DNA-Activated Protein Kinase

DNA 活化蛋白激酶
  • 文章类型: Journal Article
    将HIV-1基因组的DNA拷贝整合到细胞基因组中会导致一系列的损伤,修复对于病毒的成功复制至关重要。我们以前已经证明了ATM和DNA-PK激酶,通常负责修复细胞DNA中的双链断裂,需要启动HIV-1DNA整合后修复,即使整合不会导致DNA双链断裂。在这项研究中,我们分析了ATM磷酸化状态的变化(pSer1981),DNA-PK(pSer2056),及其相关激酶ATR(pSer428),以及他们的目标:Chk1(pSer345),Chk2(pThr68),H2AX(pSer139),和p53(pSer15)在HIV-1DNA整合后修复。我们已经证明ATM和DNA-PK,但不是ATR,在整合后的DNA修复过程中经历自磷酸化,并磷酸化其靶蛋白Chk2和H2AX。这些数据表明了HIV-1DNA的双链DNA断裂修复和整合后修复之间的共同信号机制。
    Integration of the DNA copy of HIV-1 genome into the cellular genome results in series of damages, repair of which is critical for successful replication of the virus. We have previously demonstrated that the ATM and DNA-PK kinases, normally responsible for repairing double-strand breaks in the cellular DNA, are required to initiate the HIV-1 DNA postintegrational repair, even though integration does not result in DNA double-strand breaks. In this study, we analyzed changes in phosphorylation status of ATM (pSer1981), DNA-PK (pSer2056), and their related kinase ATR (pSer428), as well as their targets: Chk1 (pSer345), Chk2 (pThr68), H2AX (pSer139), and p53 (pSer15) during the HIV-1 DNA postintegrational repair. We have shown that ATM and DNA-PK, but not ATR, undergo autophosphorylation during postintegrational DNA repair and phosphorylate their target proteins Chk2 and H2AX. These data indicate common signaling mechanisms between the double-strand DNA break repair and postintegrational repair of HIV-1 DNA.
    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
    为了克服人软骨肉瘤细胞对放射治疗的抵抗力,用DNA依赖性蛋白激酶催化亚基(DNA-PKcs)抑制剂AZD7648进行碳离子(C-离子)和参考光子(X射线)照射(IR)的联合治疗来预防有效的DNA修复基因表达分析,流式细胞术,蛋白质磷酸化,端粒长度缩短。联合治疗后细胞增殖标志物和细胞周期分布显著改变,揭示了一个突出的G2/M逮捕。G2/M检查点基因细胞周期蛋白B的表达,通过单独的IR和联合治疗,CDK1和WEE1显着降低。虽然单独的IR没有显示出影响,额外的AZD7648治疗导致AKT磷酸化的剂量依赖性减少和Chk2磷酸化的增加.IR后24小时,联合治疗减少了DNA修复机制的关键基因,导致DNA修复受损和放射敏感性增加。用AZD7648和8GyX射线/C-离子IR联合处理后,在两种细胞系中均观察到端粒长度的时间依赖性缩短。我们的数据表明,抑制DNA-PKcs可能会削弱其在DNA修复机制和端粒末端保护中的功能,从而增加对X射线和C离子IR的敏感性。
    In order to overcome the resistance to radiotherapy in human chondrosarcoma cells, the prevention from efficient DNA repair with a combined treatment with the DNA-dependent protein kinase catalytic subunit (DNA-PKcs) inhibitor AZD7648 was explored for carbon ion (C-ion) as well as reference photon (X-ray) irradiation (IR) using gene expression analysis, flow cytometry, protein phosphorylation, and telomere length shortening. Proliferation markers and cell cycle distribution changed significantly after combined treatment, revealing a prominent G2/M arrest. The expression of the G2/M checkpoint genes cyclin B, CDK1, and WEE1 was significantly reduced by IR alone and the combined treatment. While IR alone showed no effects, additional AZD7648 treatment resulted in a dose-dependent reduction in AKT phosphorylation and an increase in Chk2 phosphorylation. Twenty-four hours after IR, the key genes of DNA repair mechanisms were reduced by the combined treatment, which led to impaired DNA repair and increased radiosensitivity. A time-dependent shortening of telomere length was observed in both cell lines after combined treatment with AZD7648 and 8 Gy X-ray/C-ion IR. Our data suggest that the inhibition of DNA-PKcs may increase sensitivity to X-rays and C-ion IR by impairing its functional role in DNA repair mechanisms and telomere end protection.
    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
    (1)头颈部鳞状细胞癌(HNSCC)是常见的,虽然治疗很困难,死亡率很高。激酶抑制剂有望增强放射治疗的效果。我们比较了PARP抑制剂talazoparib和niraparib以及DNA-PKcs抑制剂AZD7648联合电离辐射的效果。(2)七个HNSCC细胞系,包括Cal33,CLS-354,底特律562,HSC4,RPMI2650(HPV阴性),UD-SCC-2和UM-SCC-47(HPV阳性),和两个健康的成纤维细胞细胞系,研究了SBLF8和SBLF9。流式细胞术用于分析凋亡和坏死诱导(AnnexinV/7AAD)和细胞周期分布(Hoechst)。通过集落形成测定法研究细胞失活。(3)AZD7648效应最强,放射增敏所有HNSCC细胞系,几乎总是以超相加的方式。Talazoparib和niraparib在两种HPV阳性细胞系中均有效,但仅在一种和两种HPV阴性细胞系中均有效。分别。健康的成纤维细胞不受凋亡和坏死诱导或G2/M期停滞的任何联合治疗的影响。AZD7648单独对健康成纤维细胞没有毒性,而与电离辐射的结合降低了克隆性。(4)总之,talazoparib,尼拉帕利和,最有力的,AZD7648可以改善HNSCC的放射治疗。健康的成纤维细胞单独耐受AZD7648非常好,但是辐射诱导的效应可能会发生。我们的结果证明了体内研究的正确性。
    (1) Head and neck squamous cell carcinoma (HNSCC) is common, while treatment is difficult, and mortality is high. Kinase inhibitors are promising to enhance the effects of radiotherapy. We compared the effects of the PARP inhibitors talazoparib and niraparib and that of the DNA-PKcs inhibitor AZD7648, combined with ionizing radiation. (2) Seven HNSCC cell lines, including Cal33, CLS-354, Detroit 562, HSC4, RPMI2650 (HPV-negative), UD-SCC-2 and UM-SCC-47 (HPV-positive), and two healthy fibroblast cell lines, SBLF8 and SBLF9, were studied. Flow cytometry was used to analyze apoptosis and necrosis induction (AnnexinV/7AAD) and cell cycle distribution (Hoechst). Cell inactivation was studied by the colony-forming assay. (3) AZD7648 had the strongest effects, radiosensitizing all HNSCC cell lines, almost always in a supra-additive manner. Talazoparib and niraparib were effective in both HPV-positive cell lines but only consistently in one and two HPV-negative cell lines, respectively. Healthy fibroblasts were not affected by any combined treatment in apoptosis and necrosis induction or G2/M-phase arrest. AZD7648 alone was not toxic to healthy fibroblasts, while the combination with ionizing radiation reduced clonogenicity. (4) In conclusion, talazoparib, niraparib and, most potently, AZD7648 could improve radiation therapy in HNSCC. Healthy fibroblasts tolerated AZD7648 alone extremely well, but irradiation-induced effects might occur. Our results justify in vivo studies.
    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
    核糖核苷酸还原酶(RNR)是脱氧核糖核苷三磷酸(dNTP)生产中的限速酶,它们是辐射损伤后DNA修复的重要底物。我们探索了RNR的放射增敏特性,并研究了一种选择性RRM2抑制剂,3-AP,作为治疗转移性pNETs的放射增敏剂。我们调查了RNR亚基的作用,RRM2,在胰腺神经内分泌(pNET)细胞中以及对体外辐射的反应。我们还评估了选择性RRM2亚基抑制剂,3-AP,作为体内治疗pNET转移的放射增敏剂。敲除RNR亚基表明RRM1和RRM2亚基,p53R3在细胞增殖中起重要作用。RRM2抑制通过ATM和DNA-PK蛋白激酶而不是ATR的磷酸化激活DDR途径。RRM2抑制也诱导Chk1和Chk2磷酸化,导致G1/S期细胞周期停滞。RRM2抑制使pNET细胞对放疗敏感并在体外诱导细胞凋亡。在体内,我们利用pNET皮下和肺转移模型来研究RNR靶向治疗和3-AP作为放射增敏剂治疗pNETs的基本原理.联合治疗显着增加BON(人pNET)异种移植物的凋亡,并显着减少肺转移的负担。一起,我们的结果表明,选择性RRM2抑制可在体内和体外诱导转移性pNETs的放射敏感性。因此,用选择性RRM2抑制剂治疗,3-AP,是一种有前途的放射增敏剂,用于转移性pNETs的治疗性医疗设备。
    Ribonucleotide Reductase (RNR) is a rate-limiting enzyme in the production of deoxyribonucleoside triphosphates (dNTPs), which are essential substrates for DNA repair after radiation damage. We explored the radiosensitization property of RNR and investigated a selective RRM2 inhibitor, 3-AP, as a radiosensitizer in the treatment of metastatic pNETs. We investigated the role of RNR subunit, RRM2, in pancreatic neuroendocrine (pNET) cells and responses to radiation in vitro. We also evaluated the selective RRM2 subunit inhibitor, 3-AP, as a radiosensitizer to treat pNET metastases in vivo. Knockdown of RNR subunits demonstrated that RRM1 and RRM2 subunits, but not p53R3, play significant roles in cell proliferation. RRM2 inhibition activated DDR pathways through phosphorylation of ATM and DNA-PK protein kinases but not ATR. RRM2 inhibition also induced Chk1 and Chk2 phosphorylation, resulting in G1/S phase cell cycle arrest. RRM2 inhibition sensitized pNET cells to radiotherapy and induced apoptosis in vitro. In vivo, we utilized pNET subcutaneous and lung metastasis models to examine the rationale for RNR-targeted therapy and 3-AP as a radiosensitizer in treating pNETs. Combination treatment significantly increased apoptosis of BON (human pNET) xenografts and significantly reduced the burden of lung metastases. Together, our results demonstrate that selective RRM2 inhibition induced radiosensitivity of metastatic pNETs both in vitro and in vivo. Therefore, treatment with the selective RRM2 inhibitor, 3-AP, is a promising radiosensitizer in the therapeutic armamentarium for metastatic pNETs.
    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
    三阴性乳腺癌(TNBC)仍然是最致命的乳腺癌亚型,其特点是对当前化疗的应答率很低,并且缺乏其他有效的治疗选择。虽然大约30%的患者对基于蒽环类和紫杉烷的标准治疗化疗方案反应良好,大多数患者的临床结果改善有限,强调了在TNBC中提高蒽环类/紫杉烷类化疗有效性的策略的迫切需要。在这项研究中,我们报道了DNA-PK抑制剂的潜力,peposertib,为了提高拓扑异构酶II(TOPOII)抑制剂的有效性,尤其是蒽环类药物,在TNBC。我们的体外研究证明了peposertib与多柔比星的协同抗增殖活性,多种TNBC细胞系中的表柔比星和依托泊苷。下游分析显示在联合治疗下ATM依赖性代偿信号和p53途径激活的诱导。这些体外发现通过在携带皮下植入肿瘤的小鼠中观察到的明显的抗肿瘤作用得到证实。我们建立了一种耐受性良好的临床前治疗方案,将peposertib与聚乙二醇化脂质体多柔比星(PLD)相结合,并在体内细胞系来源和患者来源的TNBC异种移植模型中显示出强大的抗肿瘤功效。一起来看,我们的研究结果提供了证据,证明与peposertib联合治疗有可能增强蒽环类/TOPOII型化疗的疗效,它为改善TNBC患者的治疗结果提供了一个有希望的策略。
    Triple-negative breast cancer (TNBC) remains the most lethal subtype of breast cancer, characterized by poor response rates to current chemotherapies and a lack of additional effective treatment options. While approximately 30% of patients respond well to anthracycline- and taxane-based standard-of-care chemotherapy regimens, the majority of patients experience limited improvements in clinical outcomes, highlighting the critical need for strategies to enhance the effectiveness of anthracycline/taxane-based chemotherapy in TNBC. In this study, we report on the potential of a DNA-PK inhibitor, peposertib, to improve the effectiveness of topoisomerase II (TOPO II) inhibitors, particularly anthracyclines, in TNBC. Our in vitro studies demonstrate the synergistic antiproliferative activity of peposertib in combination with doxorubicin, epirubicin and etoposide in multiple TNBC cell lines. Downstream analysis revealed the induction of ATM-dependent compensatory signaling and p53 pathway activation under combination treatment. These in vitro findings were substantiated by pronounced anti-tumor effects observed in mice bearing subcutaneously implanted tumors. We established a well-tolerated preclinical treatment regimen combining peposertib with pegylated liposomal doxorubicin (PLD) and demonstrated strong anti-tumor efficacy in cell-line-derived and patient-derived TNBC xenograft models in vivo. Taken together, our findings provide evidence that co-treatment with peposertib has the potential to enhance the efficacy of anthracycline/TOPO II-based chemotherapies, and it provides a promising strategy to improve treatment outcomes for TNBC patients.
    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:放射性肺纤维化(RIPF)是一种慢性,进步,放疗后发展的不可逆的肺间质疾病。尽管先前的一些研究集中在肺上皮细胞中上皮-间质转化(EMT)的机制上,参与这一过程的基本因素仍然知之甚少。当细胞遭受辐射诱导的损伤时,DNA依赖性蛋白激酶催化亚基(DNA-PKcs)表现出很强的修复能力;DNA-PKcs在RIPF期间是否调节EMT尚不清楚。
    目的:探讨DNA-PKcs在RIPF中的作用及分子机制,为利用DNA-PKcs靶向药物预防RIPF提供重要的理论依据。
    方法:通过Cas9/sgRNA技术产生DNA-PKcs敲除(DPK-/-)小鼠,并以20Gy剂量进行全胸部电离辐射(IR)。在整个胸部IR之前,小鼠胃内给药DNA-PKcs靶向药物VND3207.在IR后1和5个月收集肺组织。
    结果:肺纤维化(PF)患者中DNA-PKcs的表达较低。DNA-PKcs缺乏通过促进肺上皮细胞的EMT而显著加剧RIPF。机械上,shRNA或抑制剂NU7441的DNA-PKcs缺失维持了Twist1的蛋白质稳定性。此外,AKT1介导DNA-PKcs与Twist1的相互作用。胰岛素样生长因子-1(IGF-1)阻断了DNA-PKcs缺失引起的Twist1高表达和EMT相关变化,AKT1激动剂。辐射防护药物VND3207通过刺激DNA-PKcs的激酶活性来预防IR诱导的EMT并减轻小鼠的RIPF。
    结论:我们的研究阐明了DNA-PKcs在RIPF中的关键作用和机制,并表明它可能是预防RIPF的潜在靶标。
    BACKGROUND: Radiation-induced pulmonary fibrosis (RIPF) is a chronic, progressive, irreversible lung interstitial disease that develops after radiotherapy. Although several previous studies have focused on the mechanism of epithelial-mesenchymal transition (EMT) in lung epithelial cells, the essential factors involved in this process remain poorly understood. The DNA-dependent protein kinase catalytic subunit (DNA-PKcs) exhibits strong repair capacity when cells undergo radiation-induced damage; whether DNA-PKcs regulates EMT during RIPF remains unclear.
    OBJECTIVE: To investigate the role and molecular mechanism of DNA-PKcs in RIPF and provide an important theoretical basis for utilising DNA-PKcs-targeted drugs for preventing RIPF.
    METHODS: DNA-PKcs knockout (DPK-/-) mice were generated via the Cas9/sgRNA technique and subjected to whole chest ionizing radiation (IR) at a 20 Gy dose. Before whole chest IR, the mice were intragastrically administered the DNA-PKcs-targeted drug VND3207. Lung tissues were collected at 1 and 5 months after IR.
    RESULTS: The expression of DNA-PKcs is low in pulmonary fibrosis (PF) patients. DNA-PKcs deficiency significantly exacerbated RIPF by promoting EMT in lung epithelial cells. Mechanistically, DNA-PKcs deletion by shRNA or inhibitor NU7441 maintained the protein stability of Twist1. Furthermore, AKT1 mediated the interaction between DNA-PKcs and Twist1. High Twist1 expression and EMT-associated changes caused by DNA-PKcs deletion were blocked by insulin-like growth factor-1 (IGF-1), an AKT1 agonist. The radioprotective drug VND3207 prevented IR-induced EMT and alleviated RIPF in mice by stimulating the kinase activity of DNA-PKcs.
    CONCLUSIONS: Our study clarified the critical role and mechanism of DNA-PKcs in RIPF and showed that it could be a potential target for preventing RIPF.
    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
    放疗的有效性取决于“正常”细胞和癌细胞对所施用的辐射剂量的敏感性。通过抑制DNA损伤修复来提高癌症的放射敏感性是当前许多研究的目标。然而,成功取决于避免在治疗期间不可避免地照射正常组织的伴随致敏。在这项研究中,我们通过检查对增殖和静止细胞群体的影响,研究了DNA-PK和PARP抑制剂的放射增敏机制。在体外和体内在BRCA1/2null和野生型亲本癌症模型中进行实验。总体上,AZD7648相对于Olaparib具有更大的放射增敏活性,BRCA2缺陷模型显示出最大的敏感性。然而,DNA-PK抑制剂AZD7648在所有照射的小鼠中也产生更大的毒性。虽然DNA-PK和PARP抑制使野生型肿瘤细胞对辐射敏感,在BRCA1/2缺陷型细胞中,奥拉帕尼对PARP的抑制具有有限的放射增敏能力.静止细胞比增殖细胞更耐辐射,这些也被AZD7648有效致敏,而Olaparib无法增加辐射诱导的细胞杀伤,即使在BRCA1/2null单元格中。这些发现强调了DNA-PK和PARP抑制剂的放射增敏的不同机制。虽然DNA-PK抑制剂能够靶向增殖和非增殖肿瘤细胞,以获得更大的整体抗癌益处,它们的应用受到正常组织毒性恶化的限制。相反,PARP抑制剂对增殖细胞具有选择性活性,提供了一种靶向癌症的机制,但是由于非增殖细胞的活性差,它们对肿瘤生长控制的影响总体上降低了。这项研究强调了用DNA损伤修复抑制辐射致敏策略创建治疗比例的重要性。
    The effectiveness of radiotherapy depends on the sensitivities of \'normal\' and cancer cells to the administered radiation dose. Increasing the radiosensitivity of cancers by inhibiting DNA damage repair is a goal of much current research, however success depends on avoiding concomitant sensitization of normal tissues inevitably irradiated during therapy. In this study we investigated the mechanisms of radiosensitization for DNA-PK and PARP inhibitors by examining the impacts on proliferating vs quiescent cell populations. Experiments were performed in BRCA1/2null and wild-type parental cancer models in vitro and in vivo. Overall AZD7648 has greater radiosensitizing activity relative to Olaparib, with BRCA2-deficient models showing the greatest sensitivity. However, DNA-PK inhibitor AZD7648 also produced greater toxicity in all irradiated mice. While both DNA-PK and PARP inhibition sensitizes wild type tumor cells to radiation, in BRCA1/2 deficient cells PARP inhibition by Olaparib had limited radiosensitization capacity. Quiescent cells are more radioresistant than proliferating cells, and these were also effectively sensitized by AZD7648 while Olaparib was unable to increase radiation-induced cell kill, even in BRCA1/2null cells. These findings underscore the distinct mechanisms of radiosensitization for DNA-PK and PARP inhibitors. While DNA-PK inhibitors are able to target both proliferating and non-proliferating tumor cells for greater overall anti-cancer benefit, their application is limited by exacerbation of normal tissue toxicities. Conversely, PARP inhibitors exhibit selective activity for proliferating cells, providing a mechanism for targeting activity to cancers, but due to poor activity in non-proliferating cells they have an overall reduced impact on tumor growth control. This study highlights the importance of creating a therapeutic ratio with DNA damage repair inhibition radiation sensitizing strategies.
    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
    尽管DNA-PK抑制剂(DNA-PK-i)已用于癌症治疗的临床试验,DNA-PK-i在肿瘤细胞抑制中的生物标志物和作用机制尚不清楚.这里,我们观察到,低剂量的DNA-PK-i和PARP抑制剂(PARP-i)可合成抑制BRCA缺陷型肿瘤细胞,而不诱导DNA双链断裂(DSB).相反,我们发现一部分DNA-PK位于核仁内部,我们没有观察到明显的DSB。此外,Ku蛋白识别pre-rRNA,其促进DNA-PKcs自磷酸化而不依赖于DNA损伤。核糖体蛋白也被DNA-PK磷酸化,调节前rRNA生物发生。此外,DNA-PK-i与PARP-i一起作用以抑制前rRNA生物发生和肿瘤细胞生长。总的来说,我们的研究揭示了DNA-PK-i在肿瘤抑制中的DNA损伤修复非依赖性作用.
    Although DNA-PK inhibitors (DNA-PK-i) have been applied in clinical trials for cancer treatment, the biomarkers and mechanism of action of DNA-PK-i in tumor cell suppression remain unclear. Here, we observed that a low dose of DNA-PK-i and PARP inhibitor (PARP-i) synthetically suppresses BRCA-deficient tumor cells without inducing DNA double-strand breaks (DSBs). Instead, we found that a fraction of DNA-PK localized inside of nucleoli, where we did not observe obvious DSBs. Moreover, the Ku proteins recognize pre-rRNA that facilitates DNA-PKcs autophosphorylation independent of DNA damage. Ribosomal proteins are also phosphorylated by DNA-PK, which regulates pre-rRNA biogenesis. In addition, DNA-PK-i acts together with PARP-i to suppress pre-rRNA biogenesis and tumor cell growth. Collectively, our studies reveal a DNA damage repair-independent role of DNA-PK-i in tumor suppression.
    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
    作为激活STING-TBK1信号级联的主要DNA传感器,cGAS主要存在于细胞质中。最近的许多报道表明,cGAS在细胞核中也起着关键作用。我们以前的工作首次证明了cGAS在DNA损伤发生后易位到细胞核并抑制同源重组(HR),DNA双链断裂(DSB)修复的两条主要途径之一。然而,核cGAS是否调节另一个DSB修复途径,非同源末端连接(NHEJ),可以进一步分为不易出错的规范NHEJ(c-NHEJ)和更诱变的替代NHEJ(alt-NHEJ)亚途径,没有被定性。这里,我们证明cGAS使两个NHEJ亚途径的平衡向c-NHEJ倾斜。机械上,cGAS-Ku80复合物增强了DNA-PKcs和去泛素酶USP7之间的相互作用,以提高DNA-PKcs蛋白的稳定性,从而促进c-NHEJ。相比之下,cGAS-Ku80复合物通过直接结合Polθ的启动子来抑制alt-NHEJ,从而抑制其转录。一起,这些发现揭示了核cGAS在调节DSB修复中的新功能,表明cGAS在细胞核中的存在对维持基因组完整性也很重要。
    As the major DNA sensor that activates the STING-TBK1 signaling cascade, cGAS is mainly present in the cytosol. A number of recent reports have indicated that cGAS also plays critical roles in the nucleus. Our previous work demonstrated for the first time that cGAS is translocated to the nucleus upon the occurrence of DNA damage and inhibits homologous recombination (HR), one of the two major pathways of DNA double strand break (DSB) repair. However, whether nuclear cGAS regulates the other DSB repair pathway, nonhomologous end joining (NHEJ), which can be further divided into the less error-prone canonical NHEJ (c-NHEJ) and more mutagenic alternative NHEJ (alt-NHEJ) subpathways, has not been characterized. Here, we demonstrated that cGAS tipped the balance of the two NHEJ subpathways toward c-NHEJ. Mechanistically, the cGAS-Ku80 complex enhanced the interaction between DNA-PKcs and the deubiquitinase USP7 to improve DNA-PKcs protein stability, thereby promoting c-NHEJ. In contrast, the cGAS-Ku80 complex suppressed alt-NHEJ by directly binding to the promoter of Polθ to suppress its transcription. Together, these findings reveal a novel function of nuclear cGAS in regulating DSB repair, suggesting that the presence of cGAS in the nucleus is also important in the maintenance of genome integrity.
    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
    卵巢癌是妇科癌症死亡的主要原因。在最具创新性的抗癌方法中,合成致死性的遗传概念是多基因的突变协同作用以影响细胞死亡。先前的研究发现,尽管牛痘相关激酶1(VRK1)与DNA损伤修复蛋白相关,其潜在机制仍不清楚。这里,我们发现VRK1在卵巢肿瘤中高表达,VRK1耗竭能显著促进细胞凋亡和细胞周期阻滞。VRK1敲低对细胞凋亡的影响表现为DNA损伤增加,基因组不稳定性,和细胞凋亡,并通过使DNA-PK不稳定来阻断非同源末端连接(NHEJ)。Further,我们验证了VRK1耗竭增强了对PARP抑制剂(PARPi)的敏感性,奥拉帕利,通过DNA损伤促进细胞凋亡,特别是在VRK1高表达的卵巢癌细胞系中。与DNA损伤反应有关的蛋白质是开发新的抗癌治疗策略的合适靶标。它们的组合可以代表合成杀伤力的另一种形式。因此,通过将奥拉帕尼与VRK1的消除相结合,可以损害正常的保护性DNA损伤反应,并且可以用于减少药物剂量及其相关毒性。总之,VRK1代表PARPi敏感性的潜在生物标志物,和一个新的DDR相关的治疗靶点,卵巢癌。
    Ovarian cancer is the leading cause of gynecologic cancer death. Among the most innovative anti-cancer approaches, the genetic concept of synthetic lethality is that mutations in multiple genes work synergistically to effect cell death. Previous studies found that although vaccinia-related kinase-1 (VRK1) associates with DNA damage repair proteins, its underlying mechanisms remain unclear. Here, we found high VRK1 expression in ovarian tumors, and that VRK1 depletion can significantly promote apoptosis and cell cycle arrest. The effect of VRK1 knockdown on apoptosis was manifested by increased DNA damage, genomic instability, and apoptosis, and also blocked non-homologous end joining (NHEJ) by destabilizing DNA-PK. Further, we verified that VRK1 depletion enhanced sensitivity to a PARP inhibitor (PARPi), olaparib, promoting apoptosis through DNA damage, especially in ovarian cancer cell lines with high VRK1 expression. Proteins implicated in DNA damage responses are suitable targets for the development of new anti-cancer therapeutic strategies, and their combination could represent an alternative form of synthetic lethality. Therefore, normal protective DNA damage responses are impaired by combining olaparib with elimination of VRK1 and could be used to reduce drug dose and its associated toxicity. In summary, VRK1 represents both a potential biomarker for PARPi sensitivity, and a new DDR-associated therapeutic target, in ovarian cancer.
    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