DNA-Activated Protein Kinase

DNA 活化蛋白激酶
  • 文章类型: Journal Article
    关键的DNA修复酶DNA-PKcs具有几个重要的细胞功能。小鼠DNA-PKcs活性的丧失揭示了在免疫和神经系统中的重要作用。在人类中,DNA-PKcs是大脑发育和功能的关键因素,因为prkdc基因的突变会导致严重的神经功能缺损,如小头畸形和癫痫发作。预测DNA-PKcs在神经元中的未知作用。在这里,我们表明DNA-PKcs调节突触可塑性。我们证明DNA-PKcs定位于突触并在新鉴定的控制PSD-95蛋白稳定性的残基处磷酸化PSD-95。DNA-PKcs-/-小鼠的特征是长期增强(LTP)受损,神经元形态的变化,和降低突触后蛋白的水平。当在DNA-PKcs-/-小鼠中过表达时,组成型磷酸化的PSD-95突变体挽救LTP损伤。我们的研究确定了DNA-PKcs在调节神经元可塑性方面的新兴生理功能,超越基因组稳定性。
    The key DNA repair enzyme DNA-PKcs has several and important cellular functions. Loss of DNA-PKcs activity in mice has revealed essential roles in immune and nervous systems. In humans, DNA-PKcs is a critical factor for brain development and function since mutation of the prkdc gene causes severe neurological deficits such as microcephaly and seizures, predicting yet unknown roles of DNA-PKcs in neurons. Here we show that DNA-PKcs modulates synaptic plasticity. We demonstrate that DNA-PKcs localizes at synapses and phosphorylates PSD-95 at newly identified residues controlling PSD-95 protein stability. DNA-PKcs -/- mice are characterized by impaired Long-Term Potentiation (LTP), changes in neuronal morphology, and reduced levels of postsynaptic proteins. A PSD-95 mutant that is constitutively phosphorylated rescues LTP impairment when over-expressed in DNA-PKcs -/- mice. Our study identifies an emergent physiological function of DNA-PKcs in regulating neuronal plasticity, beyond genome stability.
    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
    DNA-PKcs是参与DNA修复和反应途径的关键蛋白质靶标,其异常活性与各种癌症的发生和进展密切相关。在这项研究中,我们采用了基于深度学习的筛选和基于分子动力学(MD)模拟的管道,确定八个候选DNA-PKcs目标。随后的实验揭示了三种小分子(5025-0002、M769-1095和V008-1080)对DNA-PKcs介导的细胞增殖的有效抑制。这些分子表现出抗癌活性,IC50(抑制浓度为50%)值为152.6μM,30.71μM,和74.84μM,分别。值得注意的是,V008-1080增强由CRISPR/Cas9介导的同源定向修复(HDR),同时抑制非同源末端连接(NHEJ)效率。对结构-活性关系的进一步研究揭示了这些小分子与DNA-PKcs之间的结合位点和关键相互作用。这是DeepBindGCN_RG在实际药物筛选任务中的初次运用,一种新型DNA-PKcs抑制剂的成功发现证明了其作为筛选流程中核心成分的有效性。此外,这项研究为探索新的抗癌疗法和通过靶向DNA-PKcs推进基因编辑技术的发展提供了重要的见解。
    DNA-PKcs is a crucial protein target involved in DNA repair and response pathways, with its abnormal activity closely associated with the occurrence and progression of various cancers. In this study, we employed a deep learning-based screening and molecular dynamics (MD) simulation-based pipeline, identifying eight candidates for DNA-PKcs targets. Subsequent experiments revealed the effective inhibition of DNA-PKcs-mediated cell proliferation by three small molecules (5025-0002, M769-1095, and V008-1080). These molecules exhibited anticancer activity with IC50 (inhibitory concentration at 50%) values of 152.6 μM, 30.71 μM, and 74.84 μM, respectively. Notably, V008-1080 enhanced homology-directed repair (HDR) mediated by CRISPR/Cas9 while inhibiting non-homologous end joining (NHEJ) efficiency. Further investigations into the structure-activity relationships unveiled the binding sites and critical interactions between these small molecules and DNA-PKcs. This is the first application of DeepBindGCN_RG in a real drug screening task, and the successful discovery of a novel DNA-PKcs inhibitor demonstrates its efficiency as a core component in the screening pipeline. Moreover, this study provides important insights for exploring novel anticancer therapeutics and advancing the development of gene editing techniques by targeting DNA-PKcs.
    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
    非同源末端连接在修复辐射诱导的双链断裂中的主导作用将DNA依赖性蛋白激酶(DNA-PK)确定为开发放射增敏剂的出色靶标。我们报道了一类新的咪唑并[4,5-c]吡啶-2-酮DNA-PK抑制剂的发现。与相关的磷酸肌醇3激酶(PI3K)和PI3K样激酶(PIKK)家族和更广泛的激酶相比,结构活性研究最终确定了78作为nMDNA-PK抑制剂,对DNA-PK具有优异的选择性。并显示HAP1细胞的DNA-PK依赖性放射增敏作用。化合物78在体外证明了对多种癌细胞的强大放射增敏作用。显示出高口服生物利用度,和致敏的结直肠癌(HCT116/54C)和头颈部鳞状细胞癌(UT-SCC-74B)肿瘤异种移植物对放射。化合物78还与放射组合提供了对HCT116/54C肿瘤异种移植物的显著肿瘤生长抑制。化合物78代表一种新的,强力,和选择性的DNA-PK抑制剂类,具有作为癌症治疗的放射增敏剂的巨大潜力。
    The dominant role of non-homologous end-joining in the repair of radiation-induced double-strand breaks identifies DNA-dependent protein kinase (DNA-PK) as an excellent target for the development of radiosensitizers. We report the discovery of a new class of imidazo[4,5-c]pyridine-2-one DNA-PK inhibitors. Structure-activity studies culminated in the identification of 78 as a nM DNA-PK inhibitor with excellent selectivity for DNA-PK compared to related phosphoinositide 3-kinase (PI3K) and PI3K-like kinase (PIKK) families and the broader kinome, and displayed DNA-PK-dependent radiosensitization of HAP1 cells. Compound 78 demonstrated robust radiosensitization of a broad range of cancer cells in vitro, displayed high oral bioavailability, and sensitized colorectal carcinoma (HCT116/54C) and head and neck squamous cell carcinoma (UT-SCC-74B) tumor xenografts to radiation. Compound 78 also provided substantial tumor growth inhibition of HCT116/54C tumor xenografts in combination with radiation. Compound 78 represents a new, potent, and selective class of DNA-PK inhibitors with significant potential as radiosensitizers for cancer treatment.
    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
    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
    部署DNA损伤反应(DDR)对抗各种形式的DNA损伤,确保基因组稳定性。癌细胞对基因组不稳定性的倾向提供了通过抑制DDR途径选择性杀死癌细胞的治疗机会。DNA依赖性蛋白激酶(DNA-PK),核丝氨酸/苏氨酸激酶,对于DNA双链断裂(DSB)修复中的非同源末端连接(NHEJ)途径至关重要。因此,靶向DNA-PK是一种有前途的癌症治疗策略.本文综述了DNA-PK及其相关大蛋白的结构,以及DNA-PK抑制剂的发展过程,以及其临床应用的最新进展。我们强调我们对基于不同支架的DNA-PK抑制剂的开发过程和结构-活性关系(SARs)的分析。我们希望这篇综述将为未来寻求开发新的DNA-PK抑制剂的研究人员提供实用信息。
    The deployment of DNA damage response (DDR) combats various forms of DNA damage, ensuring genomic stability. Cancer cells\' propensity for genomic instability offers therapeutic opportunities to selectively kill cancer cells by suppressing the DDR pathway. DNA-dependent protein kinase (DNA-PK), a nuclear serine/threonine kinase, is crucial for the non-homologous end joining (NHEJ) pathway in the repair of DNA double-strand breaks (DSBs). Therefore, targeting DNA-PK is a promising cancer treatment strategy. This review elaborates on the structures of DNA-PK and its related large protein, as well as the development process of DNA-PK inhibitors, and recent advancements in their clinical application. We emphasize our analysis of the development process and structure-activity relationships (SARs) of DNA-PK inhibitors based on different scaffolds. We hope this review will provide practical information for researchers seeking to develop novel DNA-PK inhibitors in the future.
    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
    KRAS突变癌症,由于它们的蛋白质靶向复杂性,目前显著的治疗障碍。鉴定这些癌症中的巨噬细胞表型已成为有希望的替代治疗靶标。我们的研究介绍了MPD1,一种针对巨噬细胞吞噬的靶向肽-药物偶联物(PDC),用于治疗KRAS突变癌症。该PDC被特别设计为通过其半胱天冬酶-3可切割特性触发正反馈回路。然而,我们观察到,在癌细胞中,DNA-PK介导的DNA损伤修复过程阻碍了该环。为了克服这个障碍,我们使用AZD7648,一种DNA-PK抑制剂。有趣的是,在KRAS突变异种移植模型中,MPD1和AZD7648联合治疗的完全缓解率达到100%.我们专注于它的协同机制。我们发现AZD7648在KRAS突变的癌细胞中特异性增强巨成细胞作用。进一步的分析揭示了巨噬细胞增多和PI3K信号之间的显著相关性,由AMPK途径驱动。此外,AZD7648加强了正反馈回路,导致升级的细胞凋亡和增强的有效载荷在肿瘤内的积累。AZD7648在增强纳米大小的药物递送和防止DNA修复抗性方面具有广泛的应用。有希望的功效和明显的协同作用强调了将MPD1与AZD7648组合作为治疗KRAS突变癌症的策略的潜力。
    KRAS-mutant cancers, due to their protein targeting complexity, present significant therapeutic hurdles. The identification of the macropinocytic phenotype in these cancers has emerged as a promising alternative therapeutic target. Our study introduces MPD1, an macropinocytosis-targeting peptide-drug conjugates (PDC), which is developed to treat KRAS mutant cancers. This PDC is specifically designed to trigger a positive feedback loop through its caspase-3 cleavable characteristic. However, we observe that this loop is hindered by DNA-PK mediated DNA damage repair processes in cancer cells. To counter this impediment, we employ AZD7648, a DNA-PK inhibitor. Interestingly, the combined treatment of MPD1 and AZD7648 resulted in a 100% complete response rate in KRAS-mutant xenograft model. We focus on the synergic mechanism of it. We discover that AZD7648 specifically enhances macropinocytosis in KRAS-mutant cancer cells. Further analysis uncovers a significant correlation between the increase in macropinocytosis and PI3K signaling, driven by AMPK pathways. Also, AZD7648 reinforces the positive feedback loop, leading to escalated apoptosis and enhanced payload accumulation within tumors. AZD7648 possesses broad applications in augmenting nano-sized drug delivery and preventing DNA repair resistance. The promising efficacy and evident synergy underscore the potential of combining MPD1 with AZD7648 as a strategy for treating KRAS-mutant cancers.
    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:靶向DNA损伤修复因子,如DNA依赖性蛋白激酶催化亚基(DNA-PKcs),可能提供有效治疗多发性骨髓瘤(MM)的机会。结合DNA损伤诱导剂,该策略已被证明可以通过升高的细胞溶质DNA水平激活cGAS-STING途径来部分改善化疗。然而,由于cGAS主要被细胞核中的染色质隔离,目前尚不清楚cGAS如何在DNA损伤后从染色质中释放并易位到细胞质中,导致cGAS-STING激活。
    方法:我们通过进行质谱和机制研究,检查了DNA-PKcs抑制对cGAS-STING介导的MM化学敏感性的作用。
    结果:这里,我们发现DNA-PKcs抑制通过激活cGAS-STING信号增强DNA损伤诱导剂阿霉素诱导的抗MM作用。MM细胞中的cGAS-STING激活部分通过IRF3-NOXA-BAK轴引起细胞死亡,并诱导巨噬细胞的M1极化。此外,这种激活不是由经典的非同源末端连接(c-NHEJ)缺陷引起的。相反,多柔比星诱导的DNA损伤,抑制DNA-PKcs促进cGAS从细胞质染色质片段释放,并增加细胞溶质cGAS和DNA的量,激活cGAS-STING。
    结论:抑制DNA-PKcs可以通过去螯合cGAS来改善多柔比星治疗MM的疗效。
    BACKGROUND: Targeting DNA damage repair factors, such as DNA-dependent protein kinase catalytic subunit (DNA-PKcs), may offer an opportunity for effective treatment of multiple myeloma (MM). In combination with DNA damage-inducing agents, this strategy has been shown to improve chemotherapies partially via activation of cGAS-STING pathway by an elevated level of cytosolic DNA. However, as cGAS is primarily sequestered by chromatin in the nucleus, it remains unclear how cGAS is released from chromatin and translocated into the cytoplasm upon DNA damage, leading to cGAS-STING activation.
    METHODS: We examined the role of DNA-PKcs inhibition on cGAS-STING-mediated MM chemosensitivity by performing mass spectrometry and mechanism study.
    RESULTS: Here, we found DNA-PKcs inhibition potentiated DNA damage-inducing agent doxorubicin-induced anti-MM effect by activating cGAS-STING signaling. The cGAS-STING activation in MM cells caused cell death partly via IRF3-NOXA-BAK axis and induced M1 polarization of macrophages. Moreover, this activation was not caused by defective classical non-homologous end joining (c-NHEJ). Instead, upon DNA damage induced by doxorubicin, inhibition of DNA-PKcs promoted cGAS release from cytoplasmic chromatin fragments and increased the amount of cytosolic cGAS and DNA, activating cGAS-STING.
    CONCLUSIONS: Inhibition of DNA-PKcs could improve the efficacy of doxorubicin in treatment of MM by de-sequestrating cGAS in damaged chromatin.
    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
    驱动神经胶质瘤进展的因素仍然知之甚少。这里,表观遗传调节因子TRIM24被确定为神经胶质瘤进展的驱动因素,其中TRIM24过表达促进HRasV12间变性星形细胞瘤(AA)进展为上皮样GBM(Ep-GBM)样肿瘤。TRIM24与HRasV12的共转染还诱导具有肿瘤蛋白p53基因(TP53)敲低的人神经干细胞(hNSC)的Ep-GBM样转化。此外,TRIM24在临床Ep-GBM标本中高表达。使用单细胞RNA测序(scRNA-Seq),作者表明TRIM24过表达影响肿瘤内异质性和肿瘤微环境.机械上,HRasV12激活RNA输出(PHAX)的磷酸化衔接子,并上调U3小核仁RNA(U3snoRNA)以招募Ku依赖性DNA依赖性蛋白激酶催化亚基(DNA-PKcs)。过表达的TRIM24也被PHAX招募到U3snoRNA,从而促进TRIM24在S767/768残基处的DNA-PKcs磷酸化。磷酸化TRIM24诱导表观基因组和转录因子网络重编程并促进Ep-GBM样转化。用小分子抑制剂NU7441靶向DNA-PKcs与替莫唑胺协同作用以降低Ep-GBM致瘤性并延长动物存活。这些发现为Ep-GBM样转化的表观遗传调控提供了新的见解,并为Ep-GBM患者提出了潜在的治疗策略。
    The factors driving glioma progression remain poorly understood. Here, the epigenetic regulator TRIM24 is identified as a driver of glioma progression, where TRIM24 overexpression promotes HRasV12 anaplastic astrocytoma (AA) progression into epithelioid GBM (Ep-GBM)-like tumors. Co-transfection of TRIM24 with HRasV12 also induces Ep-GBM-like transformation of human neural stem cells (hNSCs) with tumor protein p53 gene (TP53) knockdown. Furthermore, TRIM24 is highly expressed in clinical Ep-GBM specimens. Using single-cell RNA-sequencing (scRNA-Seq), the authors show that TRIM24 overexpression impacts both intratumoral heterogeneity and the tumor microenvironment. Mechanically, HRasV12 activates phosphorylated adaptor for RNA export (PHAX) and upregulates U3 small nucleolar RNAs (U3 snoRNAs) to recruit Ku-dependent DNA-dependent protein kinase catalytic subunit (DNA-PKcs). Overexpressed TRIM24 is also recruited by PHAX to U3 snoRNAs, thereby facilitating DNA-PKcs phosphorylation of TRIM24 at S767/768 residues. Phosphorylated TRIM24 induces epigenome and transcription factor network reprogramming and promotes Ep-GBM-like transformation. Targeting DNA-PKcs with the small molecule inhibitor NU7441 synergizes with temozolomide to reduce Ep-GBM tumorigenicity and prolong animal survival. These findings provide new insights into the epigenetic regulation of Ep-GBM-like transformation and suggest a potential therapeutic strategy for patients with Ep-GBM.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号