首页 > 最新文献

Oncogene最新文献

英文 中文
Identification of Raptor and GLI1 as USP37 substrates highlight its context-specific function in medulloblastoma cells. Raptor和GLI1作为USP37底物的鉴定突出了其在成神经管细胞瘤细胞中的上下文特异性功能。
IF 7.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-18 DOI: 10.1038/s41388-025-03651-2
Ashutosh Singh, Donghang Cheng, Amanda R Haltom, Yanwen Yang, Tara Dobson, Rashieda Hatcher, Veena Rajaram, Vidya Gopalakrishnan

The USP37 gene encodes a deubiquitylase (DUB), which catalyzes the proteolytic removal of ubiquitin moieties from proteins to modulate their stability, cellular localization or activity. Its expression is downregulated in a subgroup of medulloblastomas driven by constitutive activation of sonic hedgehog (SHH) signaling. Patients with SHH-driven medulloblastomas with elevated expression of the RE1 silencing transcription factor (REST) and reduced expression of USP37 have poor outcomes. In previous studies, we showed sustained proliferation of SHH-medulloblastoma cells due to blockade of terminal cell cycle exit and neuronal differentiation stemming from a failure in USP37-dependent stabilization of its target, the cyclin-dependent kinase inhibitor (CDKI)-p27. This finding suggested a tumor suppressive function for USP37. Interestingly, the current study also uncovered Raptor, a component of the mTORC1 complex, as a novel target of USP37. Under conditions of low-USP37 expression, reduced Raptor stability and mTORC1 activity caused a decline in phosphorylation of 4E-binding protein 1 (4EBP1) and increased its interaction with eukaryotic elongation factor 4E (eIF4E), which is known to inhibit CAP-dependent translation initiation. Surprisingly, a subset of patients with SHH-driven medulloblastomas with elevated expression of USP37 and the Glioma-associated Oncogene 1 (GLI1), also exhibited poor outcomes. Using genetic and biochemical analyses, we showed that USP37-mediated stabilization of GLI1, a terminal effector of SHH signaling, increases pathway activity and upregulates expression of its target oncogene product, CCND1, to drive cell proliferation. These data indicate that USP37 elevation in SHH-driven medulloblastomas has the potential to promote non-canonical activation of SHH signaling. Overall, our findings suggest that USP37 may have context-specific oncogenic and tumor suppressive roles in medulloblastoma cells.

USP37基因编码一种去泛素化酶(DUB),该酶催化蛋白质水解去除泛素部分,从而调节蛋白质的稳定性、细胞定位或活性。在成神经管细胞瘤的一个亚组中,它的表达被sonic hedgehog (SHH)信号的组成性激活所下调。伴有RE1沉默转录因子(REST)表达升高和USP37表达降低的shh驱动型髓母细胞瘤患者预后较差。在之前的研究中,我们发现shh -髓母细胞瘤细胞的持续增殖是由于usp37依赖的靶标细胞周期蛋白依赖性激酶抑制剂(CDKI)-p27的稳定失败而导致细胞周期末退出和神经元分化受阻。这一发现提示USP37具有肿瘤抑制功能。有趣的是,目前的研究还发现了Raptor, mTORC1复合物的一个组成部分,作为USP37的新靶点。在usp37低表达的条件下,Raptor稳定性和mTORC1活性降低导致4E结合蛋白1 (4EBP1)磷酸化下降,并增加其与真核延伸因子4E (eIF4E)的相互作用,已知其抑制cap依赖的翻译起始。令人惊讶的是,一部分shh驱动的髓母细胞瘤患者USP37和胶质瘤相关癌基因1 (GLI1)表达升高,也表现出较差的预后。通过遗传和生化分析,我们发现usp37介导的GLI1 (SHH信号的末端效应物)的稳定可以增加通路活性并上调其靶癌基因产物CCND1的表达,从而驱动细胞增殖。这些数据表明,在SHH驱动的髓母细胞瘤中,USP37的升高有可能促进SHH信号的非典型激活。总之,我们的研究结果表明USP37可能在成神经管细胞瘤细胞中具有上下文特异性的致癌和肿瘤抑制作用。
{"title":"Identification of Raptor and GLI1 as USP37 substrates highlight its context-specific function in medulloblastoma cells.","authors":"Ashutosh Singh, Donghang Cheng, Amanda R Haltom, Yanwen Yang, Tara Dobson, Rashieda Hatcher, Veena Rajaram, Vidya Gopalakrishnan","doi":"10.1038/s41388-025-03651-2","DOIUrl":"https://doi.org/10.1038/s41388-025-03651-2","url":null,"abstract":"<p><p>The USP37 gene encodes a deubiquitylase (DUB), which catalyzes the proteolytic removal of ubiquitin moieties from proteins to modulate their stability, cellular localization or activity. Its expression is downregulated in a subgroup of medulloblastomas driven by constitutive activation of sonic hedgehog (SHH) signaling. Patients with SHH-driven medulloblastomas with elevated expression of the RE1 silencing transcription factor (REST) and reduced expression of USP37 have poor outcomes. In previous studies, we showed sustained proliferation of SHH-medulloblastoma cells due to blockade of terminal cell cycle exit and neuronal differentiation stemming from a failure in USP37-dependent stabilization of its target, the cyclin-dependent kinase inhibitor (CDKI)-p27. This finding suggested a tumor suppressive function for USP37. Interestingly, the current study also uncovered Raptor, a component of the mTORC1 complex, as a novel target of USP37. Under conditions of low-USP37 expression, reduced Raptor stability and mTORC1 activity caused a decline in phosphorylation of 4E-binding protein 1 (4EBP1) and increased its interaction with eukaryotic elongation factor 4E (eIF4E), which is known to inhibit CAP-dependent translation initiation. Surprisingly, a subset of patients with SHH-driven medulloblastomas with elevated expression of USP37 and the Glioma-associated Oncogene 1 (GLI1), also exhibited poor outcomes. Using genetic and biochemical analyses, we showed that USP37-mediated stabilization of GLI1, a terminal effector of SHH signaling, increases pathway activity and upregulates expression of its target oncogene product, CCND1, to drive cell proliferation. These data indicate that USP37 elevation in SHH-driven medulloblastomas has the potential to promote non-canonical activation of SHH signaling. Overall, our findings suggest that USP37 may have context-specific oncogenic and tumor suppressive roles in medulloblastoma cells.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145775259","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
IRF2BPL transcriptionally regulates IGFBP2 to promote tumor progression and suppresses immune cell infiltration in esophageal squamous cell carcinoma. IRF2BPL通过转录调控IGFBP2促进食管鳞状细胞癌的肿瘤进展,抑制免疫细胞浸润。
IF 7.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-18 DOI: 10.1038/s41388-025-03658-9
Yueguang Wu, Heyang Cui, Longlong Wang, Ning Ding, Yongjia Weng, Yikun Cheng, Shanshan Bi, Heng Xiao, Mingwei Gao, Huijuan Liu, Qiqin Song, Weimin Zhang, Yongping Cui

Numerous ubiquitination-related proteases (URPs) have been identified as facilitators of disease progression through the disruption of ubiquitination homeostasis in substrate proteins. Notably, some URPs have exhibited non-classical biological functions. In this study, we experimentally elucidate the role of the E3 ubiquitin ligase IRF2BPL as transcriptional activator that promotes malignant phenotypes in esophageal squamous cell carcinoma (ESCC) and inhibits the infiltration of various immune cells within the tumor microenvironment. Specifically, we found that IRF2BPL is highly expressed in ESCC cells and promotes IGFBP2 transcription, thereby facilitating ESCC development both in vivo and in vitro. Moreover, the chemical drug ONC201 was shown to effectively impede ESCC progression induced by the hyperactive IRF2BPL-IGFBP2 axis in tumor cells. Collectively, our findings verified that the IRF2BPL-IGFBP2 axis plays a critical role in enhancing ESCC progression by increasing the malignancy of ESCC cells and fostering an immune-deficient tumor microenvironment. Targeting the IRF2BPL-IGFBP2 axis may represent a promising therapeutic strategy for ESCC.

许多泛素化相关蛋白酶(URPs)已被确定为通过破坏底物蛋白的泛素化稳态来促进疾病进展的促进者。值得注意的是,一些URPs表现出非经典的生物学功能。在本研究中,我们通过实验阐明了E3泛素连接酶IRF2BPL作为转录激活因子在食管鳞状细胞癌(ESCC)中促进恶性表型的作用,并抑制肿瘤微环境中各种免疫细胞的浸润。具体来说,我们发现IRF2BPL在ESCC细胞中高表达,并促进IGFBP2的转录,从而促进ESCC在体内和体外的发展。此外,化学药物ONC201被证明可以有效阻止肿瘤细胞中由过度活跃的IRF2BPL-IGFBP2轴诱导的ESCC进展。总之,我们的研究结果证实,IRF2BPL-IGFBP2轴通过增加ESCC细胞的恶性程度和培养免疫缺陷的肿瘤微环境,在促进ESCC进展中起着关键作用。靶向IRF2BPL-IGFBP2轴可能是ESCC的一种有前景的治疗策略。
{"title":"IRF2BPL transcriptionally regulates IGFBP2 to promote tumor progression and suppresses immune cell infiltration in esophageal squamous cell carcinoma.","authors":"Yueguang Wu, Heyang Cui, Longlong Wang, Ning Ding, Yongjia Weng, Yikun Cheng, Shanshan Bi, Heng Xiao, Mingwei Gao, Huijuan Liu, Qiqin Song, Weimin Zhang, Yongping Cui","doi":"10.1038/s41388-025-03658-9","DOIUrl":"https://doi.org/10.1038/s41388-025-03658-9","url":null,"abstract":"<p><p>Numerous ubiquitination-related proteases (URPs) have been identified as facilitators of disease progression through the disruption of ubiquitination homeostasis in substrate proteins. Notably, some URPs have exhibited non-classical biological functions. In this study, we experimentally elucidate the role of the E3 ubiquitin ligase IRF2BPL as transcriptional activator that promotes malignant phenotypes in esophageal squamous cell carcinoma (ESCC) and inhibits the infiltration of various immune cells within the tumor microenvironment. Specifically, we found that IRF2BPL is highly expressed in ESCC cells and promotes IGFBP2 transcription, thereby facilitating ESCC development both in vivo and in vitro. Moreover, the chemical drug ONC201 was shown to effectively impede ESCC progression induced by the hyperactive IRF2BPL-IGFBP2 axis in tumor cells. Collectively, our findings verified that the IRF2BPL-IGFBP2 axis plays a critical role in enhancing ESCC progression by increasing the malignancy of ESCC cells and fostering an immune-deficient tumor microenvironment. Targeting the IRF2BPL-IGFBP2 axis may represent a promising therapeutic strategy for ESCC.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145781331","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Dual-target CAR-T therapy for ovarian cancer: synergistic targeting of MSLN and B7H3 enhances anti-tumor efficacy and overcomes antigen heterogeneity. 卵巢癌双靶点CAR-T治疗:MSLN和B7H3协同靶向增强抗肿瘤疗效,克服抗原异质性
IF 7.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-17 DOI: 10.1038/s41388-025-03663-y
Feng Ji, Kai Yan, Bo Ding, Yuxin Zhu, Mengchen Rao, Kexing Gao, Hao Lin, Yiyang Shan, Sicong Liu, Zhongdang Xiao, Yang Shen

Ovarian cancer (OC) remains a lethal malignancy with limited treatment options owing to antigen heterogeneity and an immunosuppressive tumor microenvironment (TME). Here, we designed a unique chimeric Antigen Receptor T-Cell (CAR-T) construct (B4M3) that integrates an anti-MSLN scFv linked to the CD3ζ activation domain and an anti-B7H3 scFv linked to the 4-1BB co-stimulatory domain. In vitro, B4M3 CAR-T cells exhibited robust cytotoxicity against OC cell lines with enhanced degranulation (CD107a) and efficient tumor cell killing, even at low effector-to-target ratios. In vivo, B4M3 CAR-T cells significantly inhibited tumor growth and prolonged survival and demonstrated superior tumor infiltration and persistence in OC xenograft models. Imaging mass cytometry (IMC) revealed that B4M3 treatment reshaped the TME, increased cytotoxic T lymphocyte (CTL) infiltration, and reduced regulatory T cells (Tregs). Mechanistically, B4M3 therapy upregulated TGF-β, promoting Th17 differentiation and CTL recruitment, thereby enhancing anti-tumor immunity. Our findings demonstrate that B4M3 CAR-T cells effectively address antigen heterogeneity and enhance therapeutic efficacy in OC, thereby offering a promising strategy for solid tumor immunotherapy.

卵巢癌(OC)仍然是一种致命的恶性肿瘤,由于抗原异质性和免疫抑制肿瘤微环境(TME),治疗选择有限。在这里,我们设计了一种独特的嵌合抗原受体t细胞(CAR-T)结构体(B4M3),该结构体整合了连接CD3ζ激活域的抗msln scFv和连接4-1BB共刺激域的抗b7h3 scFv。在体外,B4M3 CAR-T细胞对OC细胞系表现出强大的细胞毒性,具有增强的脱颗粒(CD107a)和有效的肿瘤细胞杀伤,即使在低效靶比下也是如此。在体内,B4M3 CAR-T细胞显著抑制肿瘤生长和延长存活时间,并在OC异种移植模型中表现出优越的肿瘤浸润和持久性。成像细胞计数(IMC)显示B4M3治疗可重塑TME,增加细胞毒性T淋巴细胞(CTL)浸润,减少调节性T细胞(Tregs)。机制上,B4M3治疗上调TGF-β,促进Th17分化和CTL募集,从而增强抗肿瘤免疫。我们的研究结果表明,B4M3 CAR-T细胞有效地解决了OC的抗原异质性并提高了治疗效果,从而为实体瘤免疫治疗提供了一种有希望的策略。
{"title":"Dual-target CAR-T therapy for ovarian cancer: synergistic targeting of MSLN and B7H3 enhances anti-tumor efficacy and overcomes antigen heterogeneity.","authors":"Feng Ji, Kai Yan, Bo Ding, Yuxin Zhu, Mengchen Rao, Kexing Gao, Hao Lin, Yiyang Shan, Sicong Liu, Zhongdang Xiao, Yang Shen","doi":"10.1038/s41388-025-03663-y","DOIUrl":"https://doi.org/10.1038/s41388-025-03663-y","url":null,"abstract":"<p><p>Ovarian cancer (OC) remains a lethal malignancy with limited treatment options owing to antigen heterogeneity and an immunosuppressive tumor microenvironment (TME). Here, we designed a unique chimeric Antigen Receptor T-Cell (CAR-T) construct (B4M3) that integrates an anti-MSLN scFv linked to the CD3ζ activation domain and an anti-B7H3 scFv linked to the 4-1BB co-stimulatory domain. In vitro, B4M3 CAR-T cells exhibited robust cytotoxicity against OC cell lines with enhanced degranulation (CD107a) and efficient tumor cell killing, even at low effector-to-target ratios. In vivo, B4M3 CAR-T cells significantly inhibited tumor growth and prolonged survival and demonstrated superior tumor infiltration and persistence in OC xenograft models. Imaging mass cytometry (IMC) revealed that B4M3 treatment reshaped the TME, increased cytotoxic T lymphocyte (CTL) infiltration, and reduced regulatory T cells (Tregs). Mechanistically, B4M3 therapy upregulated TGF-β, promoting Th17 differentiation and CTL recruitment, thereby enhancing anti-tumor immunity. Our findings demonstrate that B4M3 CAR-T cells effectively address antigen heterogeneity and enhance therapeutic efficacy in OC, thereby offering a promising strategy for solid tumor immunotherapy.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145775209","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Unlocking the potential of targeting the angiotensin II type 1 receptor in cancer. 释放靶向血管紧张素II型1受体在癌症中的潜力。
IF 7.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-17 DOI: 10.1038/s41388-025-03666-9
David R Butcher, Christopher N Parris, Scott J Crichton, Fiona C Dempsey, Hussein N Al-Ali

The renin-angiotensin system is a key regulator of blood pressure homeostasis, with its primary effector, the angiotensin II type 1 receptor (AT1R), mediating vasoconstriction and processes fundamental to cancer progression, including proliferation, angiogenesis, and metastasis. Elevated AT1R expression is consistently linked to poor prognosis and therapeutic resistance across various malignancies. Preclinical studies provide compelling evidence that AT1R activation drives key cancer related processes, while its inhibition by angiotensin receptor blockers (ARBs) suppresses tumour growth, induces apoptosis, reduces angiogenesis, and inhibits metastasis across a wide range of cancer models. Critically, ARBs effectively modulate the tumour microenvironment (TME), alleviating fibrosis, promoting anti-tumour immune cell phenotypes, and enhancing the efficacy of targeted therapies, chemotherapies, and immunotherapies. Despite this strong preclinical evidence and supporting retrospective population studies, clinical translation of ARBs in oncology remains inconsistent, with trials often limited by design, patient heterogeneity, and supra-therapeutic ARB dosages required for acute anti-cancer effects. This review seeks to summarise the current understanding of AT1R's role in cancer, highlight preclinical and clinical investigations of targeting RAS, and suggest further strategies to unlock its therapeutic potential. Realising the full therapeutic promise of AT1R targeting in oncology requires a multifaceted approach, including the development of innovative delivery systems, such as TME-activated ARBs, and the exploration of advanced therapeutic modalities, such as antibody based AT1R inhibitors. Rigorously designed clinical trials that include biomarker-driven patient stratification to identify responsive cohorts are crucial to define the context-dependent role of AT1R and conclusively establish its clinical utility as a combinatorial strategy to enhance patient outcomes.

肾素-血管紧张素系统是血压稳态的关键调节因子,其主要效应器是血管紧张素II型1受体(AT1R),介导血管收缩和癌症进展的基本过程,包括增殖、血管生成和转移。在各种恶性肿瘤中,AT1R表达升高始终与预后不良和治疗耐药有关。临床前研究提供了令人信服的证据,表明AT1R激活驱动关键的癌症相关过程,而血管紧张素受体阻滞剂(ARBs)对其的抑制可以抑制肿瘤生长,诱导细胞凋亡,减少血管生成,并抑制多种癌症模型的转移。关键是,arb有效调节肿瘤微环境(TME),减轻纤维化,促进抗肿瘤免疫细胞表型,增强靶向治疗、化疗和免疫治疗的疗效。尽管有强有力的临床前证据和支持回顾性人群研究,但ARB在肿瘤学中的临床转化仍然不一致,试验往往受到设计、患者异质性和急性抗癌作用所需的超治疗ARB剂量的限制。本综述旨在总结目前对AT1R在癌症中的作用的理解,重点介绍靶向RAS的临床前和临床研究,并提出进一步的策略来释放其治疗潜力。实现AT1R靶向在肿瘤学中的全部治疗前景需要多方面的方法,包括开发创新的递送系统,如tme激活的arb,以及探索先进的治疗方式,如基于抗体的AT1R抑制剂。严格设计的临床试验,包括生物标志物驱动的患者分层,以确定响应队列,对于定义AT1R的环境依赖作用至关重要,并最终确定其作为一种组合策略的临床效用,以提高患者的预后。
{"title":"Unlocking the potential of targeting the angiotensin II type 1 receptor in cancer.","authors":"David R Butcher, Christopher N Parris, Scott J Crichton, Fiona C Dempsey, Hussein N Al-Ali","doi":"10.1038/s41388-025-03666-9","DOIUrl":"https://doi.org/10.1038/s41388-025-03666-9","url":null,"abstract":"<p><p>The renin-angiotensin system is a key regulator of blood pressure homeostasis, with its primary effector, the angiotensin II type 1 receptor (AT1R), mediating vasoconstriction and processes fundamental to cancer progression, including proliferation, angiogenesis, and metastasis. Elevated AT1R expression is consistently linked to poor prognosis and therapeutic resistance across various malignancies. Preclinical studies provide compelling evidence that AT1R activation drives key cancer related processes, while its inhibition by angiotensin receptor blockers (ARBs) suppresses tumour growth, induces apoptosis, reduces angiogenesis, and inhibits metastasis across a wide range of cancer models. Critically, ARBs effectively modulate the tumour microenvironment (TME), alleviating fibrosis, promoting anti-tumour immune cell phenotypes, and enhancing the efficacy of targeted therapies, chemotherapies, and immunotherapies. Despite this strong preclinical evidence and supporting retrospective population studies, clinical translation of ARBs in oncology remains inconsistent, with trials often limited by design, patient heterogeneity, and supra-therapeutic ARB dosages required for acute anti-cancer effects. This review seeks to summarise the current understanding of AT1R's role in cancer, highlight preclinical and clinical investigations of targeting RAS, and suggest further strategies to unlock its therapeutic potential. Realising the full therapeutic promise of AT1R targeting in oncology requires a multifaceted approach, including the development of innovative delivery systems, such as TME-activated ARBs, and the exploration of advanced therapeutic modalities, such as antibody based AT1R inhibitors. Rigorously designed clinical trials that include biomarker-driven patient stratification to identify responsive cohorts are crucial to define the context-dependent role of AT1R and conclusively establish its clinical utility as a combinatorial strategy to enhance patient outcomes.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145775257","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
MDC1 counteracts replication fork reversal and mediates chemosensitivity in BRCA1/2-deficient tumors. MDC1在brca1 /2缺陷肿瘤中抵消复制叉逆转并介导化学敏感性。
IF 7.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-17 DOI: 10.1038/s41388-025-03659-8
Hülya Dogan, Martin Liptay, Joana S Barbosa, Ewa Gogola, Alexandra A Duarte, Jonas A Schmid, Ismar Klebic, Merve Mutlu, Myriam Siffert, Paola Francica, Israel Salguero, Marieke van de Ven, Renske de Korte-Grimmerink, Stephen P Jackson, Jos Jonkers, Massimo Lopes, Diego Dibitetto, Sven Rottenberg

MDC1 is a key protein in DNA damage signaling. When DNA double-strand breaks (DSBs) occur, MDC1 localizes to the sites of DNA damage to promote the recruitment of other factors, including the 53BP1-mediated DSB repair pathway. By studying mechanisms of poly (ADP-ribose) polymerase inhibitor (PARPi) resistance in BRCA2; p53-deficient mouse mammary tumors, we identified a thus far unknown role of MDC1 in replication fork biology. Our results show that MDC1 localizes at active replication forks during normal DNA replication and regulates replication fork progression. It suppresses spontaneous fork reversal and regulates fork nucleolytic processing thereby promoting sensitivity to PARPi and cisplatin. In this way, MDC1 loss improves DNA damage tolerance and causes chemoresistance in BRCA1/2-deficient cells. We demonstrate that limiting MRE11 activity abolishes the reduced fork speed while MRE11 inhibition/depletion overcomes PARPi resistance in these cells. Overall, our data provides new insights into the role of MDC1 in replication fork progression that mediates PARPi- and cisplatin-induced DNA damage, in addition to its role in DSB repair.

MDC1是DNA损伤信号传导的关键蛋白。当DNA双链断裂(DSB)发生时,MDC1定位于DNA损伤位点,促进其他因子的募集,包括53bp1介导的DSB修复途径。通过研究BRCA2中聚(adp -核糖)聚合酶抑制剂(PARPi)耐药机制;p53缺陷小鼠乳腺肿瘤,我们确定了MDC1在复制叉生物学中迄今未知的作用。我们的研究结果表明,在正常的DNA复制过程中,MDC1定位于活跃的复制叉,并调节复制叉的进展。它抑制自发叉逆转并调节叉核裂解过程,从而促进对PARPi和顺铂的敏感性。通过这种方式,MDC1缺失改善了brca1 /2缺陷细胞的DNA损伤耐受性并引起化疗耐药。我们证明,限制MRE11活性可以消除降低的分叉速度,而MRE11抑制/耗尽可以克服这些细胞中的PARPi抗性。总的来说,我们的数据为MDC1在复制叉进程中的作用提供了新的见解,该进程介导PARPi和顺铂诱导的DNA损伤,以及它在DSB修复中的作用。
{"title":"MDC1 counteracts replication fork reversal and mediates chemosensitivity in BRCA1/2-deficient tumors.","authors":"Hülya Dogan, Martin Liptay, Joana S Barbosa, Ewa Gogola, Alexandra A Duarte, Jonas A Schmid, Ismar Klebic, Merve Mutlu, Myriam Siffert, Paola Francica, Israel Salguero, Marieke van de Ven, Renske de Korte-Grimmerink, Stephen P Jackson, Jos Jonkers, Massimo Lopes, Diego Dibitetto, Sven Rottenberg","doi":"10.1038/s41388-025-03659-8","DOIUrl":"https://doi.org/10.1038/s41388-025-03659-8","url":null,"abstract":"<p><p>MDC1 is a key protein in DNA damage signaling. When DNA double-strand breaks (DSBs) occur, MDC1 localizes to the sites of DNA damage to promote the recruitment of other factors, including the 53BP1-mediated DSB repair pathway. By studying mechanisms of poly (ADP-ribose) polymerase inhibitor (PARPi) resistance in BRCA2; p53-deficient mouse mammary tumors, we identified a thus far unknown role of MDC1 in replication fork biology. Our results show that MDC1 localizes at active replication forks during normal DNA replication and regulates replication fork progression. It suppresses spontaneous fork reversal and regulates fork nucleolytic processing thereby promoting sensitivity to PARPi and cisplatin. In this way, MDC1 loss improves DNA damage tolerance and causes chemoresistance in BRCA1/2-deficient cells. We demonstrate that limiting MRE11 activity abolishes the reduced fork speed while MRE11 inhibition/depletion overcomes PARPi resistance in these cells. Overall, our data provides new insights into the role of MDC1 in replication fork progression that mediates PARPi- and cisplatin-induced DNA damage, in addition to its role in DSB repair.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145775245","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
KRT81 promotes metastasis of colorectal cancer by acting as a protein scaffold for ezrin. KRT81作为ezrin蛋白支架促进结直肠癌转移。
IF 7.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-17 DOI: 10.1038/s41388-025-03665-w
Minghan Huang, Wenqing Xie, Meihua Wu, Zhimei Ou, Caibin Li, Shuhui Ji, Wanjun Liu, Min Zhi, Daici Chen

Metastasis to distant organs represents the most fatal prognostic factor for colorectal cancer (CRC). The distant metastasis of tumor cells results from the collaborative effort of multiple subcellular structures, with dynamic cytoskeletal remodeling underlying this entire process. Here, we found that knockdown of KRT81 expression (shKRT81) inhibited the proliferation, invasion, and migration, while ectopic overexpression of KRT81 enhanced the CRC cells migration. Furthermore, we identified a potential downstream effector of KRT81, ezrin, a member of the ezrin/radixin/moesin (ERM) protein family that regulates cell morphology and motility. Phenotypically, the shKRT81 attenuated ezrin protein expression and reduced the number and length of filopodia in CRC cells, which were restored when KRT81 was re-overexpressed. Mechanistically, KRT81 formed a complex with ezrin, and recruitment of ezrin to the membrane and phosphorylation at the Thr567 residue were significantly abolished in shKRT81 cells. Interestingly, we found that Myosin 1B (MYO1B) might provide the driving force for the recruitment of ezrin. Notably, combinatorial inhibition (shKRT81 + ezrin-specific inhibitor) exerted significantly greater suppression of CRC cell migration and invasion than either intervention alone. Consistently, KRT81 expression was increased in CRC, and relatively high expression of KRT81 was associated with a poor prognosis. In summary, we identified a novel regulatory axis that involves KRT81, MYO1B, and ezrin, which regulates filopodia formation and migration behavior in CRC. Therefore, KRT81 may serve as a therapeutic target for CRC.

远处器官转移是结直肠癌(CRC)最致命的预后因素。肿瘤细胞的远端转移是多个亚细胞结构协同作用的结果,整个过程背后是动态的细胞骨架重塑。本研究发现,KRT81表达下调(shKRT81)可抑制CRC细胞的增殖、侵袭和迁移,而KRT81异位过表达可增强CRC细胞的迁移。此外,我们还发现了KRT81的潜在下游效应物ezrin,它是ezrin/radixin/moesin (ERM)蛋白家族的一员,可以调节细胞形态和运动。表型上,shKRT81降低了CRC细胞中ezrin蛋白的表达,减少了丝状足的数量和长度,当KRT81再次过表达时,丝状足的数量和长度得以恢复。在机制上,KRT81与ezrin形成复合物,在shKRT81细胞中,ezrin的膜募集和Thr567残基的磷酸化被显著消除。有趣的是,我们发现Myosin 1B (MYO1B)可能为ezrin的募集提供了驱动力。值得注意的是,组合抑制(shKRT81 + ezrin特异性抑制剂)对结直肠癌细胞迁移和侵袭的抑制作用明显大于单独干预。同样,KRT81在结直肠癌中表达升高,KRT81的相对高表达与预后不良相关。总之,我们发现了一个涉及KRT81、MYO1B和ezrin的新的调控轴,它调节CRC中丝状足的形成和迁移行为。因此,KRT81可能作为结直肠癌的治疗靶点。
{"title":"KRT81 promotes metastasis of colorectal cancer by acting as a protein scaffold for ezrin.","authors":"Minghan Huang, Wenqing Xie, Meihua Wu, Zhimei Ou, Caibin Li, Shuhui Ji, Wanjun Liu, Min Zhi, Daici Chen","doi":"10.1038/s41388-025-03665-w","DOIUrl":"https://doi.org/10.1038/s41388-025-03665-w","url":null,"abstract":"<p><p>Metastasis to distant organs represents the most fatal prognostic factor for colorectal cancer (CRC). The distant metastasis of tumor cells results from the collaborative effort of multiple subcellular structures, with dynamic cytoskeletal remodeling underlying this entire process. Here, we found that knockdown of KRT81 expression (shKRT81) inhibited the proliferation, invasion, and migration, while ectopic overexpression of KRT81 enhanced the CRC cells migration. Furthermore, we identified a potential downstream effector of KRT81, ezrin, a member of the ezrin/radixin/moesin (ERM) protein family that regulates cell morphology and motility. Phenotypically, the shKRT81 attenuated ezrin protein expression and reduced the number and length of filopodia in CRC cells, which were restored when KRT81 was re-overexpressed. Mechanistically, KRT81 formed a complex with ezrin, and recruitment of ezrin to the membrane and phosphorylation at the Thr567 residue were significantly abolished in shKRT81 cells. Interestingly, we found that Myosin 1B (MYO1B) might provide the driving force for the recruitment of ezrin. Notably, combinatorial inhibition (shKRT81 + ezrin-specific inhibitor) exerted significantly greater suppression of CRC cell migration and invasion than either intervention alone. Consistently, KRT81 expression was increased in CRC, and relatively high expression of KRT81 was associated with a poor prognosis. In summary, we identified a novel regulatory axis that involves KRT81, MYO1B, and ezrin, which regulates filopodia formation and migration behavior in CRC. Therefore, KRT81 may serve as a therapeutic target for CRC.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145775188","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
YTHDC2 inhibits the resistance of lung cancer to EGFR-TKI through cuproptosis. YTHDC2通过cuprosis抑制肺癌对EGFR-TKI的抵抗。
IF 7.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-16 DOI: 10.1038/s41388-025-03660-1
Jizhuang Luo, Xin Xu, Yaohui Chen, Yiwen Huang, Yiman Huang, Yajuan Zhang, Lifang Ma, Tianxiang Chen

While third-generation EGFR tyrosine kinase inhibitors (EGFR-TKIs), such as osimertinib, have significantly improved patient survival in non-small cell lung cancer (NSCLC), acquired resistance remains a major clinical challenge, and its underlying mechanisms are incompletely understood. In this study, we demonstrate that YTHDC2 expression is significantly downregulated in osimertinib-resistant patient-derived xenograft (PDX) tissues and lung cancer cell lines compared to their osimertinib-sensitive counterparts. Further investigation revealed that YTHDC2 overcomes osimertinib resistance in lung cancer cells by promoting cuproptosis. Mechanistically, YTHDC2 binds to m6A-modified sites (specifically at nucleotides A1223 and A2824) within the mRNA of the copper transporter SLC31A1 in an m6A-dependent manner. This interaction enhances SLC31A1 mRNA stability and protein expression, thereby increasing intracellular copper transport and inducing cuproptosis in tumor cells. Additionally, we found that the copper ionophore disulfiram (DSF) overcame osimertinib resistance by augmenting YTHDC2 expression. Collectively, our findings elucidate a novel YTHDC2-SLC31A1-cuproptosis axis as a key mechanism underlying EGFR-TKI resistance and propose new therapeutic strategies for its reversal.

虽然第三代EGFR酪氨酸激酶抑制剂(EGFR- tkis),如奥西替尼,显著提高了非小细胞肺癌(NSCLC)患者的生存率,但获得性耐药仍然是一个主要的临床挑战,其潜在机制尚不完全清楚。在这项研究中,我们证明了YTHDC2的表达在奥西替尼耐药的患者来源的异种移植(PDX)组织和肺癌细胞系中,与对奥西替尼敏感的组织和肺癌细胞系相比,显著下调。进一步研究发现,YTHDC2通过促进铜增生来克服肺癌细胞对奥西替尼的耐药。在机制上,YTHDC2以m6a依赖的方式结合铜转运体SLC31A1的mRNA中m6a修饰位点(特别是核苷酸A1223和A2824)。这种相互作用增强了SLC31A1 mRNA的稳定性和蛋白的表达,从而增加了肿瘤细胞内铜的运输,诱导了铜增生。此外,我们发现铜离子载体二硫醚(DSF)通过增加YTHDC2表达克服了奥希替尼耐药性。总之,我们的研究结果阐明了新的ythdc2 - slc31a1 - cuprotosis轴是EGFR-TKI耐药的关键机制,并提出了新的治疗策略来逆转它。
{"title":"YTHDC2 inhibits the resistance of lung cancer to EGFR-TKI through cuproptosis.","authors":"Jizhuang Luo, Xin Xu, Yaohui Chen, Yiwen Huang, Yiman Huang, Yajuan Zhang, Lifang Ma, Tianxiang Chen","doi":"10.1038/s41388-025-03660-1","DOIUrl":"https://doi.org/10.1038/s41388-025-03660-1","url":null,"abstract":"<p><p>While third-generation EGFR tyrosine kinase inhibitors (EGFR-TKIs), such as osimertinib, have significantly improved patient survival in non-small cell lung cancer (NSCLC), acquired resistance remains a major clinical challenge, and its underlying mechanisms are incompletely understood. In this study, we demonstrate that YTHDC2 expression is significantly downregulated in osimertinib-resistant patient-derived xenograft (PDX) tissues and lung cancer cell lines compared to their osimertinib-sensitive counterparts. Further investigation revealed that YTHDC2 overcomes osimertinib resistance in lung cancer cells by promoting cuproptosis. Mechanistically, YTHDC2 binds to m<sup>6</sup>A-modified sites (specifically at nucleotides A1223 and A2824) within the mRNA of the copper transporter SLC31A1 in an m<sup>6</sup>A-dependent manner. This interaction enhances SLC31A1 mRNA stability and protein expression, thereby increasing intracellular copper transport and inducing cuproptosis in tumor cells. Additionally, we found that the copper ionophore disulfiram (DSF) overcame osimertinib resistance by augmenting YTHDC2 expression. Collectively, our findings elucidate a novel YTHDC2-SLC31A1-cuproptosis axis as a key mechanism underlying EGFR-TKI resistance and propose new therapeutic strategies for its reversal.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-12-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145768536","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Hotspot mutations in HER2 interfaces destabilize structure, causing breast cancer treatment failure. HER2界面热点突变破坏结构稳定,导致乳腺癌治疗失败。
IF 7.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-15 DOI: 10.1038/s41388-025-03653-0
Pranay Dey, Aniketh Bishnu, Janvi Patel, Shivali Mishra, Nikhil Gadewal, Pritha Ray, Sudeep Gupta, Abhijit De
{"title":"Hotspot mutations in HER2 interfaces destabilize structure, causing breast cancer treatment failure.","authors":"Pranay Dey, Aniketh Bishnu, Janvi Patel, Shivali Mishra, Nikhil Gadewal, Pritha Ray, Sudeep Gupta, Abhijit De","doi":"10.1038/s41388-025-03653-0","DOIUrl":"10.1038/s41388-025-03653-0","url":null,"abstract":"","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145757300","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Phosphorylation of CBX8 by PKD1 suppresses PRC1 activity and promotes cell senescence. PKD1磷酸化CBX8抑制PRC1活性,促进细胞衰老。
IF 7.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-15 DOI: 10.1038/s41388-025-03657-w
Zhiqiang Fang, Doudou Liu, Yuanyuan Su, Fengxin Hao, Ruodong Qin, Guodong Li, Jun Chen

The Polycomb group (PcG) protein chromobox 8 (CBX8) is the subunit of Polycomb repressive complex 1 (PRC1) and recognizes the trimethylation of histone H3 on Lysine 27 (H3K27me3), and coordinates with PRC2 complex to function as an epigenetic gene silencer. CBX8 plays a key role in cell proliferation, stem cell biology, cell senescence, and cancer development. However, the post-translational modifications of CBX8 remain poorly understood. Here, we report that protein kinase D1 (PKD1) interacts and phosphorylates CBX8 at Thr234 and Ser256 /311 residues. PKD1-mediated CBX8 phosphorylation at Thr234 reduced its expression level by promoting its ubiquitination-mediated degradation, whereas Ser256/311 phosphorylation decreased CBX8 binding to other PRC1 components BMI1 and RING1A. Overall, CBX8 phosphorylation by PKD1 impaired PRC1 complex integrity and activity, mitigated H2AK119ub1 level, caused the upregulation of multiple target genes repressed by CBX8, and decreased CBX8, H2AK119ub1, and H3K27me3 enrichment at INK4A/ARF locus, thereby derepressing p16INK4A and facilitating cellular senescence. Collectively, these results suggest that PKD1-mediated CBX8 phosphorylation at T234 and S256/311 is a key mechanism governing CBX8 function, including cell senescence.

Polycomb组(PcG)蛋白色盒8 (CBX8)是Polycomb抑制复合体1 (PRC1)的亚基,识别赖氨酸27 (H3K27me3)上组蛋白H3的三甲基化,并与PRC2复合体协同发挥表观遗传基因沉默剂的作用。CBX8在细胞增殖、干细胞生物学、细胞衰老和癌症发展中起关键作用。然而,CBX8的翻译后修饰仍然知之甚少。在这里,我们报道了蛋白激酶D1 (PKD1)相互作用并磷酸化CBX8的Thr234和Ser256 /311残基。pkd1介导的CBX8 Thr234磷酸化通过促进其泛素化介导的降解降低了其表达水平,而Ser256/311磷酸化降低了CBX8与其他PRC1组分BMI1和RING1A的结合。综上所述,PKD1磷酸化CBX8损害了PRC1复合物的完整性和活性,降低了H2AK119ub1水平,导致CBX8抑制的多个靶基因上调,降低了INK4A/ARF位点CBX8、H2AK119ub1和H3K27me3的富集,从而降低了p16INK4A的表达,促进了细胞衰老。总之,这些结果表明pkd1介导的CBX8 T234和S256/311位点磷酸化是控制CBX8功能的关键机制,包括细胞衰老。
{"title":"Phosphorylation of CBX8 by PKD1 suppresses PRC1 activity and promotes cell senescence.","authors":"Zhiqiang Fang, Doudou Liu, Yuanyuan Su, Fengxin Hao, Ruodong Qin, Guodong Li, Jun Chen","doi":"10.1038/s41388-025-03657-w","DOIUrl":"https://doi.org/10.1038/s41388-025-03657-w","url":null,"abstract":"<p><p>The Polycomb group (PcG) protein chromobox 8 (CBX8) is the subunit of Polycomb repressive complex 1 (PRC1) and recognizes the trimethylation of histone H3 on Lysine 27 (H3K27me3), and coordinates with PRC2 complex to function as an epigenetic gene silencer. CBX8 plays a key role in cell proliferation, stem cell biology, cell senescence, and cancer development. However, the post-translational modifications of CBX8 remain poorly understood. Here, we report that protein kinase D1 (PKD1) interacts and phosphorylates CBX8 at Thr234 and Ser256 /311 residues. PKD1-mediated CBX8 phosphorylation at Thr234 reduced its expression level by promoting its ubiquitination-mediated degradation, whereas Ser256/311 phosphorylation decreased CBX8 binding to other PRC1 components BMI1 and RING1A. Overall, CBX8 phosphorylation by PKD1 impaired PRC1 complex integrity and activity, mitigated H2AK119ub1 level, caused the upregulation of multiple target genes repressed by CBX8, and decreased CBX8, H2AK119ub1, and H3K27me3 enrichment at INK4A/ARF locus, thereby derepressing p16<sup>INK4A</sup> and facilitating cellular senescence. Collectively, these results suggest that PKD1-mediated CBX8 phosphorylation at T234 and S256/311 is a key mechanism governing CBX8 function, including cell senescence.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145763466","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Correction: GALNT5 uaRNA promotes gastric cancer progression through its interaction with HSP90. 更正:galnt5warna通过与HSP90的相互作用促进胃癌进展。
IF 7.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-15 DOI: 10.1038/s41388-025-03668-7
Hui Guo, Lianmei Zhao, Bianhua Shi, Jiayu Bao, Dexian Zheng, Baoguo Zhou, Juan Shi
{"title":"Correction: GALNT5 uaRNA promotes gastric cancer progression through its interaction with HSP90.","authors":"Hui Guo, Lianmei Zhao, Bianhua Shi, Jiayu Bao, Dexian Zheng, Baoguo Zhou, Juan Shi","doi":"10.1038/s41388-025-03668-7","DOIUrl":"https://doi.org/10.1038/s41388-025-03668-7","url":null,"abstract":"","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145763500","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Oncogene
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1