Pub Date : 2026-01-07DOI: 10.1038/s41388-025-03672-x
Jasper H L T van Genugten, Dean A Fennell, Paul Baas
Mesothelioma is an aggressive cancer that is often characterized by loss of the BRCA1-associated protein 1 (BAP1) tumor suppressor gene. This alteration typically occurs as an early clonal event in mesothelioma development, making it a promising candidate for both diagnostic and therapeutic applications. Functionally, BAP1 regulates gene expression through interactions with Polycomb-group complexes, and it plays roles in various other cellular processes including DNA repair, replication stress, and cell metabolism. While preclinical research has identified multiple potential vulnerabilities in BAP1-deficient tumors-including sensitivity to EZH2-, HDAC-, PARP-, and FGFR-inhibitors-translating these findings to the clinic remains a challenge. In this review, we provide a comprehensive overview of BAP1's molecular functions in mesothelioma, with a focus on their translation into clinical therapeutics for this hard-to-treat malignancy.
{"title":"BAP1-loss in mesothelioma: molecular mechanisms and clinical opportunities.","authors":"Jasper H L T van Genugten, Dean A Fennell, Paul Baas","doi":"10.1038/s41388-025-03672-x","DOIUrl":"https://doi.org/10.1038/s41388-025-03672-x","url":null,"abstract":"<p><p>Mesothelioma is an aggressive cancer that is often characterized by loss of the BRCA1-associated protein 1 (BAP1) tumor suppressor gene. This alteration typically occurs as an early clonal event in mesothelioma development, making it a promising candidate for both diagnostic and therapeutic applications. Functionally, BAP1 regulates gene expression through interactions with Polycomb-group complexes, and it plays roles in various other cellular processes including DNA repair, replication stress, and cell metabolism. While preclinical research has identified multiple potential vulnerabilities in BAP1-deficient tumors-including sensitivity to EZH2-, HDAC-, PARP-, and FGFR-inhibitors-translating these findings to the clinic remains a challenge. In this review, we provide a comprehensive overview of BAP1's molecular functions in mesothelioma, with a focus on their translation into clinical therapeutics for this hard-to-treat malignancy.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145918228","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}
Pub Date : 2026-01-07DOI: 10.1038/s41388-025-03662-z
Wanqing Xie, Qi Chu, Lourdes Brea, Guihua Zeng, Yuan Wang, Xiaodong Lu, Mohan Zheng, Corinne R Ley, Zhiquan Lei, Hongshun Shi, Joshua L Zhu, Lihu Gong, M Cynthia Martin, Xianglin Shi, Galina Gritsina, Arabela A Grigorescu, Hana Chandonnet, Xin Liu, Jonathan C Zhao, Gary E Schiltz, Jindan Yu
Enhancer of Zeste Homolog 2 (EZH2) is the enzymatic subunit of the Polycomb Repressive Complex 2 (PRC2). It catalyzes H3K27 methylation for epigenetic silencing of tumor suppressors and critically drives prostate cancer (PCa) progression. However, inhibitors of EZH2 catalytic function (EZH2i), such as EPZ-6438, showed limited efficacy in PCa. Here, we designed and developed a series of VHL-based proteolysis-targeting chimera (PROTAC) degraders of EZH2 using EPZ-6438 as a ligand and identified PROTAC-6272 as a lead compound. PROTAC-6272 effectively degraded EZH2 and other PRC2 subunits across diverse PCa cell lines. However, PROTAC-6272 and other similar EZH2i-based PROTACs were consistently unable to decrease androgen receptor (AR), a gene that is directly activated by solo EZH2. Mechanistically, EZH2 PROTACs failed to degrade EZH2 coactivators, such as p300, due to their inability to engage EZH2 outside of the PRC2 complex. Nevertheless, PROTAC-6272 exhibited anti-proliferative activities superior to EPZ-6438 in some PCa models, wherein it induced p21 expression and cellular senescence by disrupting a methylation-independent PRC2 function. In summary, while EZH2i-based PROTACs failed to target the PRC2-independent functions of EZH2, they confer added benefits over EPZ-6438 by abolishing a polycomb-dependent but methylation-independent function of EZH2, offering therapeutic advantages in some PCa.
{"title":"EZH2 PROTACs outperform catalytic inhibitors in prostate cancer by targeting a methylation-independent function of PRC2.","authors":"Wanqing Xie, Qi Chu, Lourdes Brea, Guihua Zeng, Yuan Wang, Xiaodong Lu, Mohan Zheng, Corinne R Ley, Zhiquan Lei, Hongshun Shi, Joshua L Zhu, Lihu Gong, M Cynthia Martin, Xianglin Shi, Galina Gritsina, Arabela A Grigorescu, Hana Chandonnet, Xin Liu, Jonathan C Zhao, Gary E Schiltz, Jindan Yu","doi":"10.1038/s41388-025-03662-z","DOIUrl":"https://doi.org/10.1038/s41388-025-03662-z","url":null,"abstract":"<p><p>Enhancer of Zeste Homolog 2 (EZH2) is the enzymatic subunit of the Polycomb Repressive Complex 2 (PRC2). It catalyzes H3K27 methylation for epigenetic silencing of tumor suppressors and critically drives prostate cancer (PCa) progression. However, inhibitors of EZH2 catalytic function (EZH2i), such as EPZ-6438, showed limited efficacy in PCa. Here, we designed and developed a series of VHL-based proteolysis-targeting chimera (PROTAC) degraders of EZH2 using EPZ-6438 as a ligand and identified PROTAC-6272 as a lead compound. PROTAC-6272 effectively degraded EZH2 and other PRC2 subunits across diverse PCa cell lines. However, PROTAC-6272 and other similar EZH2i-based PROTACs were consistently unable to decrease androgen receptor (AR), a gene that is directly activated by solo EZH2. Mechanistically, EZH2 PROTACs failed to degrade EZH2 coactivators, such as p300, due to their inability to engage EZH2 outside of the PRC2 complex. Nevertheless, PROTAC-6272 exhibited anti-proliferative activities superior to EPZ-6438 in some PCa models, wherein it induced p21 expression and cellular senescence by disrupting a methylation-independent PRC2 function. In summary, while EZH2i-based PROTACs failed to target the PRC2-independent functions of EZH2, they confer added benefits over EPZ-6438 by abolishing a polycomb-dependent but methylation-independent function of EZH2, offering therapeutic advantages in some PCa.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145918172","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}
Pub Date : 2026-01-07DOI: 10.1038/s41388-025-03669-6
Wenjing Wang, Qiang He, Tianfei Fan, Yang Xiong, Yimin Xiong, Qinhui Liu, Na Yang, Yining Xu, Yanping Li, Jinhan He
Histone lactylation, a recently discovered epigenetic modification, has been shown to play a critical role in regulating gene expression and cellular functions. However, its involvement in cisplatin (CDDP) resistance in non-small cell lung cancer (NSCLC) remains poorly understood. In this study, we demonstrated that histone lactylation is closely associated with CDDP resistance and correlates with poor prognosis of NSCLC. Mechanistically, H4K12la (histone e 4 lysine 12 lactylation) levels and CEBPB (CCAAT/enhancer-binding protein beta) had a cooperative effect on the regulation of AKR1C2 (Aldo-Keto reductase 1C2). Furthermore, AKR1C2 knockdown activates the mTOR oncogenic signaling pathway. Importantly, genetic manipulation of AKR1C2 or the combination of CDDP with an mTOR inhibitor effectively reverse CDDP resistance in NSCLC/CDDP cells. These findings highlighted the potential of AKR1C2 as a predictive biomarker for patient response to CDDP therapy. Additionally, our study established a novel link between histone lactylation and CDDP resistance, providing new insights into the epigenetic regulation in NSCLC.
{"title":"An H4K12la/CEBPB-AKR1C2 signaling axis modulates the mTOR pathway to regulate cisplatin resistance in lung cancer.","authors":"Wenjing Wang, Qiang He, Tianfei Fan, Yang Xiong, Yimin Xiong, Qinhui Liu, Na Yang, Yining Xu, Yanping Li, Jinhan He","doi":"10.1038/s41388-025-03669-6","DOIUrl":"https://doi.org/10.1038/s41388-025-03669-6","url":null,"abstract":"<p><p>Histone lactylation, a recently discovered epigenetic modification, has been shown to play a critical role in regulating gene expression and cellular functions. However, its involvement in cisplatin (CDDP) resistance in non-small cell lung cancer (NSCLC) remains poorly understood. In this study, we demonstrated that histone lactylation is closely associated with CDDP resistance and correlates with poor prognosis of NSCLC. Mechanistically, H4K12la (histone e 4 lysine 12 lactylation) levels and CEBPB (CCAAT/enhancer-binding protein beta) had a cooperative effect on the regulation of AKR1C2 (Aldo-Keto reductase 1C2). Furthermore, AKR1C2 knockdown activates the mTOR oncogenic signaling pathway. Importantly, genetic manipulation of AKR1C2 or the combination of CDDP with an mTOR inhibitor effectively reverse CDDP resistance in NSCLC/CDDP cells. These findings highlighted the potential of AKR1C2 as a predictive biomarker for patient response to CDDP therapy. Additionally, our study established a novel link between histone lactylation and CDDP resistance, providing new insights into the epigenetic regulation in NSCLC.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2026-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145918241","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}
Pub Date : 2026-01-04DOI: 10.1038/s41388-025-03671-y
Shengpan Wu, Baojun Wang, Hongzhao Li, Hanfeng Wang, Songliang Du, Xing Huang, Yang Fan, Yu Gao, Liangyou Gu, Qingbo Huang, Jianjun Chen, Xu Zhang, Yan Huang, Xin Ma
While Stimulator-of-interferon genes (STING) is an innate immune adapter crucial for sensing cytosolic DNA and modulating immune microenvironment, its tumor-promoting role in tumor survival and immune evasion remains largely unknown. Here we reported that renal cancer cells are exceptionally dependent on STING for survival and evading immunosurveillance via suppressing ER stress-mediated pyroptosis. We found that STING is significantly amplified and upregulated in clear cell renal cell carcinoma (ccRCC), and its elevated expression is associated with worse clinical outcomes. Mechanically, STING depletion in RCC cells specifically triggers activation of the PERK/eIF2α/ATF4/CHOP pathway and activates cleavage of Caspase-8, thereby inducing GSDMD-mediated pyroptosis, which is independent of the innate immune pathway of STING. Moreover, animal study results revealed that STING depletion promoted infiltration of CD4+ and CD8+ T cells, consequently boosting robust antitumor immunity via pyroptosis-induced inflammation. From the perspective of targeted therapy, we found that Compound SP23, a PROTAC STING degrader, demonstrated comparable efficacy to STING depletion both in vitro and in vivo for treatment of ccRCC. These findings collectively unveiled an unforeseen function of STING in regulating GSDMD-dependent pyroptosis, thus regulating immune response in RCC. Consequently, pharmacological degradation of STING by SP23 may become an attractive strategy for treatment of advanced RCC.
{"title":"Targeting STING elicits GSDMD-dependent pyroptosis and boosts anti-tumor immunity in renal cell carcinoma.","authors":"Shengpan Wu, Baojun Wang, Hongzhao Li, Hanfeng Wang, Songliang Du, Xing Huang, Yang Fan, Yu Gao, Liangyou Gu, Qingbo Huang, Jianjun Chen, Xu Zhang, Yan Huang, Xin Ma","doi":"10.1038/s41388-025-03671-y","DOIUrl":"https://doi.org/10.1038/s41388-025-03671-y","url":null,"abstract":"<p><p>While Stimulator-of-interferon genes (STING) is an innate immune adapter crucial for sensing cytosolic DNA and modulating immune microenvironment, its tumor-promoting role in tumor survival and immune evasion remains largely unknown. Here we reported that renal cancer cells are exceptionally dependent on STING for survival and evading immunosurveillance via suppressing ER stress-mediated pyroptosis. We found that STING is significantly amplified and upregulated in clear cell renal cell carcinoma (ccRCC), and its elevated expression is associated with worse clinical outcomes. Mechanically, STING depletion in RCC cells specifically triggers activation of the PERK/eIF2α/ATF4/CHOP pathway and activates cleavage of Caspase-8, thereby inducing GSDMD-mediated pyroptosis, which is independent of the innate immune pathway of STING. Moreover, animal study results revealed that STING depletion promoted infiltration of CD4<sup>+</sup> and CD8<sup>+</sup> T cells, consequently boosting robust antitumor immunity via pyroptosis-induced inflammation. From the perspective of targeted therapy, we found that Compound SP23, a PROTAC STING degrader, demonstrated comparable efficacy to STING depletion both in vitro and in vivo for treatment of ccRCC. These findings collectively unveiled an unforeseen function of STING in regulating GSDMD-dependent pyroptosis, thus regulating immune response in RCC. Consequently, pharmacological degradation of STING by SP23 may become an attractive strategy for treatment of advanced RCC.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2026-01-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145896709","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}
Pub Date : 2026-01-03DOI: 10.1038/s41388-025-03640-5
Marwa Taya, Daniel Fishman, Fahim Kanani, Lotem Zinger, Keren Merenbakh-Lamin, Shaked Elfasi Sosner, Anil Khushalrao Shendge, Anat Klein Goldberg, Uri Wolf, Galit Winkler, Eric Shifrut, Shmuel Cohen, Ido Wolf, Israel Sekler, Tami Rubinek
Activating mutations in the ligand-binding domain of the estrogen receptor (ER)-encoding (ESR1) gene are present in up to 40% of metastatic breast cancer (BC) patients and are strongly associated with a high risk of liver metastasis (LM) formation. Using the MCF-7 BC model, we investigated whether the increased hepatic tropism of ESR1-mutated BC cells is driven by their metabolic adaptation to the liver microenvironment. Indeed, metabolomic analysis revealed elevated metabolites related to the urea cycle (UC) in LM-forming ESR1-mutated cells compared to wild-type (WT) ER-expressing cells, which failed to generate LM. The subsequent proteomic, western blotting, and qPCR analyses demonstrated a dramatic upregulation of the UC constituent, the mitochondrial ornithine/citrulline transporter SLC25A15, in liver-predilected ESR1-mutated cells relative to their WT counterpart cells. Unlike WT cells, ESR1-mutated cells readily formed spheroids and exhibited enhanced migration in liver mimicking hepatocyte-conditioned media. In addition, we employed a novel ex vivo approach where ESR1 mutated cells were seeded onto colonized fresh liver tissue-which was abolished by SLC25A15 knockout. Moreover, SLC25A15 knockout robustly reduced the ability of ESR1-mutated cells to establish LM in vivo. These findings highlight SLC25A15-mediated dysregulation of the UC as a critical driver of BC hepatic metastasis and identify SLC25A15 as a potential therapeutic target for disrupting metastatic spread of BC to the liver.
{"title":"The role of the SLC25A15 transporter in the formation of liver metastasis in ESR1-mutated breast cancer.","authors":"Marwa Taya, Daniel Fishman, Fahim Kanani, Lotem Zinger, Keren Merenbakh-Lamin, Shaked Elfasi Sosner, Anil Khushalrao Shendge, Anat Klein Goldberg, Uri Wolf, Galit Winkler, Eric Shifrut, Shmuel Cohen, Ido Wolf, Israel Sekler, Tami Rubinek","doi":"10.1038/s41388-025-03640-5","DOIUrl":"https://doi.org/10.1038/s41388-025-03640-5","url":null,"abstract":"<p><p>Activating mutations in the ligand-binding domain of the estrogen receptor (ER)-encoding (ESR1) gene are present in up to 40% of metastatic breast cancer (BC) patients and are strongly associated with a high risk of liver metastasis (LM) formation. Using the MCF-7 BC model, we investigated whether the increased hepatic tropism of ESR1-mutated BC cells is driven by their metabolic adaptation to the liver microenvironment. Indeed, metabolomic analysis revealed elevated metabolites related to the urea cycle (UC) in LM-forming ESR1-mutated cells compared to wild-type (WT) ER-expressing cells, which failed to generate LM. The subsequent proteomic, western blotting, and qPCR analyses demonstrated a dramatic upregulation of the UC constituent, the mitochondrial ornithine/citrulline transporter SLC25A15, in liver-predilected ESR1-mutated cells relative to their WT counterpart cells. Unlike WT cells, ESR1-mutated cells readily formed spheroids and exhibited enhanced migration in liver mimicking hepatocyte-conditioned media. In addition, we employed a novel ex vivo approach where ESR1 mutated cells were seeded onto colonized fresh liver tissue-which was abolished by SLC25A15 knockout. Moreover, SLC25A15 knockout robustly reduced the ability of ESR1-mutated cells to establish LM in vivo. These findings highlight SLC25A15-mediated dysregulation of the UC as a critical driver of BC hepatic metastasis and identify SLC25A15 as a potential therapeutic target for disrupting metastatic spread of BC to the liver.</p>","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2026-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145896755","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}
Indolent lung cancer is characterized by slow progression, and the patient may die of other causes before it becomes symptomatic. The increasing usage of lung cancer screening technologies has increased the detection of early-stage cancers, improved survival, and reduced mortality, but it also brings overdiagnosis or overtreatment for indolent lung cancer patients. In this study, we performed high-throughput whole-exome sequencing in lung adenocarcinoma (LUAD) patients with a tumor doubling time of more than 600 days (the indolent group) and another group with a tumor doubling time of less than 30 days (the aggressive group). We identified a novel germline variation in PDZRN4, rs74955204 (p.G121E, c.362 G > A) in the indolent group. The rs74955204 variant resulted in upregulated PDZRN4 expression and was positively correlated with the survival of LUAD patients. We confirmed that overexpression of PDZRN4 inhibits the ubiquitylation of HIF-1A mediated by TRIM28, thus activating HIF-1A-IGFBP3 signaling to promote apoptosis, and inducing NDRG1 to inhibit EGFR-AKT signaling, restricting lung cancer cell growth in vitro and in vivo. With the EGFRL858R-driven lung cancer mouse model as an example, we found that PDZRN4 knockout promoted, whereas PDZRN4 overexpression suppressed LUAD growth in vivo. In addition, recombinant PDZRN4 or screened small molecules retinoic acid showed potent inhibitory effects on EGFR-driven LUAD growth and amelioration of osimertinib resistance. Overall, our findings suggest that targeted PDZRN4 is a potential therapeutic strategy for treating LUAD. Examination of PDZRN4 rs74955204 variant may be an effective approach for the early detection of indolent lung cancer and to avoid overdiagnosis.
惰性肺癌的特点是进展缓慢,患者可能在出现症状之前死于其他原因。肺癌筛查技术的日益普及,增加了早期癌症的发现,提高了生存率,降低了死亡率,但也给惰性肺癌患者带来了过度诊断或过度治疗。在本研究中,我们对肿瘤加倍时间超过600天的肺腺癌(LUAD)患者(惰性组)和肿瘤加倍时间小于30天的肺腺癌(LUAD)患者(侵袭组)进行了高通量全外显子组测序。我们在惰性组中发现了PDZRN4, rs74955204 (p.G121E, c.362 G > a)的一种新的种系变异。rs74955204变异导致PDZRN4表达上调,与LUAD患者的生存呈正相关。我们在体外和体内证实,PDZRN4过表达可抑制TRIM28介导的HIF-1A泛素化,从而激活HIF-1A- igfbp3信号通路促进细胞凋亡,诱导NDRG1抑制EGFR-AKT信号通路,限制肺癌细胞生长。以egfrl858r驱动的肺癌小鼠模型为例,我们发现PDZRN4敲除促进体内LUAD的生长,而PDZRN4过表达抑制体内LUAD的生长。此外,重组PDZRN4或筛选的小分子维甲酸对egfr驱动的LUAD生长和改善奥西替尼耐药性有明显的抑制作用。总之,我们的研究结果表明,靶向PDZRN4是治疗LUAD的潜在治疗策略。检测PDZRN4 rs74955204变异可能是早期发现惰性肺癌,避免过度诊断的有效途径。摘要PDZRN4的种系变异rs74955204 (p.G121E, c.g 362 G > A)显著上调PDZRN4的表达,导致LUAD的惰性生长。过表达的PDZRN4可抑制TRIM28对HIF-1A的泛素化,激活HIF-1A- igfbp3信号通路促进细胞凋亡,诱导NDRG1抑制EGFR-AKT信号通路,从而限制肺癌细胞生长。
{"title":"PDZRN4 suppresses lung adenocarcinoma progression via inhibiting ubiquitin-mediated HIF-1A degradation","authors":"Honghao Li, Junfeng Wu, Hua Dai, Jiaming Li, Xiaoyan Bai, Yueli Sun, Jiaxin Lin, Yangsi Li, Peng Li, Lingchao Shi, Zhen Wang, Haiyan Tu, Ee Ke","doi":"10.1038/s41388-025-03670-z","DOIUrl":"10.1038/s41388-025-03670-z","url":null,"abstract":"Indolent lung cancer is characterized by slow progression, and the patient may die of other causes before it becomes symptomatic. The increasing usage of lung cancer screening technologies has increased the detection of early-stage cancers, improved survival, and reduced mortality, but it also brings overdiagnosis or overtreatment for indolent lung cancer patients. In this study, we performed high-throughput whole-exome sequencing in lung adenocarcinoma (LUAD) patients with a tumor doubling time of more than 600 days (the indolent group) and another group with a tumor doubling time of less than 30 days (the aggressive group). We identified a novel germline variation in PDZRN4, rs74955204 (p.G121E, c.362 G > A) in the indolent group. The rs74955204 variant resulted in upregulated PDZRN4 expression and was positively correlated with the survival of LUAD patients. We confirmed that overexpression of PDZRN4 inhibits the ubiquitylation of HIF-1A mediated by TRIM28, thus activating HIF-1A-IGFBP3 signaling to promote apoptosis, and inducing NDRG1 to inhibit EGFR-AKT signaling, restricting lung cancer cell growth in vitro and in vivo. With the EGFRL858R-driven lung cancer mouse model as an example, we found that PDZRN4 knockout promoted, whereas PDZRN4 overexpression suppressed LUAD growth in vivo. In addition, recombinant PDZRN4 or screened small molecules retinoic acid showed potent inhibitory effects on EGFR-driven LUAD growth and amelioration of osimertinib resistance. Overall, our findings suggest that targeted PDZRN4 is a potential therapeutic strategy for treating LUAD. Examination of PDZRN4 rs74955204 variant may be an effective approach for the early detection of indolent lung cancer and to avoid overdiagnosis.","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":"45 4","pages":"577-590"},"PeriodicalIF":7.3,"publicationDate":"2025-12-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145857129","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}
Pub Date : 2025-12-27DOI: 10.1038/s41388-025-03661-0
Haoran Wang, Lichen Ge, Jianing Li, Ke Zhong, Shanzhi Li, Ningjing Ma, Lijun Tao, Jiawang Zhou, Zhaotong Wang, Xing Chang, Yunqing Lu, Yalan Rui, Guoyou Xie, Weifeng Yang, Zuanzong Xu, Abdulaziz Ahmed A. Saad, Xiansong Wang, Zhuojia Chen, Wanglin Li, Cheng Yi, Hongsheng Wang, Kun Zhang
RNA modification recognition proteins are crucial in cancer development and progression. Among all RNA modification-related proteins (RMRPs), Weighted Gene Co-expression Network Analysis (WGCNA), combined with comprehensive bioinformatic analysis, suggests that NAT10—the sole known writer of N4-acetylcytidine (ac4C)—is a critical regulatory protein in colorectal cancer (CRC) progression. NAT10 facilitates the malignancy phenotypes and DNA damage repair in CRC cells via its ac4C transferase activity and regulation of PPAN. Specifically, NAT10 enhances the translation efficiency of PPAN via acetylation at the C744 and C747 sites. In addition, NAT10 promotes the translation of ac4C-modified MYC mRNA. MYC protein then enhances PPAN transcription through binding to the PPAN promoter. The newly identified ac4C reader protein MYBBP1A mediates NAT10-induced translation of both PPAN and MYC. We further found that VDR binds to the NAT10 promoter to activate its transcription, resulting in the high expression of NAT10 in CRC. Xenograft studies and clinical data confirmed the role of the NAT10-PPAN axis in promoting CRC development and DNA damage repair. Collectively, our study reveals the role and underlying mechanism of mRNA ac4C modification in CRC progression, providing critical potential targets for CRC drug development.
{"title":"NAT10 triggers colorectal cancer progression via promoting PPAN-regulated DNA damage repair","authors":"Haoran Wang, Lichen Ge, Jianing Li, Ke Zhong, Shanzhi Li, Ningjing Ma, Lijun Tao, Jiawang Zhou, Zhaotong Wang, Xing Chang, Yunqing Lu, Yalan Rui, Guoyou Xie, Weifeng Yang, Zuanzong Xu, Abdulaziz Ahmed A. Saad, Xiansong Wang, Zhuojia Chen, Wanglin Li, Cheng Yi, Hongsheng Wang, Kun Zhang","doi":"10.1038/s41388-025-03661-0","DOIUrl":"10.1038/s41388-025-03661-0","url":null,"abstract":"RNA modification recognition proteins are crucial in cancer development and progression. Among all RNA modification-related proteins (RMRPs), Weighted Gene Co-expression Network Analysis (WGCNA), combined with comprehensive bioinformatic analysis, suggests that NAT10—the sole known writer of N4-acetylcytidine (ac4C)—is a critical regulatory protein in colorectal cancer (CRC) progression. NAT10 facilitates the malignancy phenotypes and DNA damage repair in CRC cells via its ac4C transferase activity and regulation of PPAN. Specifically, NAT10 enhances the translation efficiency of PPAN via acetylation at the C744 and C747 sites. In addition, NAT10 promotes the translation of ac4C-modified MYC mRNA. MYC protein then enhances PPAN transcription through binding to the PPAN promoter. The newly identified ac4C reader protein MYBBP1A mediates NAT10-induced translation of both PPAN and MYC. We further found that VDR binds to the NAT10 promoter to activate its transcription, resulting in the high expression of NAT10 in CRC. Xenograft studies and clinical data confirmed the role of the NAT10-PPAN axis in promoting CRC development and DNA damage repair. Collectively, our study reveals the role and underlying mechanism of mRNA ac4C modification in CRC progression, providing critical potential targets for CRC drug development.","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":"45 4","pages":"558-576"},"PeriodicalIF":7.3,"publicationDate":"2025-12-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145846820","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}
Pub Date : 2025-12-20DOI: 10.1038/s41388-025-03664-x
Yikang Wang, Chang Yuan, Zhichao Liu, Ning Ma, Cong Qi, Boyao Yu, Haizhen Jin, Chenyang Tian, Chunguang Li, Zhigang Li
Lymph node metastasis (LNM) is a major determinant of poor prognosis in esophageal squamous cell carcinoma (ESCC), with patients remaining at high risk of postoperative recurrence in the absence of reliable predictors or effective interventions. To address this unmet clinical need, we perform proteomic and bioinformatic analyses and identify lysosome-associated membrane protein 1 (LAMP1) as a critical driver of LNM in ESCC. LAMP1 is markedly overexpressed in primary tumors that with LNM and correlates with worse survival outcomes across independent cohorts. Gain- and loss-of-function experiments demonstrate that LAMP1 promotes lymphangiogenesis and lymphatic metastasis both in vitro and in vivo. Mechanistically, LAMP1 interacts with DNA-PKcs, mediates AKT activation, leading to VEGFC upregulation and secretion, thereby driving lymphangiogenesis and metastasis. Importantly, treatment with the LAMP1 inhibitor Parishin C significantly suppresses LNM in preclinical ESCC models. These findings uncover a previously unrecognized LAMP1/DNA-PKcs/AKT/VEGFC signaling axis that promotes ESCC progression and highlight LAMP1 as a promising biomarker and therapeutic target for preventing LNM and improving patient outcomes.
{"title":"LAMP1 enhances DNA-PKcs-mediated AKT phosphorylation and VEGFC secretion to promote lymph node metastasis in esophageal squamous cell carcinoma","authors":"Yikang Wang, Chang Yuan, Zhichao Liu, Ning Ma, Cong Qi, Boyao Yu, Haizhen Jin, Chenyang Tian, Chunguang Li, Zhigang Li","doi":"10.1038/s41388-025-03664-x","DOIUrl":"10.1038/s41388-025-03664-x","url":null,"abstract":"Lymph node metastasis (LNM) is a major determinant of poor prognosis in esophageal squamous cell carcinoma (ESCC), with patients remaining at high risk of postoperative recurrence in the absence of reliable predictors or effective interventions. To address this unmet clinical need, we perform proteomic and bioinformatic analyses and identify lysosome-associated membrane protein 1 (LAMP1) as a critical driver of LNM in ESCC. LAMP1 is markedly overexpressed in primary tumors that with LNM and correlates with worse survival outcomes across independent cohorts. Gain- and loss-of-function experiments demonstrate that LAMP1 promotes lymphangiogenesis and lymphatic metastasis both in vitro and in vivo. Mechanistically, LAMP1 interacts with DNA-PKcs, mediates AKT activation, leading to VEGFC upregulation and secretion, thereby driving lymphangiogenesis and metastasis. Importantly, treatment with the LAMP1 inhibitor Parishin C significantly suppresses LNM in preclinical ESCC models. These findings uncover a previously unrecognized LAMP1/DNA-PKcs/AKT/VEGFC signaling axis that promotes ESCC progression and highlight LAMP1 as a promising biomarker and therapeutic target for preventing LNM and improving patient outcomes.","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":"45 4","pages":"534-548"},"PeriodicalIF":7.3,"publicationDate":"2025-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145800090","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}
Pub Date : 2025-12-20DOI: 10.1038/s41388-025-03667-8
Charlotte Andrieu, Jojanneke Stoof, Dalal AlSultan, Laura Ivers, Jose Javier Berenguer Pina, Darko Skrobo, Jo Ballot, Debbie O’Reilly, Denis M. Collins, Alex J. Eustace, Cecily Quinn, Janice M. Walshe, Giuseppe Gullo, Naomi Walsh, John Crown
Metastatic breast cancer (MBC) is generally an incurable neoplasm. A small cohort of patients with HER2-positive MBC, however, achieve such prolonged remission without relapse following anti-HER2 therapy and chemotherapy, that it is speculated they might be cured. The genomes of these patients might provide insights into the underlying mechanisms for their successful treatment. Here, a total of 243 HER2-positive patients diagnosed with MBC between 2000 and 2015 were studied. Of these, 29 patients were identified as exceptional responders (ExR) with an overall survival (OS) > 60 months and no evidence of relapse, 54 patients with an OS > 60 months but who relapsed or developed progressive disease were defined as exceptional survivors (ExS), and 160 patients with an OS < 60 months were identified as short-term responders (STR). Whole-Genome Sequencing and centromere copy number (CCN) analysis was performed on 27 patients (12 ExR; 4 ExS; 11 STR). A significant amplification was observed in the centromeric regions of ExR, exhibiting higher CCN compared to the ExS and STR. Digital PCR validation of chromosome 4 centromere region D4Z1 copy number was not associated with ExR OS. Our results suggest that the amplification of centromere regions are associated with very prolonged remission and survival in patients with HER2-positive MBC.
{"title":"Chromosome centromere copy number amplification associated with exceptional response in HER2-positive metastatic breast cancer patients","authors":"Charlotte Andrieu, Jojanneke Stoof, Dalal AlSultan, Laura Ivers, Jose Javier Berenguer Pina, Darko Skrobo, Jo Ballot, Debbie O’Reilly, Denis M. Collins, Alex J. Eustace, Cecily Quinn, Janice M. Walshe, Giuseppe Gullo, Naomi Walsh, John Crown","doi":"10.1038/s41388-025-03667-8","DOIUrl":"10.1038/s41388-025-03667-8","url":null,"abstract":"Metastatic breast cancer (MBC) is generally an incurable neoplasm. A small cohort of patients with HER2-positive MBC, however, achieve such prolonged remission without relapse following anti-HER2 therapy and chemotherapy, that it is speculated they might be cured. The genomes of these patients might provide insights into the underlying mechanisms for their successful treatment. Here, a total of 243 HER2-positive patients diagnosed with MBC between 2000 and 2015 were studied. Of these, 29 patients were identified as exceptional responders (ExR) with an overall survival (OS) > 60 months and no evidence of relapse, 54 patients with an OS > 60 months but who relapsed or developed progressive disease were defined as exceptional survivors (ExS), and 160 patients with an OS < 60 months were identified as short-term responders (STR). Whole-Genome Sequencing and centromere copy number (CCN) analysis was performed on 27 patients (12 ExR; 4 ExS; 11 STR). A significant amplification was observed in the centromeric regions of ExR, exhibiting higher CCN compared to the ExS and STR. Digital PCR validation of chromosome 4 centromere region D4Z1 copy number was not associated with ExR OS. Our results suggest that the amplification of centromere regions are associated with very prolonged remission and survival in patients with HER2-positive MBC.","PeriodicalId":19524,"journal":{"name":"Oncogene","volume":"45 4","pages":"549-557"},"PeriodicalIF":7.3,"publicationDate":"2025-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.nature.comhttps://www.nature.com/articles/s41388-025-03667-8.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145800087","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}