关于TLR10敲入小鼠的研究证明了其对B细胞的内在抑制作用,表明TLR10是自身免疫性疾病的潜在药物靶点。在这项研究中,我们研究了TLR10在人浆细胞样树突状细胞(pDCs)中的表达和功能。我们已经看到原发性人类pDCs,B细胞,和单核细胞组成型表达TLR10。与抗TLR10Ab预温育后,pDC中细胞因子的产生在DNA和RNA病毒的刺激下被下调。在进一步调查可能的机制后,我们记录了Ab介导的TLR10接合后STAT3的磷酸化。这导致细胞因子信号传导3(SOCS3)表达的抑制性分子抑制剂的诱导。我们还记录了TLR10接合后pDC中转录因子IFN调节因子7(IRF7)的核易位的抑制。我们的数据提供了(据我们所知)TLR10在人pDC的表面上组成性表达并作为其先天反应的调节剂的第一个证据。我们的发现表明通过Ab介导的TLR10靶向利用pDCs功能的潜力,这可能为自身免疫性疾病开辟新的治疗途径。
    TLRs are the most thoroughly studied group of pattern-recognition receptors that play a central role in innate immunity. Among them, TLR10 (CD290) remains the only TLR family member without a known ligand and clearly defined functions. One major impediment to studying TLR10 is its absence in mice. A recent study on TLR10 knock-in mice demonstrated its intrinsic inhibitory role in B cells, indicating that TLR10 is a potential drug target in autoimmune diseases. In this study, we interrogated the expression and function of TLR10 in human plasmacytoid dendritic cells (pDCs). We have seen that primary human pDCs, B cells, and monocytes constitutively express TLR10. Upon preincubation with an anti-TLR10 Ab, production of cytokines in pDCs was downregulated in response to stimulation with DNA and RNA viruses. Upon further investigation into the possible mechanism, we documented phosphorylation of STAT3 upon Ab-mediated engagement of TLR10. This leads to the induction of inhibitory molecule suppressor of cytokine signaling 3 (SOCS3) expression. We have also documented the inhibition of nuclear translocation of transcription factor IFN regulatory factor 7 (IRF7) in pDCs following TLR10 engagement. Our data provide the (to our knowledge) first evidence that TLR10 is constitutively expressed on the surface of human pDCs and works as a regulator of their innate response. Our findings indicate the potential of harnessing the function of pDCs by Ab-mediated targeting of TLR10 that may open a new therapeutic avenue for autoimmune disorders.
    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
    干燥综合征(SS)是一种自身免疫性疾病,其中唾液腺(SGs)和泪腺(LGs)受到淋巴细胞浸润和炎症的影响。据报道,浆细胞样树突状细胞(pDCs)释放的干扰素-α(IFN-α)有助于SS的病理,和ART已被证明有效改善SS。尽管目前的研究努力,ART在SS治疗中的作用机制仍有待完全阐明。ART是否可以通过抑制IFN-α来治疗SS尚不清楚。在研究期间在体内和体外设置中测试了该假设。SS模型小鼠,用ART治疗,显示与干燥相关的症状改善。RNA-seq分析揭示了ART治疗后强烈的抗IFN-α信号应答。另外的体外研究进一步证实了ART的应用抑制了MyD88蛋白的表达和IRF7的核易位。这表明ART对TLR-MyD88-IRF7途径的干预在SS的治疗方法中起作用。总之,这项研究强调了ART在SS中的治疗潜力,并且ART通过TLR-MyD88-IRF7途径抑制pDC中的IFN-α信号传导。
    Sjögren\'s syndrome (SS) is an autoimmune disease in which the salivary glands (SGs) and the lacrimal glands (LGs) are affected by lymphocytic infiltration and inflammation. It has been reported that interferon-α (IFN-α) released by plasmacytoid dendritic cells (pDCs) contribute to the pathology of SS, and ART has been shown to effectively ameliorates SS. Despite the current research endeavors, the mechanism of how ART works in the treatment of SS remains to be fully elucidated. Whether ART can treat SS by inhibiting IFN-α remains unclear. This hypothesis was tested both in vivo and in vitro settings during the study. The SS model mice, which were treated with ART, showed amelioration in symptoms related to dryness. RNA-seq analysis revealed strong anti-IFN-α signaling response upon ART treatment. Additional in vitro studies provided further confirmation that the application of ART inhibits the MyD88 protein expression and the nuclear translocation of IRF7. This suggests that the intervention of ART in the TLR-MyD88-IRF7 pathway plays a role in the therapeutic approach for SS. In summary, this study highlighted the therapeutic potential of ART in SS and ART inhibited the IFN-α signaling in pDCs via the TLR-MyD88-IRF7 pathway.
    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
    I型干扰素(IFN-I)是具有多种活性的多效性因子,在先天和适应性免疫中起重要作用。虽然许多研究表明IFN-I诱导剂对广谱抗病毒发挥有利作用,免疫调节,通过诱导内源性IFN-I和IFN刺激基因(ISGs)进行抗肿瘤,它们在骨稳态中的作用仍需进一步探索。这里,我们的研究证明了两种不同的IFN-I诱导剂,diABZI和聚(I:C),作为缓解骨质溶解和骨质疏松症的潜在疗法。首先,IFN-I诱导剂在体外抑制控制破骨细胞(OC)分化和活性的基因。此外,diABZI通过抑制OC分化和功能在体内减轻Ti颗粒诱导的骨溶解和卵巢切除(OVX)诱导的骨质疏松症中的骨丢失。此外,在来自Ifnar1-/-小鼠的巨噬细胞中未观察到IFN-I诱导剂对OC分化的抑制作用,这表明IFN-I诱导剂对OC的抑制作用是IFNAR依赖性的。机械上,OC前体中IRF7和IFIT3的RNAi介导的沉默削弱了IFN-I诱导剂对OC分化的抑制作用。一起来看,这些结果表明,IFN-I诱导剂通过IFN-β信号通路诱导OC特异性介质,通过限制破骨细胞生成和骨吸收,在骨转换中起保护作用。
    Type I interferons (IFN-I) are pleiotropic factors endowed with multiple activities that play important roles in innate and adaptive immunity. Although many studies indicate that IFN-I inducers exert favorable effects on broad-spectrum antivirus, immunomodulation, and anti-tumor activities by inducing endogenous IFN-I and IFN-stimulated genes, their function in bone homeostasis still needs further exploration. Here, our study demonstrates 2 distinct IFN-I inducers, diABZI and poly(I:C), as potential therapeutics to alleviate osteolysis and osteoporosis. First, IFN-I inducers suppress the genes that control osteoclast (OC) differentiation and activity in vitro. Moreover, diABZI alleviates bone loss in Ti particle-induced osteolysis and ovariectomized -induced osteoporosis in vivo by inhibiting OC differentiation and function. In addition, the inhibitory effects of IFN-I inducers on OC differentiation are not observed in macrophages derived from Ifnar1-/-mice, which indicate that the suppressive effect of IFN-I inducers on OC is IFNAR-dependent. Mechanistically, RNAi-mediated silencing of IRF7 and IFIT3 in OC precursors impairs the suppressive effect of the IFN-I inducers on OC differentiation. Taken together, these results demonstrate that IFN-I inducers play a protective role in bone turnover by limiting osteoclastogenesis and bone resorption through the induction of OC-specific mediators via the IFN-I signaling pathway.
    OCs are responsible for bone resorption, and their excessive differentiation and enhanced activity will lead to bone resorption diseases such as osteoporosis and osteolysis. Here, our study demonstrates 2 distinct IFN-I inducers, diABZI and poly(I:C), as potential therapeutics to alleviate osteolysis and osteoporosis. IFN-I inducers suppress OC differentiation, and particularly diABZI alleviates bone loss in osteolysis and osteoporosis mouse models. Taken together, IFN-I inducers play a protective role in bone turnover by limiting osteoclastogenesis and bone resorption through the induction of OC-specific mediators via the IFN-I signaling pathway. Our in-depth and comprehensive discovery of the IFN-I inducer would provide new insight into OC biology and therapeutic targets for osteoclastic bone resorption diseases.
    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
    I型干扰素表现出抗增殖和抗癌活性,但它们在癌症中的详细调控机制尚未完全阐明。RNA结合蛋白是基因调控的主要协调者,与肿瘤进展密切相关。在这里,我们显示上调的RNA结合蛋白RBM45与乳腺癌的不良预后相关。在培养细胞和异种移植小鼠模型中,RBM45的耗尽均抑制乳腺癌进展。机械上,RBM45消融通过调节I型干扰素信号抑制乳腺癌进展,特别是通过提高IFN-β的生产。重要的是,RBM45将TRIM28招募到IRF7,并刺激其磺酰化,从而抑制IFNB1转录。RBM45的缺失通过减少TRIM28和IRF7之间的相互作用来促进IFNB1转录,从而降低IRF7的SUMO化,导致抑制乳腺癌进展。一起来看,我们的发现揭示了RBM45在调节I型干扰素信号和癌症侵袭性进展中的重要作用,提示RBM45是乳腺癌的潜在治疗靶点。
    Type I interferons exhibit anti-proliferative and anti-cancer activities, but their detailed regulatory mechanisms in cancer have not been fully elucidated yet. RNA binding proteins are master orchestrators of gene regulation, which are closely related to tumor progression. Here we show that the upregulated RNA binding protein RBM45 correlates with poor prognosis in breast cancer. Depletion of RBM45 suppresses breast cancer progression both in cultured cells and xenograft mouse models. Mechanistically, RBM45 ablation inhibits breast cancer progression through regulating type I interferon signaling, particularly by elevating IFN-β production. Importantly, RBM45 recruits TRIM28 to IRF7 and stimulates its SUMOylation, thereby repressing IFNB1 transcription. Loss of RBM45 reduced the SUMOylation of IRF7 by reducing the interaction between TRIM28 and IRF7 to promote IFNB1 transcription, leading to the inhibition of breast cancer progression. Taken together, our finding uncovers a vital role of RBM45 in modulating type I interferon signaling and cancer aggressive progression, implicating RBM45 as a potential therapeutic target in breast cancer.
    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
    干扰素(IFN)是主要参与启动针对病原体的先天免疫应答和促进免疫细胞成熟的信号蛋白。干扰素调节因子7(IRF7)在IFN信号通路中起关键作用。IRF7的激活过程是由外源或异常核酸激发的,然后通过模式识别受体(PRR)和随后的信号级联进行鉴定。激活后,IRF7调节IFN的表达和炎症基因调节。作为一种多功能转录因子,IRF7主要表达于免疫细胞,然而它的存在也在角质形成细胞中检测到,成纤维细胞,和各种真皮细胞类型。在这些细胞中,IRF7对皮肤免疫至关重要,炎症,和纤维化。IRF7失调可能导致自身免疫和炎性皮肤病,包括系统性硬皮病(SSc),系统性红斑狼疮(SLE),特应性皮炎(AD)和牛皮癣。本综述旨在广泛阐明IRF7及其信号通路在免疫细胞和角质形成细胞中的作用。强调其在皮肤相关和结缔组织疾病中的重要性。
    Interferons (IFNs) are signalling proteins primarily involved in initiating innate immune responses against pathogens and promoting the maturation of immune cells. Interferon Regulatory Factor 7 (IRF7) plays a pivotal role in the IFNs signalling pathway. The activation process of IRF7 is incited by exogenous or abnormal nucleic acids, which is followed by the identification via pattern recognition receptors (PRRs) and the ensuing signalling cascades. Upon activation, IRF7 modulates the expression of both IFNs and inflammatory gene regulation. As a multifunctional transcription factor, IRF7 is mainly expressed in immune cells, yet its presence is also detected in keratinocytes, fibroblasts, and various dermal cell types. In these cells, IRF7 is critical for skin immunity, inflammation, and fibrosis. IRF7 dysregulation may lead to autoimmune and inflammatory skin conditions, including systemic scleroderma (SSc), systemic lupus erythematosus (SLE), Atopic dermatitis (AD) and Psoriasis. This comprehensive review aims to extensively elucidate the role of IRF7 and its signalling pathways in immune cells and keratinocytes, highlighting its significance in skin-related and connective tissue 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