MYC

Myc
  • 文章类型: Journal Article
    尽管在治疗皮肤黑色素瘤方面取得了进展,肢端和粘膜(A/M)黑色素瘤患者的治疗选择仍然有限,预后较差.我们分析了156例黑色素瘤(101例皮肤,28肢,和27粘膜)使用FoundationOne癌症基因特异性临床测试平台,并确定了新的,A/M黑色素瘤特定解剖部位的潜在靶向基因组改变(GA)。使用新的A/M黑色素瘤临床前模型,我们证明了与皮肤黑素瘤相关的几种GA和相应的致癌途径在A/M黑素瘤中具有相似的靶向性。其他改动,包括MYC和CRKL扩增,是A/M黑色素瘤特有的,并且对使用BRD4抑制剂JQ1或Src/ABL抑制剂达沙替尼的间接靶向敏感,分别。我们进一步确定了新的,可操作的A/M特定更改,包括体内对达沙替尼反应的粘膜黑色素瘤中NF2融合失活。我们的研究强调了皮肤和A/M黑色素瘤之间新的分子差异,在A/M内的不同解剖部位,这可能会改变这些罕见黑素瘤的临床试验和治疗模式。
    Despite advancements in treating cutaneous melanoma, patients with acral and mucosal (A/M) melanomas still have limited therapeutic options and poor prognoses. We analyzed 156 melanomas (101 cutaneous, 28 acral, and 27 mucosal) using the Foundation One cancer-gene specific clinical testing platform and identified new, potentially targetable genomic alterations (GAs) in specific anatomic sites of A/M melanomas. Using novel pre-clinical models of A/M melanoma, we demonstrate that several GAs and corresponding oncogenic pathways associated with cutaneous melanomas are similarly targetable in A/M melanomas. Other alterations, including MYC and CRKL amplifications, were unique to A/M melanomas and susceptible to indirect targeting using the BRD4 inhibitor JQ1 or Src/ABL inhibitor dasatinib, respectively. We further identified new, actionable A/M-specific alterations, including an inactivating NF2 fusion in a mucosal melanoma responsive to dasatinib in vivo. Our study highlights new molecular differences between cutaneous and A/M melanomas, and across different anatomic sites within A/M, which may change clinical testing and treatment paradigms for these rare melanomas.
    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
    背景:LMO2是参与B细胞个体发育的相关基因,是侵袭性大B细胞淋巴瘤(aLBCL)的生存预测因子。大多数评估LMO2mRNA表达的研究依赖于微阵列平台或qRT-PCR方法,俯瞰组织形态。在这项研究中,我们通过显色原位杂交(CISH)在正常组织和一系列82aLBCL中评估LMO2RNA的表达。
    方法:在福尔马林固定石蜡包埋的组织中进行LMO2CISH,通过三种不同的方法得分,并与转录组相关。
    结果:我们获得了与LMO2蛋白表达和基因表达结果的评估方法相关的统计学显著结果。正常的扁桃体组织显示出高水平的LMO2,特别是在生发中心的光区内。相反,在aLBCL中,注意到LMO2表达显着降低,在携带MYC重排的情况下尤为明显。此外,通过总生存期和Cox回归生存分析获得了显著结果,纳入国际预后指数数据和LMO2表达水平。
    结论:我们显示了一种可靠的方法来鉴定CISH的LMO2mRNA表达,有效捕获LMO2的许多已报道的生物学特征。
    BACKGROUND: LMO2 is a relevant gene involved in B-cell ontogeny and a survival predictor of aggressive large B-cell lymphomas (aLBCL). Most studies assessing LMO2 mRNA expression have relied on microarray platforms or qRT-PCR methods, overlooking tissue morphology. In this study, we evaluate LMO2 RNA expression by chromogenic in situ hybridization (CISH) in normal tissue and in a series of 82 aLBCL.
    METHODS: LMO2 CISH was performed in formalin-fixed paraffin-embedded tissues, scored by three different methods, and correlated with a transcriptome panel.
    RESULTS: We obtained statistically significant results correlating the methods of evaluation with LMO2 protein expression and gene expression results. Normal tonsil tissue showed high levels of LMO2, particularly within the light zone of the germinal center. Conversely, in aLBCL, a notable reduction in LMO2 expression was noted, remarkably in cases carrying MYC rearrangements. Furthermore, significant results were obtained through overall survival and Cox regression survival analysis, incorporating International Prognostic Index data alongside LMO2 expression levels.
    CONCLUSIONS: We show a reliable method to identify LMO2 mRNA expression by CISH, effectively capturing many of the reported biologic features of LMO2.
    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
    心脏纤维化是各种心血管疾病中常见的病理生理过程。比如冠心病,高血压,和心肌病。心肌成纤维细胞转分化为肌成纤维细胞(MFs)是心肌纤维化的关键环节。LncRNAPVT1参与多个器官的纤维化疾病;然而,其在心脏纤维化中的作用和机制尚不清楚。用TGF-β1刺激人心肌成纤维细胞(HCFs)诱导成肌纤维细胞;免疫荧光染色,免疫印迹,用荧光原位杂交技术检测肌成纤维细胞表型和lncPVT1表达。CCK-8、流式细胞术检测PVT1敲低或过表达诱导的细胞生物学表型,和免疫印迹。使用异丙肾上腺素(ISO)诱导心肌纤维化的小鼠模型,心脏功能通过超声心动图检查,心脏组织由H&E,和Masson三色染色.在这项研究中,TGF-β1诱导HCF转化为肌成纤维细胞,表现为α-SMA水平显著增加,波形蛋白,胶原蛋白I,和胶原蛋白III;通过TGF-β1刺激,lncPVT1的表达水平显着增加。lncPVT1敲低也降低了TGF-β1,TGFBR1和TGFBR2的蛋白水平。在TGF-β1刺激下,lncPVT1敲低降低FN1、α-SMA、胶原蛋白I,和胶原蛋白III含量,抑制HCF细胞活力,增强细胞凋亡,并抑制Smad2/3磷酸化。在有或没有TGF-β1刺激的情况下,LncPVT1正调节MYC表达;在TGF-β1刺激的HCF中MYC过表达显着减弱了lncPVT1敲低对HCF增殖和转分化为MF的影响。在ISO诱导的心肌纤维化模型中,lncPVT1敲低部分减少纤维化面积,改善心脏功能,并降低纤维化标志物的水平。此外,lncPVT1敲低降低了小鼠心脏组织中的MYC和CDK4水平,但增加了E-cadherin。lncPVT1在心脏纤维化和TGF-β1刺激的HCFs中上调。LncPVT1敲低部分改善TGF-β1诱导的HCF活化和转分化为MF的体外和ISO诱导的心肌纤维化,可能通过与MYC相互作用和上调MYC。
    Cardiac fibrosis is a commonly seen pathophysiological process in various cardiovascular disorders, such as coronary heart disorder, hypertension, and cardiomyopathy. Cardiac fibroblast trans-differentiation into myofibroblasts (MFs) is a key link in myocardial fibrosis. LncRNA PVT1 participates in fibrotic diseases in multiple organs; however, its role and mechanism in cardiac fibrosis remain largely unknown. Human cardiac fibroblasts (HCFs) were stimulated with TGF-β1 to induce myofibroblast; Immunofluorescent staining, Immunoblotting, and fluorescence in situ hybridization were used to detect the myofibroblasts phenotypes and lnc PVT1 expression. Cell biological phenotypes induced by lnc PVT1 knockdown or overexpression were detected by CCK-8, flow cytometry, and Immunoblotting. A mouse model of myocardial fibrosis was induced using isoproterenol (ISO), and the cardiac functions were examined by echocardiography measurements, cardiac tissues by H&E, and Masson trichrome staining. In this study, TGF-β1 induced HCF transformation into myofibroblasts, as manifested as significantly increased levels of α-SMA, vimentin, collagen I, and collagen III; the expression level of lnc PVT1 expression showed to be significantly increased by TGF-β1 stimulation. The protein levels of TGF-β1, TGFBR1, and TGFBR2 were also decreased by lnc PVT1 knockdown. Under TGF-β1 stimulation, lnc PVT1 knockdown decreased FN1, α-SMA, collagen I, and collagen III protein contents, inhibited HCF cell viability and enhanced cell apoptosis, and inhibited Smad2/3 phosphorylation. Lnc PVT1 positively regulated MYC expression with or without TGF-β1 stimulation; MYC overexpression in TGF-β1-stimulated HCFs significantly attenuated the effects of lnc PVT1 knockdown on HCF proliferation and trans-differentiation to MFs. In the ISO-induced myocardial fibrosis model, lnc PVT1 knockdown partially reduced fibrotic area, improved cardiac functions, and decreased the levels of fibrotic markers. In addition, lnc PVT1 knockdown decreased MYC and CDK4 levels but increased E-cadherin in mice heart tissues. lnc PVT1 is up-regulated in cardiac fibrosis and TGF-β1-stimulated HCFs. Lnc PVT1 knockdown partially ameliorates TGF-β1-induced HCF activation and trans-differentiation into MFs in vitro and ISO-induced myocardial fibrosis in vivo, potentially through interacting with MYC and up-regulating MYC.
    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
    背景:KRAS突变经常发生在癌症中,尤其是胰腺导管腺癌,结直肠癌,和非小细胞肺癌。尽管最近批准了KRASG12C抑制剂,对于所有KRAS突变癌症,目前尚未建立有效的精准治疗.KRAS突变癌症的许多治疗方法,包括表观基因组靶向药物,目前正在调查中。小泛素样修饰蛋白(SUMO)是通过称为SUMO化和去SUMO化的过程与细胞中的其他蛋白质共价连接并分离的小蛋白质家族。我们评估了SUMO化抑制在KRAS突变的癌细胞中是否有效。
    方法:在多种人和小鼠KRAS突变的癌细胞系中评估了第一类SUMO激活酶E抑制剂TAK-981(subasumstat)的功效。使用TaqMan阵列的基因表达测定用于鉴定TAK-981功效的生物标志物。使用免疫印迹分析研究了SUMO化抑制的生物学作用和随后的调节机制,免疫荧光测定,和老鼠模型。
    结果:我们发现TAK-981下调了目前无法用药的MYC的表达,并有效抑制了不同组织类型中表达MYC的KRAS突变癌症的生长。此外,通过MYC过表达使TAK-981抗性细胞对SUMO化抑制敏感。TAK-981通过改变SUMO化和泛素化之间的平衡并促进泛素-蛋白酶体系统中的关键因素MYC和Fbxw7的结合来诱导MYC的蛋白酶体降解。在使用小鼠来源的CMT167细胞系的免疫活性和免疫缺陷小鼠模型中,TAK-981单一疗法的功效是显著的但适度的。由于MAPK抑制KRAS下游途径在KRAS突变癌症中至关重要,我们预计SUMO化和MEK的共同抑制可能是一个很好的选择。令人惊讶的是,TAK-981和曲美替尼联合治疗可显著诱导多种细胞系和基因工程小鼠来源的类器官细胞凋亡.此外,在使用不同组织类型的细胞系的小鼠模型中,联合治疗导致肿瘤长期消退.最后,我们发现联合治疗可补充抑制Rad51和BRCA1并累积DNA损伤.
    结论:我们发现在KRAS突变的癌细胞中,MYC通过SUMO化抑制发生下调。我们的发现表明,SUMO化和MEK的双重抑制可能是通过增强DNA损伤积累来治疗表达MYC的KRAS突变癌症的有希望的治疗方法。
    BACKGROUND: KRAS mutations frequently occur in cancers, particularly pancreatic ductal adenocarcinoma, colorectal cancer, and non-small cell lung cancer. Although KRASG12C inhibitors have recently been approved, effective precision therapies have not yet been established for all KRAS-mutant cancers. Many treatments for KRAS-mutant cancers, including epigenome-targeted drugs, are currently under investigation. Small ubiquitin-like modifier (SUMO) proteins are a family of small proteins covalently attached to and detached from other proteins in cells via the processes called SUMOylation and de-SUMOylation. We assessed whether SUMOylation inhibition was effective in KRAS-mutant cancer cells.
    METHODS: The efficacy of the first-in-class SUMO-activating enzyme E inhibitor TAK-981 (subasumstat) was assessed in multiple human and mouse KRAS-mutated cancer cell lines. A gene expression assay using a TaqMan array was used to identify biomarkers of TAK-981 efficacy. The biological roles of SUMOylation inhibition and subsequent regulatory mechanisms were investigated using immunoblot analysis, immunofluorescence assays, and mouse models.
    RESULTS: We discovered that TAK-981 downregulated the expression of the currently undruggable MYC and effectively suppressed the growth of MYC-expressing KRAS-mutant cancers across different tissue types. Moreover, TAK-981-resistant cells were sensitized to SUMOylation inhibition via MYC-overexpression. TAK-981 induced proteasomal degradation of MYC by altering the balance between SUMOylation and ubiquitination and promoting the binding of MYC and Fbxw7, a key factor in the ubiquitin-proteasome system. The efficacy of TAK-981 monotherapy in immunocompetent and immunodeficient mouse models using a mouse-derived CMT167 cell line was significant but modest. Since MAPK inhibition of the KRAS downstream pathway is crucial in KRAS-mutant cancer, we expected that co-inhibition of SUMOylation and MEK might be a good option. Surprisingly, combination treatment with TAK-981 and trametinib dramatically induced apoptosis in multiple cell lines and gene-engineered mouse-derived organoids. Moreover, combination therapy resulted in long-term tumor regression in mouse models using cell lines of different tissue types. Finally, we revealed that combination therapy complementally inhibited Rad51 and BRCA1 and accumulated DNA damage.
    CONCLUSIONS: We found that MYC downregulation occurred via SUMOylation inhibition in KRAS-mutant cancer cells. Our findings indicate that dual inhibition of SUMOylation and MEK may be a promising treatment for MYC-expressing KRAS-mutant cancers by enhancing DNA damage accumulation.
    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
    通常在多种肿瘤中上调的78kDa葡萄糖调节蛋白(GRP78)是重要的预后标志物,也是抑制肿瘤发生和治疗抗性的有希望的靶标。虽然GRP78已被公认为具有抗凋亡特性的主要内质网(ER)伴侣和未折叠蛋白反应的主要调节因子,它作为癌蛋白表达调节剂的新作用刚刚出现。MYC在约70%的人类癌症中失调,是最常见的活化癌蛋白。然而,尽管最近取得了进展,MYC的治疗靶向仍然具有挑战性.在这里,我们将GRP78确定为抑制MYC表达的新靶标。使用多种MYC依赖性癌症模型,包括头颈部鳞状细胞癌及其顺铂耐药克隆,乳腺和胰腺腺癌,我们的研究表明,通过siRNA敲低GRP78或通过小分子抑制剂(YUM70或HA15)抑制其活性降低c-MYC表达,导致细胞凋亡和细胞活力的丧失。这在2D细胞培养中观察到,3D球体和异种移植模型。机械上,我们确定c-MYC的抑制处于转录后水平,YUM70和HA15处理有效上调真核翻译抑制剂4E-BP1,其靶向对c-MYC翻译起始至关重要的eIF4E.此外,通过siRNA敲除4E-BP1拯救了YUM70介导的c-MYC抑制。由于YUM70还能够抑制N-MYC表达,这项研究提供了一种通过敲低或抑制GRP78来抑制MYC蛋白表达的新方法。
    The 78-kDa glucose regulated protein (GRP78) commonly upregulated in a wide variety of tumors is an important prognostic marker and a promising target for suppressing tumorigenesis and treatment resistance. While GRP78 is well established as a major endoplasmic reticulum (ER) chaperone with anti-apoptotic properties and a master regulator of the unfolded protein response, its new role as a regulator of oncoprotein expression is just emerging. MYC is dysregulated in about 70 % of human cancers and is the most commonly activated oncoprotein. However, despite recent advances, therapeutic targeting of MYC remains challenging. Here we identify GRP78 as a new target for suppression of MYC expression. Using multiple MYC-dependent cancer models including head and neck squamous cell carcinoma and their cisplatin-resistant clones, breast and pancreatic adenocarcinoma, our studies revealed that GRP78 knockdown by siRNA or inhibition of its activity by small molecule inhibitors (YUM70 or HA15) reduced c-MYC expression, leading to onset of apoptosis and loss of cell viability. This was observed in 2D cell culture, 3D spheroid and in xenograft models. Mechanistically, we determined that the suppression of c-MYC is at the post-transcriptional level and that YUM70 and HA15 treatment potently upregulated the eukaryotic translation inhibitor 4E-BP1, which targets eIF4E critical for c-MYC translation initiation. Furthermore, knock-down of 4E-BP1 via siRNA rescued YUM70-mediated c-MYC suppression. As YUM70 is also capable of suppressing N-MYC expression, this study offers a new approach to suppress MYC protein expression through knockdown or inhibition of GRP78.
    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
    在受伤的斑马鱼视网膜中,Müller胶质细胞(MG)重新编程以采用视网膜干细胞特性并再生受损的神经元。最强的斑马鱼重编程因子可能是刺激哺乳动物MG类似再生反应的良好候选者。Myc蛋白是有效的重编程因子,可以刺激分化细胞的细胞可塑性;然而,它们在MG重编程和视网膜再生中的作用尚不清楚。在这里,我们报道了视网膜损伤刺激mycb和mych表达,尽管Mycb和Mych都刺激MG重编程和增殖,只有Mych增强视网膜神经元凋亡。Wt的RNAseq分析,mychmut,Mycbmut鱼显示Mycb和Mych调节40%和16%,分别,有助于MG再生相关转录组的基因。在这些基因中,那些被诱导的偏向于调节核糖体生物发生,蛋白质合成,DNA合成,和细胞分裂是受视网膜损伤调节的顶级细胞过程,这表明Mycb和Mych是有效的MG重编程因子。与此一致,这些蛋白质中的任一种的强制表达足以刺激未损伤的视网膜中的MG增殖。
    In the injured zebrafish retina, Müller glial cells (MG) reprogram to adopt retinal stem cell properties and regenerate damaged neurons. The strongest zebrafish reprogramming factors might be good candidates for stimulating a similar regenerative response by mammalian MG. Myc proteins are potent reprogramming factors that can stimulate cellular plasticity in differentiated cells; however, their role in MG reprogramming and retina regeneration remains poorly explored. Here we report that retinal injury stimulates mycb and mych expression and that although both Mycb and Mych stimulate MG reprogramming and proliferation, only Mych enhances retinal neuron apoptosis. RNAseq analysis of Wt, mychmut, and mycbmut fish revealed Mycb and Mych regulate ∼40% and ∼16%, respectively, of the genes contributing to MG\'s regeneration-associated transcriptome. Of these genes, those that are induced are biased towards regulating ribosome biogenesis, protein synthesis, DNA synthesis, and cell division which are the top cellular processes regulated by retinal injury and this suggests Mycb and Mych are potent MG reprogramming factors. Consistent with this, forced expression of either of these proteins is sufficient to stimulate MG proliferation in the uninjured retina.
    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:WNT信号传导是空间组织排列的核心,调节干细胞活性,并代表胃肠道癌症的标志。虽然它在驱动肠道肿瘤中的作用是很好的特征,WNT在胃癌发生中的作用仍然难以捉摸。
    方法:我们开发了小鼠模型来控制B-CATENIN致癌形式的特异性表达,并结合MYC在胃窦的Lgr5+细胞中的激活。我们使用在体内和类器官模型中应用的多组学方法来表征它们在驱动胃肿瘤发生中的合作。
    结果:我们报道了胃中的组成型B-CATENIN稳定具有可忽略的致癌作用,并且需要激活MYC来诱导胃肿瘤形成。虽然胃腺中生理上较低的MYC水平限制了B-CATENIN转录活性,MYC表达的增加释放了WNT致癌转录程序,在没有直接转录合作的情况下促进B-CATENIN增强子入侵。MYC活化通过mTOR和ERK活化以及MiT/TFE抑制诱导抑制溶酶体生物发生的代谢重组。这可以防止EPCAM通过巨噬细胞增多而降解,促进B-CATENIN染色质积累和WNT致癌转录的激活。
    结论:我们的结果揭示了一个新的信号传导框架,对控制胃上皮结构和WNT依赖性致癌转化具有重要意义。
    OBJECTIVE: WNT signaling is central to spatial tissue arrangement, regulating stem cell activity, and represents the hallmark of gastrointestinal cancers. While its role in driving intestinal tumors is well characterized, WNT\'s role in gastric tumorigenesis remains elusive.
    METHODS: We have developed mouse models to control the specific expression of an oncogenic form of B-CATENIN in combination with MYC activation in Lgr5+ cells of the gastric antrum. We used multi-omics approaches applied in vivo and in organoid models to characterize their cooperation in driving gastric tumorigenesis.
    RESULTS: We report that constitutive B-CATENIN stabilization in the stomach has negligible oncogenic effects and requires MYC activation to induce gastric tumour formation. While physiologically low MYC levels in gastric glands limit B-CATENIN transcriptional activity, increased MYC expression unleashes the WNT oncogenic transcriptional program, promoting B-CATENIN enhancer invasion without a direct transcriptional cooperation. MYC activation induces a metabolic rewiring that suppresses lysosomal biogenesis through mTOR and ERK activation and MiT/TFE inhibition. This prevents EPCAM degradation by macropinocytosis, promoting B-CATENIN chromatin accumulation and activation of WNT oncogenic transcription.
    CONCLUSIONS: Our results uncovered a new signaling framework with important implications for the control of gastric epithelial architecture and WNT-dependent oncogenic transformation.
    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
    MYC与多种人类肿瘤的发病机理有关,多年来一直被描述为一种转录因子,可调节具有多效性功能的基因以促进致瘤生长。然而,尽管在确定MYC的特定靶基因方面进行了大量努力,这些靶基因可能单独促进肿瘤发生,该领域尚未达成共识,这是否是MYC的关键功能。最近的工作将对MYC功能的看法从基因特异性转录因子转变为必需的应激复原因子。在高度增殖的细胞中,MYC通过促进核心启动子的DNA修复来保持细胞完整性,保护停滞的复制叉,和/或防止转录-复制冲突。此外,越来越多的证据表明,MYC不仅通过驱动细胞自主生长来促进肿瘤发生,还能使肿瘤逃避宿主的免疫系统。在这次审查中,我们总结了我们目前对MYC如何损害抗肿瘤免疫的理解,以及为什么这种功能在进化上与MYC蛋白家族的生物学联系在一起。我们展示了为什么MYC的细胞自主和免疫逃避功能是相互依赖的,并讨论了在癌症治疗中靶向MYC蛋白的方法。
    MYC has been implicated in the pathogenesis of a wide range of human tumors and has been described for many years as a transcription factor that regulates genes with pleiotropic functions to promote tumorigenic growth. However, despite extensive efforts to identify specific target genes of MYC that alone could be responsible for promoting tumorigenesis, the field is yet to reach a consensus whether this is the crucial function of MYC. Recent work shifts the view on MYC\'s function from being a gene-specific transcription factor to an essential stress resilience factor. In highly proliferating cells, MYC preserves cell integrity by promoting DNA repair at core promoters, protecting stalled replication forks, and/or preventing transcription-replication conflicts. Furthermore, an increasing body of evidence demonstrates that MYC not only promotes tumorigenesis by driving cell-autonomous growth, but also enables tumors to evade the host\'s immune system. In this review, we summarize our current understanding of how MYC impairs antitumor immunity and why this function is evolutionarily hard-wired to the biology of the MYC protein family. We show why the cell-autonomous and immune evasive functions of MYC are mutually dependent and discuss ways to target MYC proteins in cancer therapy.
    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
    人类骨肉瘤(OS)的特征在于TP53基因的改变。在老鼠身上,p53的丢失触发OS的发展,其中c-Myc(Myc)致瘤性是不可或缺的。然而,关于Myc靶向哪些基因促进肿瘤发生,人们知之甚少。这里,我们研究了γ-谷氨酰环基转移酶(Ggct)的作用,Ggct是谷胱甘肽稳态所必需的γ-谷氨酰循环的组成酶,在人类和小鼠操作系统开发中。我们发现GGCT是人类OS的不良预后因素,Ggct的缺失抑制了小鼠p53缺陷的骨分化。Myc通过与Ggct启动子结合直接上调Ggct,和通过基因组编辑缺失其中的Myc结合位点减弱了p53缺陷型OS细胞的致瘤潜能。一起来看,这些结果表明了GGCT在癌细胞中广泛上调并巩固了其作为抗癌药物靶标的适用性的基本原理。
    Osteosarcoma (OS) in humans is characterized by alterations in the TP53 gene. In mice, loss of p53 triggers OS development, for which c-Myc (Myc) oncogenicity is indispensable. However, little is known about which genes are targeted by Myc to promote tumorigenesis. Here, we examined the role of γ-glutamylcyclotransferase (Ggct) which is a component enzyme of the γ-glutamyl cycle essential for glutathione homeostasis, in human and mouse OS development. We found that GGCT is a poor prognostic factor for human OS, and that deletion of Ggct suppresses p53-deficient osteosarcomagenesis in mice. Myc upregulates Ggct directly by binding to the Ggct promoter, and deletion of a Myc binding site therein by genome editing attenuated the tumorigenic potential of p53-deficient OS cells. Taken together, these results show a rationale that GGCT is widely upregulated in cancer cells and solidify its suitability as a target for anticancer drugs.
    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
    高级别B细胞淋巴瘤(HGBCL),非霍奇金淋巴瘤的亚型,患者在初始治疗或挽救性化疗后复发或难治性。MYC和BCL2的双重失调是其重要致病机制之一。因此,MYC和BCL2的联合靶向似乎是一个有前途的策略。二氢乳清酸脱氢酶(DHODH)是嘧啶从头生物合成的第四限速酶。它已被证明是多种疾病的潜在治疗靶标。在这项研究中,DHODH抑制剂brequinar表现出生长抑制,细胞周期阻断,MYC和BCL2重排促进HGBCL细胞系的凋亡。brequinar和BCL2抑制剂venetoclax通过不同途径对DHL细胞的存活具有协同抑制作用。维奈托克可以上调MCL-1和MYC的表达,已被报道为BCL2抑制剂的耐药机制。Brequinar下调MCL-1和MYC,这可能会克服HGBCL细胞对维奈托克的耐药性。此外,布雷那可以下调广泛的基因,包括核糖体生物合成基因,这可能有助于其抗肿瘤作用。体内研究表明,在布基那和维奈托克联合治疗的异种移植模型中,肿瘤生长具有协同抑制作用。这些结果为在MYC和BCL2异常的HGBCL中合理组合DHODH和BCL2阻断提供了初步证据。
    High-grade B-cell lymphoma (HGBCL), the subtype of non-Hodgkin lymphoma, to be relapsed or refractory in patients after initial therapy or salvage chemotherapy. Dual dysregulation of MYC and BCL2 is one of the important pathogenic mechanisms. Thus, combined targeting of MYC and BCL2 appears to be a promising strategy. Dihydroorotate dehydrogenase (DHODH) is the fourth rate-limiting enzyme for the de novo biosynthesis of pyrimidine. It has been shown to be a potential therapeutic target for multiple diseases. In this study, the DHODH inhibitor brequinar exhibited growth inhibition, cell cycle blockade, and apoptosis promotion in HGBCL cell lines with MYC and BCL2 rearrangements. The combination of brequinar and BCL2 inhibitors venetoclax had a synergistic inhibitory effect on the survival of DHL cells through different pathways. Venetoclax could upregulate MCL-1 and MYC expression, which has been reported as a resistance mechanism of BCL2 inhibitors. Brequinar downregulated MCL-1 and MYC, which could potentially overcome drug resistance to venetoclax in HGBCL cells. Furthermore, brequinar could downregulate a broad range of genes, including ribosome biosynthesis genes, which might contribute to its anti-tumor effects. In vivo studies demonstrated synergetic tumor growth inhibition in xenograft models with brequinar and venetoclax combination treatment. These results provide preliminary evidence for the rational combination of DHODH and BCL2 blockade in HGBCL with abnormal MYC and BCL2.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号