Oncogene最新文献

筛选
英文 中文
Cooperation between SIX1 and DHX9 transcriptionally regulates integrin-focal adhesion signaling mediated metastasis and sunitinib resistance in KIRC. SIX1和DHX9之间的合作转录调节了KIRC中整合素-局灶粘附信号介导的转移和舒尼替尼耐药性。
IF 6.9 1区 医学
Oncogene Pub Date : 2024-08-22 DOI: 10.1038/s41388-024-03126-w
Shiyu Huang, Juncheng Hu, Min Hu, Yanguang Hou, Banghua Zhang, Jiachen Liu, Xiuheng Liu, Zhiyuan Chen, Lei Wang
{"title":"Cooperation between SIX1 and DHX9 transcriptionally regulates integrin-focal adhesion signaling mediated metastasis and sunitinib resistance in KIRC.","authors":"Shiyu Huang, Juncheng Hu, Min Hu, Yanguang Hou, Banghua Zhang, Jiachen Liu, Xiuheng Liu, Zhiyuan Chen, Lei Wang","doi":"10.1038/s41388-024-03126-w","DOIUrl":"https://doi.org/10.1038/s41388-024-03126-w","url":null,"abstract":"<p><p>High invasive capacity and acquired tyrosine kinase inhibitors (TKI) resistance of kidney renal clear cell carcinoma (KIRC) cells remain obstacles to prolonging the survival time of patients with advanced KIRC. In the present study, we reported that sine oculis homeobox 1 (SIX1) was upregulated in sunitinib-resistant KIRC cells and metastatic KIRC tissues. Subsequently, we found that SIX1 mediated metastasis and sunitinib resistance via Focal adhesion (FA) signaling, and knockdown of SIX1 enhanced the antitumor efficiency of sunitinib in KIRC. Mechanistically, Integrin subunit beta 1 (ITGB1), an upstream gene of FA signaling, was a direct transcriptional target of SIX1. In addition, we showed that DExH-box helicase 9 (DHX9) was an important mediator for SIX1-induced ITGB1 transcription, and silencing the subunits of SIX1/DHX9 complex significantly reduced transcription of ITGB1. Downregulation of SIX1 attenuated nuclear translocation of DHX9 and abrogated the binding of DHX9 to ITGB1 promoter. Collectively, our results unveiled a new signal axis SIX1/ITGB1/FAK in KIRC and identified a novel therapeutic strategy for metastatic KIRC patients.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":null,"pages":null},"PeriodicalIF":6.9,"publicationDate":"2024-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142036468","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
BRD4-specific PROTAC inhibits basal-like breast cancer partially through downregulating KLF5 expression. BRD4 特异性 PROTAC 部分通过下调 KLF5 的表达抑制基底样乳腺癌。
IF 6.9 1区 医学
Oncogene Pub Date : 2024-08-20 DOI: 10.1038/s41388-024-03121-1
Yanjie Kong, Tianlong Lan, Luzhen Wang, Chen Gong, Wenxin Lv, Hailin Zhang, Chengang Zhou, Xiuyun Sun, Wenjing Liu, Haihui Huang, Xin Weng, Chang Cai, Wenfeng Peng, Meng Zhang, Dewei Jiang, Chuanyu Yang, Xia Liu, Yu Rao, Ceshi Chen
{"title":"BRD4-specific PROTAC inhibits basal-like breast cancer partially through downregulating KLF5 expression.","authors":"Yanjie Kong, Tianlong Lan, Luzhen Wang, Chen Gong, Wenxin Lv, Hailin Zhang, Chengang Zhou, Xiuyun Sun, Wenjing Liu, Haihui Huang, Xin Weng, Chang Cai, Wenfeng Peng, Meng Zhang, Dewei Jiang, Chuanyu Yang, Xia Liu, Yu Rao, Ceshi Chen","doi":"10.1038/s41388-024-03121-1","DOIUrl":"https://doi.org/10.1038/s41388-024-03121-1","url":null,"abstract":"<p><p>Interest in the use of proteolysis-targeting chimeras (PROTACs) in cancer therapy has increased in recent years. Targeting bromodomain and extra terminal domain (BET) proteins, especially bromodomain-containing protein 4 (BRD4), has shown inhibitory effects on basal-like breast cancer (BLBC). However, the bioavailability of BRD4 PROTACs is restricted by their non-selective biodegradability and low tumor-targeting ability. We demonstrated that 6b (BRD4 PROTAC) suppresses BLBC cell growth by targeting BRD4, but not BRD2 and BRD3, for cereblon (CRBN)-mediated ubiquitination and proteasomal degradation. Compound 6b also inhibited expression of Krüppel-like factor 5 (KLF5) transcription factor, a key oncoprotein in BLBC, controlled by BRD4-mediated super-enhancers. Moreover, 6b inhibited HCC1806 tumor growth in a xenograft mouse model. The combination of 6b and KLF5 inhibitors showed additive effects on BLBC. These results suggest that BRD4-specific PROTAC can effectively inhibit BLBC by downregulating KLF5, and that 6b has potential as a novel therapeutic drug for BLBC.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":null,"pages":null},"PeriodicalIF":6.9,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142009171","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
Alternative splicing of ALDOA confers tamoxifen resistance in breast cancer. ALDOA 的交替剪接使乳腺癌产生他莫昔芬抗药性。
IF 6.9 1区 医学
Oncogene Pub Date : 2024-08-20 DOI: 10.1038/s41388-024-03134-w
Shiyi Yu, Rui Wu, Yue Si, Zhehao Fan, Ying Wang, Chang Yao, Rongmao Sun, Yaji Xue, Yongli Chen, Zheng Wang, Shuangshuang Dong, Ning Wang, Xinyue Ling, Zhengyan Liang, Caili Bi, Yi Yang, Weibing Dong, Haibo Sun
{"title":"Alternative splicing of ALDOA confers tamoxifen resistance in breast cancer.","authors":"Shiyi Yu, Rui Wu, Yue Si, Zhehao Fan, Ying Wang, Chang Yao, Rongmao Sun, Yaji Xue, Yongli Chen, Zheng Wang, Shuangshuang Dong, Ning Wang, Xinyue Ling, Zhengyan Liang, Caili Bi, Yi Yang, Weibing Dong, Haibo Sun","doi":"10.1038/s41388-024-03134-w","DOIUrl":"https://doi.org/10.1038/s41388-024-03134-w","url":null,"abstract":"<p><p>The cancer-associated alternative splicing (AS) events generate cancer-related transcripts which are involved in uncontrolled cell proliferation and drug resistance. However, the key AS variants implicated in tamoxifen (TAM) resistance in breast cancer remain elusive. In the current study, we investigated the landscape of AS events in nine pairs of primary and relapse breast tumors from patients receiving TAM-based therapy. We unrevealed a notable association between the inclusion of exon 7.2 in the 5'untranslated region (5'UTR) of ALDOA mRNA and TAM resistance. Mechanistically, the inclusion of ALDOA exon 7.2 enhances the translation efficiency of the transcript, resulting in increased ALDOA protein expression, mTOR pathway activity, and the promotion of TAM resistance in breast cancer cells. Moreover, the inclusion of exon 7.2 in ALDOA mRNA is mediated by MSI1 via direct interaction. In addition, elevated inclusion of ALDOA exon 7.2 or expression of MSI1 is associated with an unfavorable prognosis in patients undergoing endocrine therapy. Notably, treatment with Aldometanib, an ALDOA inhibitor, effectively restrains the growth of TAM-resistant breast cancer cells in vitro and in vivo. The present study unveils the pivotal role of an AS event in ALDOA, under the regulation of MSI1, in driving TAM resistance in breast cancer. Therefore, this study provides a promising therapeutic avenue targeting ALDOA to combat TAM resistance.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":null,"pages":null},"PeriodicalIF":6.9,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142009170","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
Mapping the breast tumor microenvironment: proximity analysis reveals spatial relationships between macrophage subtypes and metastasis-initiating cancer cells. 绘制乳腺肿瘤微环境图:接近性分析揭示了巨噬细胞亚型与转移启动癌细胞之间的空间关系。
IF 6.9 1区 医学
Oncogene Pub Date : 2024-08-20 DOI: 10.1038/s41388-024-03127-9
Eloïse M Grasset, Atul Deshpande, Jae W Lee, Yeonju Cho, Sarah M Shin, Erin M Coyne, Alexei Hernandez, Xuan Yuan, Zhehao Zhang, Ashley Cimino-Mathews, Andrew J Ewald, Won Jin Ho
{"title":"Mapping the breast tumor microenvironment: proximity analysis reveals spatial relationships between macrophage subtypes and metastasis-initiating cancer cells.","authors":"Eloïse M Grasset, Atul Deshpande, Jae W Lee, Yeonju Cho, Sarah M Shin, Erin M Coyne, Alexei Hernandez, Xuan Yuan, Zhehao Zhang, Ashley Cimino-Mathews, Andrew J Ewald, Won Jin Ho","doi":"10.1038/s41388-024-03127-9","DOIUrl":"https://doi.org/10.1038/s41388-024-03127-9","url":null,"abstract":"<p><p>Metastasis is responsible for the majority of cancer-related fatalities. We previously identified specific cancer cell populations responsible for metastatic events which are cytokeratin-14 (CK14) and E-cadherin positive in luminal tumors, and E-cadherin and vimentin positive in triple-negative tumors. Since cancer cells evolve within a complex ecosystem comprised of immune cells and stromal cells, we sought to decipher the spatial interactions of these aggressive cancer cell populations within the tumor microenvironment (TME). We used imaging mass cytometry to detect 36 proteins in tumor microarrays containing paired primary and metastatic lesions from luminal or triple-negative breast cancers (TNBC), resulting in a dataset of 1,477,337 annotated cells. Focusing on metastasis-initiating cell populations, we observed close proximity to specific fibroblast and macrophage subtypes, a relationship maintained between primary and metastatic tumors. Notably, high CK14 in luminal cancer cells and high vimentin in TNBC cells correlated with close proximity to specific macrophage subtypes (CD163<sup>int</sup>CD206<sup>int</sup>PDL1<sup>int</sup>HLA-DR<sup>+</sup> or PDL1<sup>high</sup>ARG1<sup>high</sup>). Our in-depth spatial analysis demonstrates that metastasis-initiating cancer cells consistently colocalizes with distinct cell populations within the TME, suggesting a role for these cell-cell interactions in promoting metastasis.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":null,"pages":null},"PeriodicalIF":6.9,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142009172","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
Jumonji histone demethylases are therapeutic targets in small cell lung cancer Jumonji组蛋白去甲基化酶是小细胞肺癌的治疗靶点。
IF 6.9 1区 医学
Oncogene Pub Date : 2024-08-18 DOI: 10.1038/s41388-024-03125-x
Aiden Nguyen, Clarissa G. Nuñez, Tram Anh Tran, Luc Girard, Michael Peyton, Rodrigo Catalan, Cristina Guerena, Kimberley Avila, Benjamin J. Drapkin, Raghav Chandra, John D. Minna, Elisabeth D. Martinez
{"title":"Jumonji histone demethylases are therapeutic targets in small cell lung cancer","authors":"Aiden Nguyen,&nbsp;Clarissa G. Nuñez,&nbsp;Tram Anh Tran,&nbsp;Luc Girard,&nbsp;Michael Peyton,&nbsp;Rodrigo Catalan,&nbsp;Cristina Guerena,&nbsp;Kimberley Avila,&nbsp;Benjamin J. Drapkin,&nbsp;Raghav Chandra,&nbsp;John D. Minna,&nbsp;Elisabeth D. Martinez","doi":"10.1038/s41388-024-03125-x","DOIUrl":"10.1038/s41388-024-03125-x","url":null,"abstract":"Small cell lung cancer (SCLC) is a recalcitrant cancer of neuroendocrine (NE) origin. Changes in therapeutic approaches against SCLC have been lacking over the decades. Here, we use preclinical models to identify a new therapeutic vulnerability in SCLC consisting of the targetable Jumonji lysine demethylase (KDM) family. We show that Jumonji demethylase inhibitors block malignant growth and that etoposide-resistant SCLC cell lines are particularly sensitive to Jumonji inhibition. Mechanistically, small molecule-mediated inhibition of Jumonji KDMs activates endoplasmic reticulum (ER) stress genes, upregulates ER stress signaling, and triggers apoptotic cell death. Furthermore, Jumonji inhibitors decrease protein levels of SCLC NE markers INSM1 and Secretogranin-3 and of driver transcription factors ASCL1 and NEUROD1. Genetic knockdown of KDM4A, a Jumonji demethylase highly expressed in SCLC and a known regulator of ER stress genes, induces ER stress response genes, decreases INSM1, Secretogranin-3, and NEUROD1 and inhibits proliferation of SCLC in vitro and in vivo. Lastly, we demonstrate that two different small molecule Jumonji KDM inhibitors (pan-inhibitor JIB-04 and KDM4 inhibitor SD70) block the growth of SCLC tumor xenografts in vivo. Our study highlights the translational potential of Jumonji KDM inhibitors against SCLC, a clinically feasible approach in light of recently opened clinical trials evaluating this drug class, and establishes KDM4A as a relevant target across SCLC subtypes.","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":null,"pages":null},"PeriodicalIF":6.9,"publicationDate":"2024-08-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41388-024-03125-x.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141996239","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Monoacylglycerol lipase blockades the senescence-associated secretory phenotype by interfering with NF-κB activation and promotes docetaxel efficacy in prostate cancer 单酰基甘油脂肪酶通过干扰NF-κB的激活来阻断衰老相关的分泌表型,并促进多西他赛在前列腺癌中的疗效。
IF 6.9 1区 医学
Oncogene Pub Date : 2024-08-18 DOI: 10.1038/s41388-024-03132-y
Jianpeng Yu, Minghao Zhang, Taipeng Li, Wenlong Gao, Zhao Yang, Keruo Wang, Zihao Liu, Shimiao Zhu, Simeng Wen, Yang Zhao, Qiliang Cai, Zhiqun Shang, Yong Wang, Yuanjie Niu
{"title":"Monoacylglycerol lipase blockades the senescence-associated secretory phenotype by interfering with NF-κB activation and promotes docetaxel efficacy in prostate cancer","authors":"Jianpeng Yu,&nbsp;Minghao Zhang,&nbsp;Taipeng Li,&nbsp;Wenlong Gao,&nbsp;Zhao Yang,&nbsp;Keruo Wang,&nbsp;Zihao Liu,&nbsp;Shimiao Zhu,&nbsp;Simeng Wen,&nbsp;Yang Zhao,&nbsp;Qiliang Cai,&nbsp;Zhiqun Shang,&nbsp;Yong Wang,&nbsp;Yuanjie Niu","doi":"10.1038/s41388-024-03132-y","DOIUrl":"10.1038/s41388-024-03132-y","url":null,"abstract":"Metabolic reprogramming and cellular senescence greatly contribute to cancer relapse and recurrence. In aging and treated prostate, persistent accumulating senescence-associated secretory phenotype (SASP) of cancer cells often limits the overall survival of patients. Novel strategic therapy with monoacylglycerol lipase (MGLL) upregulation that counters the cellular and docetaxel induced SASP might overcome this clinical challenge in prostate cancer (PCa). With primary comparative expression and survival analysis screening of fatty acid (FA) metabolism signature genes in the TCGA PCa dataset and our single center cohort, MGLL was detected to be downregulated in malignancy prostate tissues and its low expression predicted worse progression-free and overall survival. Functionally, overexpression of MGLL mainly suppresses NF-κB-driven SASP (N-SASP) which mostly restricts the cancer cell paracrine and autocrine tumorigenic manners and the corresponding cellular senescence. Further investigating metabolites, we determined that MGLL constitutive expression prevents lipid accumulation, decreases metabolites preferably, and consequently downregulates ATP levels. Overexpressed MGLL inhibited IκBα phosphorylation, NF-κB p65 phosphorylation, and NF-κB nuclear translocation to deactivate NF-κB transcriptional activities, and be responsible for the repressed N-SASP, partially through reducing ATP levels. Preclinically, combinational treatment with MGLL overexpression and docetaxel chemotherapy dramatically delays tumor progression in mouse models. Taken together, our findings identify MGLL as a switch for lipase-related N-SASP suppression and provide a potential drug candidate for promoting docetaxel efficacy in PCa.","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":null,"pages":null},"PeriodicalIF":6.9,"publicationDate":"2024-08-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142000443","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
Ac4C modification of lncRNA SIMALR promotes nasopharyngeal carcinoma progression through activating eEF1A2 to facilitate ITGB4/ITGA6 translation lncRNA SIMALR的Ac4C修饰通过激活eEF1A2促进ITGB4/ITGA6的翻译,从而促进鼻咽癌的进展。
IF 6.9 1区 医学
Oncogene Pub Date : 2024-08-18 DOI: 10.1038/s41388-024-03133-x
Sha Gong, Han Qiao, Jing-Yun Wang, Sheng-Yan Huang, Shi-Wei He, Yin Zhao, Xi-Rong Tan, Ming-Liang Ye, Jun-Yan Li, Ye-Lin Liang, Sai-Wei Huang, Jun Chen, Xun-Hua Zhu, Na Liu, Ying-Qing Li
{"title":"Ac4C modification of lncRNA SIMALR promotes nasopharyngeal carcinoma progression through activating eEF1A2 to facilitate ITGB4/ITGA6 translation","authors":"Sha Gong,&nbsp;Han Qiao,&nbsp;Jing-Yun Wang,&nbsp;Sheng-Yan Huang,&nbsp;Shi-Wei He,&nbsp;Yin Zhao,&nbsp;Xi-Rong Tan,&nbsp;Ming-Liang Ye,&nbsp;Jun-Yan Li,&nbsp;Ye-Lin Liang,&nbsp;Sai-Wei Huang,&nbsp;Jun Chen,&nbsp;Xun-Hua Zhu,&nbsp;Na Liu,&nbsp;Ying-Qing Li","doi":"10.1038/s41388-024-03133-x","DOIUrl":"10.1038/s41388-024-03133-x","url":null,"abstract":"The dysregulation of long non-coding RNAs (lncRNAs) are involved in regulating tumor progression in multiple manner. However, little is known about whether lncRNA is involved in the translation regulation of proteins. Here, we identified that the suppressor of inflammatory macrophage apoptosis lncRNA (SIMALR) was highly expressed in nasopharyngeal carcinoma (NPC) tissues by analyzing the lncRNA microarray. Clinically, the high expression of SIMALR served as an independent predictor for inferior prognosis in NPC patients. SIMALR functioned as an oncogenic lncRNA that promoted the proliferation and metastasis of NPC cells in vitro and in vivo. Mechanistically, SIMALR served as a critical accelerator of protein synthesis by binding to eEF1A2 (eukaryotic translation elongation factor 1 alpha 2), one of the most crucial regulators in the translation machinery of the eukaryotic cells, and enhancing its endogenous GTPase activity. Furthermore, SIMALR mediated the activation of eEF1A2 phosphorylation to accelerate the translation of ITGB4/ITGA6, ultimately promoting the malignant phenotype of NPC cells. In addition, N-acetyltransferase 10 (NAT10) enhanced the stability of SIMALR and caused its overexpression in NPC through the N4-acetylcytidine (ac4C) modification. In sum, our results illustrate SIMALR functions as an accelerator for protein translation and highlight the oncogenic role of NAT10-SIMALR-eEF1A2-ITGB4/6 axis in NPC.","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":null,"pages":null},"PeriodicalIF":6.9,"publicationDate":"2024-08-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41388-024-03133-x.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141996237","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Engineered targeting OIP5 sensitizes bladder cancer to chemotherapy resistance via TRIP12-PPP1CB-YBX1 axis 通过 TRIP12-PPP1CB-YBX1 轴设计靶向 OIP5 可使膀胱癌对化疗产生耐药性。
IF 6.9 1区 医学
Oncogene Pub Date : 2024-08-18 DOI: 10.1038/s41388-024-03136-8
Xianteng Wang, Ting Guo, Liman Niu, Binbin Zheng, Wei Huang, Haibo Xu, Weiren Huang
{"title":"Engineered targeting OIP5 sensitizes bladder cancer to chemotherapy resistance via TRIP12-PPP1CB-YBX1 axis","authors":"Xianteng Wang,&nbsp;Ting Guo,&nbsp;Liman Niu,&nbsp;Binbin Zheng,&nbsp;Wei Huang,&nbsp;Haibo Xu,&nbsp;Weiren Huang","doi":"10.1038/s41388-024-03136-8","DOIUrl":"10.1038/s41388-024-03136-8","url":null,"abstract":"Chemoresistance is an important cause of treatment failure in bladder cancer, and identifying genes that confer drug resistance is an important step toward developing new therapeutic strategies to improve treatment outcomes. In the present study, we show that gemcitabine plus cisplatin (GEM/DDP) therapy induces NF-κB signaling, which promotes p65-mediated transcriptional activation of OIP5. OIP5 recruits the E3 ubiquitin ligase TRIP12 to bind to and degrade the phosphatase PPP1CB, thereby enhancing the transcription factor activity of YBX1. This in turn upregulates drug-resistance-related genes under the transcriptional control of YBX1, leading to chemoresistance. Moreover, PPP1CB degradation can enhance the phosphorylation activity of IKKβ, triggering the NF-κB signaling cascade, which further stimulates OIP5 gene expression, thus forming a negative feedback regulatory loop. Consistently, elevated OIP5 expression was associated with chemoresistance and poor prognosis in patients with bladder cancer. Furthermore, we used a CRISPR/Cas9-based engineered gene circuit, which can monitor the progression of chemoresistance in real-time, to induce OIP5 knockout upon detection of increased NF-κB signaling. The gene circuit significantly inhibited tumor cell growth in vivo, underscoring the potential for synergy between gene therapy and chemotherapy in the treatment of cancer.","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":null,"pages":null},"PeriodicalIF":6.9,"publicationDate":"2024-08-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.com/articles/s41388-024-03136-8.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142000442","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
MiR-183-5p inhibits lung squamous cell carcinoma survival through disrupting hypoxia adaptation mediated by HIF-1α/NDUFA4L2 axis MiR-183-5p通过破坏由HIF-1α/NDUFA4L2轴介导的低氧适应,抑制肺鳞癌的存活。
IF 6.9 1区 医学
Oncogene Pub Date : 2024-08-17 DOI: 10.1038/s41388-024-03129-7
Peng Han, Boxiang Zhang, Yixing Li, Rui Gao, Xinru Li, Hui Ren, Puyu Shi, Aomei Zhao, Jianjun Xue, Aimin Yang, Yiqian Liang
{"title":"MiR-183-5p inhibits lung squamous cell carcinoma survival through disrupting hypoxia adaptation mediated by HIF-1α/NDUFA4L2 axis","authors":"Peng Han,&nbsp;Boxiang Zhang,&nbsp;Yixing Li,&nbsp;Rui Gao,&nbsp;Xinru Li,&nbsp;Hui Ren,&nbsp;Puyu Shi,&nbsp;Aomei Zhao,&nbsp;Jianjun Xue,&nbsp;Aimin Yang,&nbsp;Yiqian Liang","doi":"10.1038/s41388-024-03129-7","DOIUrl":"10.1038/s41388-024-03129-7","url":null,"abstract":"Hypoxia is a common feature of lung squamous cell carcinoma (LUSC), and hypoxia-inducible factor-1 (HIF-1) overexpression is associated with poor clinical outcome in LUSC. NADH dehydrogenase 1 alpha subcomplex subunit 4-like 2 (NDUFA4L2) is a recently identified target of HIF-1, but its roles in LUSC remain unclear. Herein, the expression and regulatory mechanisms of NDUFA4L2 were investigated in LUSC, and the influences on LUSC cell oxidative metabolism and survival of NDUFA4L2 were determined. The potential microRNA targeting to NDUFA4L2 was identified and its roles on LUSC cell were detected. We found that NDUFA4L2 were overexpressed in LUSC tissues, and that NDUFA4L2 expression correlated with shorter overall survival. NDUFA4L2 was regulated by HIF-1α under hypoxia, and NDUFA4L2 decreased mitochondrial reactive oxygen species (mitoROS) production through inhibiting mitochondrial complex I activity in LUSC cells. NDUFA4L2 silencing effectively suppressed LUSC cell growth and enhanced apoptosis by inducing mitoROS accumulation. Additionally, NDUFA4L2 was a target for miR-183-5p, and LUSC patients with high miR-183-5p levels had better prognoses. MiR-183-5p significantly induced mitoROS production and suppressed LUSC survival through negatively regulating NDUFA4L2 in vitro and in vivo. Our results suggested that regulation of NDUFA4L2 by HIF-1α is an important mechanism promoting LUSC progression under hypoxia. NDUFA4L2 inhibition using enforced miR-183-5p expression might be an effective strategy for LUSC treatment.","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":null,"pages":null},"PeriodicalIF":6.9,"publicationDate":"2024-08-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141996240","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
Cooperative genomic lesions in HRAS-mutant cancers predict resistance to farnesyltransferase inhibitors HRAS突变癌症中的合作基因组病变可预测法尼基转移酶抑制剂的耐药性。
IF 6.9 1区 医学
Oncogene Pub Date : 2024-08-16 DOI: 10.1038/s41388-024-03095-0
Aradhya Nigam, Gnana P. Krishnamoorthy, Walid K. Chatila, Katherine Berman, Mahesh Saqcena, Henry Walch, Mandakini Venkatramani, Alan L. Ho, Nikolaus Schultz, James A. Fagin, Brian R. Untch
{"title":"Cooperative genomic lesions in HRAS-mutant cancers predict resistance to farnesyltransferase inhibitors","authors":"Aradhya Nigam,&nbsp;Gnana P. Krishnamoorthy,&nbsp;Walid K. Chatila,&nbsp;Katherine Berman,&nbsp;Mahesh Saqcena,&nbsp;Henry Walch,&nbsp;Mandakini Venkatramani,&nbsp;Alan L. Ho,&nbsp;Nikolaus Schultz,&nbsp;James A. Fagin,&nbsp;Brian R. Untch","doi":"10.1038/s41388-024-03095-0","DOIUrl":"10.1038/s41388-024-03095-0","url":null,"abstract":"In the clinical development of farnesyltransferase inhibitors (FTIs) for HRAS-mutant tumors, responses varied by cancer type. Co-occurring mutations may affect responses. We aimed to uncover cooperative genetic events specific to HRAS-mutant tumors and to study their effect on sensitivity to FTIs. Using targeted sequencing data from the MSK-IMPACT and Dana-Farber Cancer Institute Genomic Evidence Neoplasia Information Exchange databases, we identified comutations that were observed predominantly in HRAS-mutant versus KRAS-mutant or NRAS-mutant cancers. HRAS-mutant cancers had a higher frequency of coaltered mutations (48.8%) in the MAPK, PI3K, or RTK pathway genes, compared with KRAS-mutant (41.4%) and NRAS-mutant (38.4%) cancers (p &lt; 0.05). Class 3 BRAF, NF1, PTEN, and PIK3CA mutations were more prevalent in HRAS-mutant lineages. To study the effects of comutations on sensitivity to FTIs, HrasG13R was transfected into “RASless” (Kraslox/lox/Hras−/−/Nras−/−/RERTert/ert) mouse embryonic fibroblasts (MEFs), which sensitized nontransfected MEFs to tipifarnib. Comutation in the form of Pten or Nf1 deletion and Pik3caH1047R transduction led to resistance to tipifarnib in HrasG13R-transfected MEFs in the presence or absence of KrasWT, whereas BrafG466E transduction led to resistance to tipifarnib only in the presence of KrasWT. Combined treatment with tipifarnib and MEK inhibition sensitized cells to tipifarnib in all settings, including in MEFs with PI3K pathway comutations. HRAS-mutant tumors demonstrate lineage-dependent MAPK or PI3K pathway alterations, which confer resistance to tipifarnib. The combined use of FTIs and MEK inhibition is a promising strategy for HRAS-mutant tumors.","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":null,"pages":null},"PeriodicalIF":6.9,"publicationDate":"2024-08-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141996238","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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信