Smoothened Receptor

光滑受体
  • 文章类型: Journal Article
    mTORC1复合物受TSC1和TSC2负调控。Hedgehog信号的激活严格依赖于Smoothened和Hedgehog信号效应子和转录因子之间的通讯,GLI2,在初级纤毛中。有关此通信的详细信息尚不清楚,我们想进一步探索这一点。在这里,我们报道了在Tsc2-/-MEFs中组成激活的mTORC1导致平滑到质膜的错误定位,结合纤毛中GLI2浓度的增加和Hedgehog信号的减少,通过Hedgehog靶基因的表达降低来测量,Gli1对mTORC1的抑制挽救了光滑纤毛的细胞定位,减少纤毛的GLI2浓度,并恢复Hedgehog信号。我们的结果揭示了GLI2两步激活过程的证据。第一步包括GLI2稳定和纤毛定位,而第二步包括与纤毛局部平滑的通信。我们发现mTORC1抑制第二步。这是mTORC1参与调节Hedgehog信号的第一个证明。
    The mTORC1-complex is negatively regulated by TSC1 and TSC2. Activation of Hedgehog signaling is strictly dependent on communication between Smoothened and the Hedgehog-signaling effector and transcription factor, GLI2, in the primary cilium. Details about this communication are not known, and we wanted to explore this further. Here we report that in Tsc2 -/- MEFs constitutively activated mTORC1 led to mis-localization of Smoothened to the plasma membrane, combined with increased concentration of GLI2 in the cilia and reduced Hedgehog signaling, measured by reduced expression of the Hedgehog target gene, Gli1 Inhibition of mTORC1 rescued the cellular localization of Smoothened to the cilia, reduced the cilia concentration of GLI2, and restored Hedgehog signaling. Our results reveal evidence for a two-step activation process of GLI2. The first step includes GLI2 stabilization and cilium localization, whereas the second step includes communication with cilia-localized Smoothened. We found that mTORC1 inhibits the second step. This is the first demonstration that mTORC1 is involved in the regulation of Hedgehog signaling.
    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
    目标:平滑(SMO),刺猬信号通路的关键组成部分,代表三阴性乳腺癌(TNBC)的治疗靶标,然而,TNBC患者的化疗反应率仅为40-50%,强调迫切需要开发新药物来有效治疗这种疾病。新化合物TPB15,一种衍生自[1,2,4]三唑并[4,3-α]吡啶的SMO抑制剂,与第一种SMO抑制剂vismodegib相比,在体外和体内均表现出优异的抗TNBC活性和较低的毒性。然而,该化合物的药代动力学特性尚不清楚。本工作旨在开发一种简单的HPLC-MS/MS方法,以描述TPB15在大鼠中的药代动力学和生物利用度,作为进一步临床研究的基础工作。
    方法:在AgilentZORBAXStableBondC18柱上通过梯度洗脱,使用乙腈和0.1%甲酸作为流动相以0.3mL/min的流速进行分离。采用m/z454.2→100.0,248.1→121.1的正模式多反应监测(MRM)来确定TPB15和内标替硝唑,分别。特异性,日内和日间精度和准确性,提取回收,稳定性,基体效应,方法的稀释完整性和残留性得到验证。通过以5mg/kg的剂量静脉注射和25mg/kg的剂量口服灌胃对大鼠进行TPB15的药代动力学和生物利用度研究。使用药代动力学软件DAS2.1.1通过非隔室分析计算药代动力学参数。
    结果:特异性值,日内和日间精度和准确性,提取回收,稳定性,基体效应,稀释完整性和残留满足可接受的限制。该方法的定量下限为10ng/mL,线性范围为10-2000ng/mL。然后通过灌胃(25mg/kg)和静脉注射(5mg/kg)将经过验证的方法应用于大鼠的药代动力学和生物利用度研究,TBP15在大鼠体内的口服生物利用度为16.4±3.5%。药代动力学参数计算如下:最大血浆浓度(Cmax)(PO:2787.17±279.45µg/L),达到最大血浆浓度的时间(Tmax)(PO:4.20±0.90h),浓度-时间曲线下面积0至时间(AUC0-t)(PO:17,373.03±2585.18ng/mL·h,IV:21,129.79±3360.84ng/mL·h),浓度-时间曲线下面积为0至无穷大(AUC0-∞)(PO:17,443.85±2597.63ng/mL·h,IV:17,443.85±2597.63ng/mL·h),终末消除半衰期(t1/2)(PO:7.26±2.16h,IV:4.78±1.09h)。
    结论:TPB15是治疗TNBC的有希望的候选药物,已在体外和体内表现出突出的疗效和安全性。这项研究建立了一个简单的,敏感,和快速HPLC-MS/MS生物分析方法,根据FDA和EMA指南开发和验证,用于进行TPB15的药代动力学和生物利用度研究。结果表明,由于其长t1/2,药代动力学特征良好。然而,下一阶段的研究应包括配方筛选,以提高生物利用度,以及临床试验,代谢途径分析,和评估潜在的药物-药物相互作用。
    OBJECTIVE: Smoothened (SMO), a key component of the hedgehog signaling pathway, represents a therapeutic target for triple negative breast cancer (TNBC), yet the chemotherapy response rate in TNBC patients is only 40-50%, underscoring the urgent need for the development of novel drugs to effectively treat this condition. The novel compound TPB15, an SMO inhibitor derived from [1,2,4] triazolo [4,3-α] pyridines, demonstrated superior anti-TNBC activity and lower toxicity compared to the first SMO inhibitor vismodegib in both in vitro and in vivo. However, the compound\'s pharmacokinetic properties remain unclear. The present work aims to develop a simple HPLC-MS/MS method to profile the pharmacokinetics and bioavailability of TPB15 in rats as a ground work for further clinical research.
    METHODS: Separation was performed on an Agilent ZORBAX StableBond C18 column by gradient elution using acetonitrile and 0.1% formic acid as mobile phase at a flow rate of 0.3 mL/min. Multiple reaction monitoring(MRM) in positive mode with the transitions of m/z 454.2 → 100.0, 248.1 → 121.1 was employed to determine TPB15 and internal standard tinidazole, respectively. The specificity, intra- and inter- day precision and accuracy, extraction recovery, stability, matrix effect, dilution integrity and carryover of the method was validated. The pharmacokinetics and bioavailability  study of TPB15 were carried out on rats through intravenous injection at the dose of 5 mg/kg and oral gavage at the dose of 25 mg/kg, and the pharmacokinetics parameters were calculated by the non-compartment analysis using the pharmacokinetics software DAS 2.1.1.
    RESULTS: The values of specificity, intra- and inter- day precision and accuracy, extraction recovery, stability, matrix effect, dilution integrity and carryover satisfied the acceptable limits. The lower limit of quantification of this method was 10 ng/mL with a linear range of 10-2000 ng/mL. The validated method was then applied to pharmacokinetics and bioavailability studies in rat by dosing with gavage (25 mg/kg) and intravenous injection(5 mg/kg), and the oral bioavailability of TBP15 in rat was calculated as 16.4 ± 3.5%. The pharmacokinetic parameters were calculated as following: maximum of plasma concentration (Cmax) (PO: 2787.17 ± 279.45 µg/L), Time to maximum plasma concentration (Tmax) (PO: 4.20 ± 0.90 h), the area under the concentration-time curve 0 to time (AUC0-t) (PO: 17,373.03 ± 2585.18 ng/mL·h, IV: 21,129.79 ± 3360.84 ng/mL·h), the area under the concentration-time curve 0 to infinity (AUC0-∞) (PO: 17,443.85 ± 2597.63 ng/mL·h, IV: 17,443.85 ± 2597.63 ng/mL·h), terminal elimination half-life (t1/2) (PO: 7.26 ± 2.16 h, IV: 4.78 ± 1.09 h).
    CONCLUSIONS: TPB15, a promising candidate for treating TNBC, has demonstrated outstanding efficacy and safety in vitro and in vivo. This study established a simple, sensitive, and rapid HPLC-MS/MS bioanalytical method, developed and validated in accordance with FDA and EMA guidelines, for conducting pharmacokinetic and bioavailability studies of TPB15. The results revealed a favorable pharmacokinetic profile owing to its long t1/2. Nevertheless, the next phase of research should include formulation screening to enhance bioavailability, as well as clinical trials, metabolism pathway analysis, and assessment of potential drug-drug interactions.
    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
    在Hedgehog(Hh)信号转导过程中的发育和疾病,非典型G蛋白偶联受体(GPCR)SMOOTHENED(SMO)通过结合蛋白激酶A催化亚基(PKA-C)并物理阻断其酶活性与GLI转录因子通信。这里,我们显示GPCR激酶2(GRK2)在原代纤毛内源性小鼠和斑马鱼Hh通路激活过程中协调这一过程。SMO激活后,GRK2从纤毛基部快速重新定位到轴,触发SMO磷酸化和PKA-C相互作用。重构研究表明,GRK2磷酸化使活性SMO能够直接结合PKA-C。最后,SMO-GRK2-PKA途径是一系列细胞和体内模型中Hh信号转导的基础。因此,纤毛SMO的GRK2磷酸化以及随后的PKA-C结合和失活是Hh信号传导中细胞内步骤的关键启动事件。更广泛地说,我们的研究表明,GRKs在实现与不同细胞内效应物的直接GPCR相互作用方面具有扩大的作用。
    During Hedgehog (Hh) signal transduction in development and disease, the atypical G protein-coupled receptor (GPCR) SMOOTHENED (SMO) communicates with GLI transcription factors by binding the protein kinase A catalytic subunit (PKA-C) and physically blocking its enzymatic activity. Here, we show that GPCR kinase 2 (GRK2) orchestrates this process during endogenous mouse and zebrafish Hh pathway activation in the primary cilium. Upon SMO activation, GRK2 rapidly relocalizes from the ciliary base to the shaft, triggering SMO phosphorylation and PKA-C interaction. Reconstitution studies reveal that GRK2 phosphorylation enables active SMO to bind PKA-C directly. Lastly, the SMO-GRK2-PKA pathway underlies Hh signal transduction in a range of cellular and in vivo models. Thus, GRK2 phosphorylation of ciliary SMO and the ensuing PKA-C binding and inactivation are critical initiating events for the intracellular steps in Hh signaling. More broadly, our study suggests an expanded role for GRKs in enabling direct GPCR interactions with diverse intracellular effectors.
    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
    髓母细胞瘤(SHH-MB)的Sonichedgehog亚群的特征在于SHH信号通路的异常激活。阳性SHH调节剂Smoothened(SMO)的抑制已证明有希望的临床疗效。然而,对SMO抑制剂的原发性和获得性耐药性限制了其疗效。有必要了解对治疗耐药的潜在分子机制,以弥合这种未满足的需求。这里,我们在鼠SMB21和人类DAOY细胞中利用全基因组CRISPR-Cas9敲除筛选,为了解开遗传依赖性和药物相关的遗传相互作用物,它们可以作为SHH-MB的替代治疗靶标。我们的屏幕加强SMB21细胞作为SHH-MB的忠实模型系统,与DAOY细胞相反,并确定表观遗传机制的成员,包括DNA甲基转移酶1(DNMT1)作为SHH依赖性肿瘤的药物靶标。我们表明,Dnmt1在正常小鼠小脑发育中起着至关重要的作用,并且是体内SHH-MB生长所必需的。此外,单独的DNMT1药理学抑制以及与SMO抑制组合有效地抑制鼠和人SHH-MB细胞模型中的肿瘤生长,并通过抑制SMO下游的SHH信号传导输出来延长SHH-MB小鼠模型的存活。总之,我们的数据强调了抑制表观遗传调节因子作为SMO抑制剂敏感和耐药SHH-MB的新治疗途径的潜力.
    Sonic hedgehog subgroup of medulloblastoma (SHH-MB) is characterized by aberrant activation of the SHH signaling pathway. An inhibition of the positive SHH regulator Smoothened (SMO) has demonstrated promising clinical efficacy. Yet, primary and acquired resistance to SMO inhibitors limit their efficacy. An understanding of underlying molecular mechanisms of resistance to therapy is warranted to bridge this unmet need. Here, we make use of genome-wide CRISPR-Cas9 knockout screens in murine SMB21 and human DAOY cells, in order to unravel genetic dependencies and drug-related genetic interactors that could serve as alternative therapeutic targets for SHH-MB. Our screens reinforce SMB21 cells as a faithful model system for SHH-MB, as opposed to DAOY cells, and identify members of the epigenetic machinery including DNA methyltransferase 1 (DNMT1) as druggable targets in SHH-dependent tumors. We show that Dnmt1 plays a crucial role in normal murine cerebellar development and is required for SHH-MB growth in vivo. Additionally, DNMT1 pharmacological inhibition alone and in combination with SMO inhibition effectively inhibits tumor growth in murine and human SHH-MB cell models and prolongs survival of SHH-MB mouse models by inhibiting SHH signaling output downstream of SMO. In conclusion, our data highlight the potential of inhibiting epigenetic regulators as a novel therapeutic avenue in SMO-inhibitor sensitive as well as resistant SHH-MBs.
    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
    Hedgehog(Hh)信号通路的异常激活与胰腺癌的发生和发展有关。出于这个原因,通过靶向G蛋白偶联受体Smoothened(SMO)的抑制剂阻断Hh途径已被认为是治疗该癌症的治疗靶标。在我们之前的工作中,我们获得了一种新的基于嘌呤支架的SMO配体(化合物I),在几种癌细胞系中显示出有趣的抗肿瘤活性。在这项工作中,我们报告了17种新嘌呤衍生物的设计和合成,与吉西他滨相比,其中一些对Mia-PaCa-2(Hh依赖性胰腺癌细胞系)具有高细胞毒性作用,对非肿瘤性HEK-293细胞具有低毒性,例如8F,8g和8h(IC50=4.56,4.11和3.08μM,分别)。这些嘌呤中的两个还显示了它们通过NanoBRET测定结合SMO的能力(pKi=5.17对于8f和5.01对于8h),对化合物I具有更高的亲和力(pKi=1.51)。此外,对接研究提供了嘌呤取代模式与SMO亲和力相关的见解。最后,对所选嘌呤的Hh抑制研究,使用基于NIH3T3Shh-LightII细胞中荧光素酶活性的转录功能测定,证明了8g在两个特定的Hh依赖性细胞模型中降低了GLI活性,IC50=6.4μM,并降低了Hh靶基因的表达,Med1细胞和Ptch1-/-小鼠胚胎成纤维细胞。因此,我们的研究结果为SMO配体的设计提供了平台,这些配体可能是治疗胰腺癌的潜在选择性细胞毒性剂.
    Aberrant activation of the Hedgehog (Hh) signalling pathway has been associated with the development and progression of pancreatic cancer. For this reason, blockade of Hh pathway by inhibitors targeting the G protein-coupled receptor Smoothened (SMO) has been considered as a therapeutic target for the treatment of this cancer. In our previous work, we obtained a new SMO ligand based on a purine scaffold (compound I), which showed interesting antitumor activity in several cancer cell lines. In this work, we report the design and synthesis of 17 new purine derivatives, some of which showed high cytotoxic effect on Mia-PaCa-2 (Hh-dependent pancreatic cancer cell lines) and low toxicity on non-neoplastic HEK-293 cells compared with gemcitabine, such as 8f, 8g and 8h (IC50 = 4.56, 4.11 and 3.08 μM, respectively). Two of these purines also showed their ability to bind to SMO through NanoBRET assays (pKi = 5.17 for 8f and 5.01 for 8h), with higher affinities to compound I (pKi = 1.51). In addition, docking studies provided insight the purine substitution pattern is related to the affinity on SMO. Finally, studies of Hh inhibition for selected purines, using a transcriptional functional assay based on luciferase activity in NIH3T3 Shh-Light II cells, demonstrated that 8g reduced GLI activity with a IC50 = 6.4 μM as well as diminished the expression of Hh target genes in two specific Hh-dependent cell models, Med1 cells and Ptch1-/- mouse embryonic fibroblasts. Therefore, our results provide a platform for the design of SMO ligands that could be potential selective cytotoxic agents for the treatment of pancreatic 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
    目的探讨BOC对胶质母细胞瘤细胞的作用及其机制。体外,在神经胶质瘤细胞系中进行BOC敲除。CCK-8和Transwell用于评估BOC对生存能力的影响,入侵,和神经胶质细胞的迁移。采用RNA-seq技术分析BOC敲低胶质瘤细胞与对照组的差异基因表达,qRT-PCR用于验证下游差异基因的表达。进行SMO过表达以研究SMO对神经胶质瘤细胞的影响。BOC敲低小鼠皮下肿瘤模型用于验证BOC对小鼠肿瘤的作用。组织芯片技术用于检测正常人脑组织和胶质瘤组织中BOC和SMO的表达。体外,BOC敲除抑制了生存能力,入侵,和神经胶质瘤细胞的迁移,以及下调下游差异基因SMO的表达,EGFR,HRAS,和MRAS。相反,SMO过表达上调了生存力,入侵,和BOC敲低细胞的迁移能力。在体内,BOC敲低抑制小鼠肿瘤生长,下调下游差异基因SMO的表达,EGFR,HRAS,和MRAS。组织芯片结果显示BOC和SMO在胶质瘤组织中均高表达。BOC在神经胶质瘤患者中异常过度表达并促进神经胶质瘤的发展。机械上,BOC通过上调SMO的表达激活Hedgehog(Hh)和RAS信号通路,EGFR,HRAS,还有MRAS,从而促进扩散,胶质瘤细胞的侵袭和迁移。
    The purpose of this study was to investigate the effects of BOC on glioblastoma cells and its underlying mechanisms. In vitro, BOC-knockdown was performed in glioma cell lines. CCK-8 and Transwell were used to assess the impact of BOC on the viability, invasion, and migration of gliobma cells. RNA-seq technology was employed to analyze the differential gene expression between BOC-knockdown glioma cells and the control group, and qRT-PCR was used to validate the expression of downstream differential genes. SMO-overexpression was performed to investigate the effects of SMO on glioma cells. A BOC-knockdown mouse subcutaneous tumor model was to verify the effects of BOC on mouse tumors. Tissue microarray technology was used to detect the expression of BOC and SMO in samples of normal human brain tissue and glioma tissue. In vitro, BOC-knockdown inhibited the viability, invasion, and migration of glioma cells, as well as downregulated the expression of downstream differential genes SMO, EGFR, HRAS, and MRAS. Conversely, SMO-overexpression upregulated the viability, invasion, and migration abilities of BOC-knockdown cells. In vivo, BOC-knockdown suppressed tumor growth in mice and downregulated the expression of downstream differential genes SMO, EGFR, HRAS, and MRAS. Tissue microarray results showed that both BOC and SMO were highly expressed in glioma tissues. BOC is aberrantly overexpressed in glioma patients and promotes glioma development. Mechanistically, BOC activates the Hedgehog (Hh) and RAS signaling pathways by upregulating the expression of SMO, EGFR, HRAS, and MRAS, thereby facilitating the Proliferation, invasion and migration of glioma cells.
    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
    哮喘是儿童气道中最常见的慢性炎症性疾病。哮喘最普遍的表型是嗜酸性粒细胞性哮喘,这是由Th2免疫反应驱动的,可以通过吸入皮质类固醇治疗有效地管理。然而,有Th17免疫应答的哮喘表型对皮质类固醇治疗不敏感,表现出更严重的表型.这种对皮质类固醇不敏感的哮喘的治疗目前尚不成熟,需要进一步关注。本研究旨在阐明Hedgehog信号通路在哮喘Th17细胞分化中的调控作用。研究表明,Smo和Gli3都是Hedgehog信号通路的关键组成部分,在体外Th17极化和体内Th17显性哮喘模型中上调。发现用小分子抑制剂抑制Smo或基因敲除Gli3可以抑制Th17极化。发现Smo在Th1,Th2,Th17和Treg极化中增加,而Gli3在Th17极化中特异性增加。ChIP-qPCR分析表明,Gli3可以直接与T细胞中的IL-6相互作用,诱导STAT3磷酸化并促进Th17细胞分化。此外,该研究表明哮喘患儿Gli3表达升高与IL-17A和IL-6表达之间存在相关性.总之,研究表明,Hedgehog信号通路在哮喘的发病机制中起着重要作用,因为它通过IL-6/STAT3信号调节Th17细胞的分化。这可能为Th17细胞驱动的皮质类固醇不敏感哮喘提供潜在的治疗靶标。
    Asthma is the most prevalent chronic inflammatory disease of the airways in children. The most prevalent phenotype of asthma is eosinophilic asthma, which is driven by a Th2 immune response and can be effectively managed by inhaled corticosteroid therapy. However, there are phenotypes of asthma with Th17 immune response that are insensitive to corticosteroid therapy and manifest a more severe phenotype. The treatment of this corticosteroid-insensitive asthma is currently immature and requires further attention. The objective of this study is to elucidate the regulation of the Hedgehog signaling pathway in Th17 cell differentiation in asthma. The study demonstrated that both Smo and Gli3, key components of the Hedgehog signaling pathway, were upregulated in Th17 polarization in vitro and in a Th17-dominant asthma model in vivo. Inhibiting Smo with a small molecule inhibitor or genetically knocking down Gli3 was found to suppress Th17 polarization. Smo was found to increase in Th1, Th2, Th17 and Treg polarization, while Gli3 specifically increased in Th17 polarization. ChIP-qPCR analyses indicated that Gli3 can directly interact with IL-6 in T cells, inducing STAT3 phosphorylation and promoting Th17 cell differentiation. Furthermore, the study demonstrated a correlation between elevated Gli3 expression and IL-17A and IL-6 expression in children with asthma. In conclusion, the study demonstrated that the Hedgehog signaling pathway plays an important role in the pathogenesis of asthma, as it regulates the differentiation of Th17 cells through the IL-6/STAT3 signaling. This may provide a potential therapeutic target for corticosteroid-insensitive asthma driven by Th17 cells.
    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
    目的:除了视网膜母细胞瘤基因的作用外,与眼科肿瘤患者预后不良相关的基因组事件尚不清楚.
    方法:我们回顾性分析了48例六种类型的眼科肿瘤患者。我们使用联合外显子组和转录组分析在这些患者中搜索高频突变基因和易感基因。
    结果:我们确定了四个明显的致病基因(TP53,PTCH1,SMO,BAP1).易感性基因分析确定热点基因,包括RUNX1、APC、IDH2和BRCA2,高频基因分析确定了几个基因,包括TP53,TTN,MUC16转录组分析确定了5868个差异表达基因,其中TOP2A和ZWINT在所有样品中上调,而CFD,伊兰,HBA1和HBB下调。《京都基因百科全书》和基因组富集分析表明,癌症信号通路中的磷酸肌醇3-激酶(PI3K)-Akt和转录失调可能与眼科肿瘤发生有关。
    结论:TP53明显参与眼科肿瘤发生,尤其是基底细胞癌,PI3K-Akt信号通路可能是参与眼科肿瘤发生的重要通路。RUNX1,SMO,TOP2A,ZWINT也很可能参与眼科肿瘤发生,但是需要进一步的功能实验来验证这些基因在调节肿瘤发生中的机制。
    OBJECTIVE: Apart from the role of the retinoblastoma gene, the genomic events associated with poor outcomes in patients with ophthalmic tumors are poorly understood.
    METHODS: We retrospectively analyzed 48 patients with six types of ophthalmic tumors. We searched for high-frequency mutated genes and susceptibility genes in these patients using combined exome and transcriptome analysis.
    RESULTS: We identified four clearly causative genes (TP53, PTCH1, SMO, BAP1). Susceptibility gene analysis identified hotspot genes, including RUNX1, APC, IDH2, and BRCA2, and high-frequency gene analysis identified several genes, including TP53, TTN, and MUC16. Transcriptome analysis identified 5868 differentially expressed genes, of which TOP2A and ZWINT were upregulated in all samples, while CFD, ELANE, HBA1, and HBB were downregulated. Kyoto Encyclopedia of Genes and Genomes enrichment analysis indicated that the phosphoinositide 3-kinase (PI3K)-Akt and Transcriptional misregulation in cancer signaling pathways may be involved in ophthalmic tumorigenesis.
    CONCLUSIONS: TP53 is clearly involved in ophthalmic tumorigenesis, especially in basal cell carcinoma, and the PI3K-Akt signaling pathway may be an essential pathway involved in ophthalmic tumorigenesis. RUNX1, SMO, TOP2A, and ZWINT are also highly likely to be involved in ophthalmic tumorigenesis, but further functional experiments are needed to verify the mechanisms of these genes in regulating tumorigenesis.
    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
    在本期《细胞化学生物学》中,Liu等人1报道了Q29的鉴定,Q29是一种合成的二萜,可阻断平滑(SMO)的共价胆固醇修饰并抑制刺猬信号传导。Q29能够抑制肿瘤细胞生长,在体外和体内,克服对SMO抑制剂的抗性。
    In this issue of Cell Chemical Biology, Liu et al.1 report the identification of Q29, a synthetic diterpenoid that blocks covalent cholesterol modification of smoothened (SMO) and inhibits hedgehog signaling. Q29 is capable of suppressing tumor cell growth, both in vitro and in vivo, and overcoming resistance to SMO inhibitors.
    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
    刺猬(Hh)信号,进化上保守的途径,在发育和肿瘤发生中起着至关重要的作用,使其成为有希望的药物靶标。已知多个负调节器来控制Hh信号;然而,活化平滑(SMO)如何参与下游GLI2和GLI3的活化尚不清楚.在这里,我们确定纤毛激酶DYRK2是GLI2和GLI3转录因子对Hh信号传导的正调节因子.转录组和相互作用组分析表明,DYRK2在纤毛碱基的进化上保守的丝氨酸残基上磷酸化GLI2和GLI3,响应于Hh途径的激活。这种磷酸化诱导GLI2/GLI3从抑制剂解离,SUFU,以及它们向细胞核的易位。小鼠体内Dyrk2的缺失会导致骨骼畸形,但神经管发育仍正常.值得注意的是,DYRK2介导的磷酸化通过控制细胞增殖来协调肢体发育。一起来看,纤毛激酶DYRK2通过调节两个过程来控制Hh信号的激活:SMO和纤毛形成下游的GLI2和GLI3的磷酸化。因此,我们对Hh信号的独特调控机制的发现扩大了对Hh相关疾病控制的认识.
    Hedgehog (Hh) signaling, an evolutionarily conserved pathway, plays an essential role in development and tumorigenesis, making it a promising drug target. Multiple negative regulators are known to govern Hh signaling; however, how activated Smoothened (SMO) participates in the activation of downstream GLI2 and GLI3 remains unclear. Herein, we identified the ciliary kinase DYRK2 as a positive regulator of the GLI2 and GLI3 transcription factors for Hh signaling. Transcriptome and interactome analyses demonstrated that DYRK2 phosphorylates GLI2 and GLI3 on evolutionarily conserved serine residues at the ciliary base, in response to activation of the Hh pathway. This phosphorylation induces the dissociation of GLI2/GLI3 from suppressor, SUFU, and their translocation into the nucleus. Loss of Dyrk2 in mice causes skeletal malformation, but neural tube development remains normal. Notably, DYRK2-mediated phosphorylation orchestrates limb development by controlling cell proliferation. Taken together, the ciliary kinase DYRK2 governs the activation of Hh signaling through the regulation of two processes: phosphorylation of GLI2 and GLI3 downstream of SMO and cilia formation. Thus, our findings of a unique regulatory mechanism of Hh signaling expand understanding of the control of Hh-associated diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号