Cellular OncologyPub Date : 2024-02-01Epub Date: 2023-08-23DOI: 10.1007/s13402-023-00861-2
Kai Jiang, Lili Zhu, Huizhen Huang, Liu Zheng, Zhuqing Wang, Xiaonan Kang
{"title":"Lactate score classification of hepatocellular carcinoma helps identify patients with tumors that respond to immune checkpoint blockade therapy.","authors":"Kai Jiang, Lili Zhu, Huizhen Huang, Liu Zheng, Zhuqing Wang, Xiaonan Kang","doi":"10.1007/s13402-023-00861-2","DOIUrl":"10.1007/s13402-023-00861-2","url":null,"abstract":"<p><strong>Purpose: </strong>Hepatocellular carcinoma (HCC) responds poorly to immunotherapy, and the durable response rate is 10-20%. Here, we aim to characterize HCC classifications based on lactate genes to identify patients who may benefit from immunotherapy.</p><p><strong>Methods: </strong>Lactate-related genes were applied for HCC classification in the current study, and lactate Cluster 1 (LC1) and lactate Cluster 2 (LC2) were defined. Differential genes from LC1 and LC2 helped define the following lactate phenotype clusters: lactate phenotype Cluster 1 (LPC1), lactate phenotype Cluster 2 (LPC2) and lactate phenotype Cluster 3 (LPC3). Based on the cluster annotation, the lactate score was defined and analyzed to evaluate the immunotherapy response.</p><p><strong>Results: </strong>All the classified clusters were analyzed, and they showed different immune signatures. The survival rate of LPC3 was higher than that of LPC2 (LPC3 vs. LPC2, P = 0.027) and LPC1 (LPC3 vs. LPC1, P = 0.027). Then, the lactate score was annotated and confirmed to be effective in predicting responses to immune checkpoint blockade therapy.</p><p><strong>Conclusion: </strong>In the current study, we developed a classification system for HCC and defined the lactate score, which was validated to be partially effective in estimating responses among tumor patients.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10899304/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10414923","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cellular OncologyPub Date : 2023-12-01Epub Date: 2023-08-30DOI: 10.1007/s13402-023-00844-3
Yu'e Liu, Shu Zhao, Yi Chen, Wencong Ma, Shiping Lu, Le He, Jie Chen, Xi Chen, Xiaoling Zhang, Yufeng Shi, Xuan Jiang, Kaijun Zhao
{"title":"Vimentin promotes glioma progression and maintains glioma cell resistance to oxidative phosphorylation inhibition.","authors":"Yu'e Liu, Shu Zhao, Yi Chen, Wencong Ma, Shiping Lu, Le He, Jie Chen, Xi Chen, Xiaoling Zhang, Yufeng Shi, Xuan Jiang, Kaijun Zhao","doi":"10.1007/s13402-023-00844-3","DOIUrl":"10.1007/s13402-023-00844-3","url":null,"abstract":"<p><strong>Purpose: </strong>Glioma has been demonstrated as one of the most malignant intracranial tumors and currently there is no effective treatment. Based on our previous RNA-sequencing data for oxidative phosphorylation (OXPHOS)-inhibition resistant and OXPHOS-inhibition sensitive cancer cells, we found that vimentin (VIM) is highly expressed in the OXPHOS-inhibition resistant cancer cells, especially in glioma cancer cells. Further study of VIM in the literature indicates that it plays important roles in cancer progression, immunotherapy suppression, cancer stemness and drug resistance. However, its role in glioma remains elusive. This study aims to decipher the role of VIM in glioma, especially its role in OXPHOS-inhibition sensitivity, which may provide a promising therapeutic target for glioma treatment.</p><p><strong>Methods: </strong>The expression of VIM in glioma and the normal tissue has been obtained from The Cancer Genome Atlas (TCGA) database, and further validated in Human Protein Atlas (HPA) and Chinese Glioma Genome Atlas (CGGA). And the single-cell sequencing data was obtained from TISCH2. The immune infiltration was calculated via Tumor Immune Estimation Resource (TIMER), Estimation of Stromal and Immune Cells in Malignant Tumors using Expression Data (ESTIMATE) and ssGSEA, and the Immunophenoscore (IPS) was calculated via R package. The differentiated expressed genes were analyzed including GO/KEGG and Gene Set Enrichment Analysis (GSEA) between the VIM-high and -low groups. The methylation of VIM was checked at the EWAS and Methsurv. The correlation between VIM expression and cancer stemness was obtained from SangerBox. We also employed DepMap data and verified the role of VIM by knocking down it in VIM-high glioma cell and over-expressing it in VIM-low glioma cells to check the cell viability.</p><p><strong>Results: </strong>Vim is highly expressed in the glioma patients compared to normal samples and its high expression negatively correlates with patients' survival. The DNA methylation in VIM promoters in glioma patients is lower than that in the normal samples. High VIM expression positively correlates with the immune infiltration and tumor progression. Furthermore, Vim is expressed high in the OXPHOS-inhibition glioma cancer cells and low in the OXPHOS-inhibition sensitive ones and its expression maintains the OXPHOS-inhibition resistance.</p><p><strong>Conclusions: </strong>In conclusion, we comprehensively deciphered the role of VIM in the progression of glioma and its clinical outcomes. Thus provide new insights into targeting VIM in glioma cancer immunotherapy in combination with the current treatment.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2023-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10111042","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"miR-4739 promotes epithelial-mesenchymal transition and angiogenesis in \"driver gene-negative\" non-small cell lung cancer via activating the Wnt/β-catenin signaling.","authors":"Wenjian Cen, Qin Yan, Wenpeng Zhou, Minjie Mao, Qitao Huang, Yaobin Lin, Neng Jiang","doi":"10.1007/s13402-023-00848-z","DOIUrl":"10.1007/s13402-023-00848-z","url":null,"abstract":"<p><strong>Purpose: </strong>\"Driver gene-negative\" non-small cell lung cancer (NSCLC) currently has no approved targeted drug, due to the lack of common actionable driver molecules. Even though miRNAs play crucial roles in various malignancies, their roles in \"driver gene-negative\" NSCLC keep unclear.</p><p><strong>Methods: </strong>miRNA expression microarrays were utilized to screen miRNAs associated with \"driver gene-negative\" NSCLC malignant progression. Quantitative real-time PCR (RT-qPCR) and in situ hybridization (ISH) were employed to validate the expression of miR-4739, and its correlation with clinicopathological characteristics was analyzed in tumor specimens using univariate and multivariate analyses. The biological functions and underlying mechanisms of miR-4739 were investigated both in vitro and in vivo.</p><p><strong>Results: </strong>our research demonstrated, for the first time, that miR-4739 was substantially increased in \"driver gene-negative\" NSCLC tumor tissues and cell lines, and overexpression of miR-4739 was related to clinical staging, metastasis, and unfavorable outcomes. Functional experiments discovered that miR-4739 dramatically enhanced tumor cell proliferation, migration, and metastasis by promoting the epithelial-to-mesenchymal transition (EMT). Meanwhile, miR-4739 can be transported from cancer cells to the site of vascular epithelial cells through exosomes, consequently facilitating the proliferation and migration of vascular epithelial cells and inducing angiogenesis. Mechanistically, miR-4739 can activate Wnt/β-catenin signaling both in tumor cells and vascular epithelial cells by targeting Wnt/β-catenin signaling antagonists APC2 and DKK3, respectively.</p><p><strong>Conclusion: </strong>Our work identifies a valuable oncogene, miR-4739, that accelerates malignant progression in \"driver gene-negative\" NSCLC and serves as a potential therapeutic target for this group of tumors.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2023-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9873703","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cellular OncologyPub Date : 2023-12-01Epub Date: 2023-06-24DOI: 10.1007/s13402-023-00835-4
Huan Deng, Jingwang Gao, Bo Cao, Ziyu Qiu, Tian Li, Ruiyang Zhao, Hanghang Li, Bo Wei
{"title":"LncRNA CCAT2 promotes malignant progression of metastatic gastric cancer through regulating CD44 alternative splicing.","authors":"Huan Deng, Jingwang Gao, Bo Cao, Ziyu Qiu, Tian Li, Ruiyang Zhao, Hanghang Li, Bo Wei","doi":"10.1007/s13402-023-00835-4","DOIUrl":"10.1007/s13402-023-00835-4","url":null,"abstract":"<p><strong>Objective: </strong>Gastric cancer (GC) is one of the most malignant tumors worldwide. Thus, it is necessary to explore the underlying mechanisms of GC progression and develop novel therapeutic regimens. Long non-coding RNAs (lncRNAs) have been demonstrated to be abnormally expressed and regulate the malignant behaviors of cancer cells. Our previous research demonstrated that lncRNA colon cancer-associated transcript 2 (CCAT2) has potential value for GC diagnosis and discrimination. However, the functional mechanisms of lncRNA CCAT2 in GC development remain to be explored.</p><p><strong>Methods: </strong>GC and normal adjacent tissues were collected to detect the expression of lncRNA CCAT2, ESRP1 and CD44 in clinical specimens and their clinical significance for GC patients. Cell counting kit-8, wound healing and transwell assays were conducted to investigate the malignant behaviors in vitro. The generation of nude mouse xenografts by subcutaneous, intraperitoneal and tail vein injection was performed to examine GC growth and metastasis in vivo. Co-immunoprecipitation, RNA-binding protein pull-down assay and fluorescence in situ hybridization were performed to reveal the binding relationships between ESRP1 and CD44.</p><p><strong>Results: </strong>In the present study, lncRNA CCAT2 was overexpressed in GC tissues compared to adjacent normal tissues and correlated with short survival time of patients. lncRNA CCAT2 promoted the proliferation, migration and invasion of GC cells. Its overexpression modulates alternative splicing of Cluster of differentiation 44 (CD44) variants and facilitates the conversion from the standard form to variable CD44 isoform 6 (CD44v6). Mechanistically, lncRNA CCAT2 upregulated CD44v6 expression by binding to epithelial splicing regulatory protein 1 (ESRP1), which subsequently mediates CD44 alternative splicing. The oncogenic role of the lncRNA CCAT2/ESRP1/CD44 axis in the promotion of malignant behaviors was verified by both in vivo and in vitro experiments.</p><p><strong>Conclusions: </strong>Our findings identified a novel mechanism by which lncRNA CCAT2, as a type of protein-binding RNA, regulates alternative splicing of CD44 and promotes GC progression. This axis may become an effective target for clinical diagnosis and treatment.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2023-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10054524","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Modulation of miR-146b by N6-methyladenosine modification remodels tumor-associated macrophages and enhances anti-PD-1 therapy in colorectal cancer.","authors":"Shuying He, Wen Song, Shudan Cui, Jiating Li, Yonghong Jiang, Xueqing Chen, Liang Peng","doi":"10.1007/s13402-023-00839-0","DOIUrl":"10.1007/s13402-023-00839-0","url":null,"abstract":"<p><strong>Purpose: </strong>MicroRNA-146b (miR-146b) alleviates experimental colitis in mice by mediating macrophage polarization and the release of inflammatory factors. Our goals were to evaluate the antitumor efficacy of miR-146b in colorectal cancer (CRC) and to investigate the underlying mechanisms.</p><p><strong>Methods: </strong>We used murine models of CRC to evaluate whether miR-146b influenced the progression of tumors independent of tumor-associated macrophages (TAMs). RNA immunoprecipitation, N6-methyladenosine (m<sup>6</sup>A) RNA immunoprecipitation and in vitro pri-miRNA processing assays were conducted to examine whether m<sup>6</sup>A mediates the maturation of pri-miR-146b/miR-146b. In a series of in vitro and in vivo experiments, we further defined the molecular mechanisms of methyltransferase-like 3 (METTL3)/miR-146b-mediated antitumor immunity and its efficacy in combination with anti-PD-1 immunotherapy.</p><p><strong>Results: </strong>We found that miR-146b deletion supported tumor progression by increasing the number of alternatively activated (M2) TAMs. Mechanistically, the m<sup>6</sup>A-related \"writer\" protein METTL3 and \"reader\" protein HNRNPA2B1 controlled miR-146b maturation by regulating the m<sup>6</sup>A modification region of pri-miR-146b. Furthermore, miR-146b deletion promoted the polarization of M2-TAMs by enhancing phosphoinositide 3-kinase (PI3K)/AKT signaling, and this effect was mediated by the class IA PI3K catalytic subunit p110β, which reduced T cell infiltration, aggravated immunosuppression and ultimately promoted tumor progression. METTL3 knockdown or miR-146b deletion induced programmed death ligand 1 (PD-L1) production via the p110β/PI3K/AKT pathway in TAMs and consequently augmented the antitumor activity of anti-PD-1 immunotherapy.</p><p><strong>Conclusions: </strong>The maturation of pri-miR-146b is m<sup>6</sup>A-dependent, and miR-146b deletion-mediated TAM differentiation promotes the development of CRC by activating the PI3K/AKT pathway, which induces upregulation of PD-L1 expression, inhibits T cell infiltration into the TME and enhances the antitumor activity of anti-PD-1 immunotherapy. The findings reveal that targeting miR-146b can serve as an adjuvant to anti-PD-1 immunotherapy.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2023-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10697876/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10128479","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cellular OncologyPub Date : 2023-12-01Epub Date: 2023-06-28DOI: 10.1007/s13402-023-00838-1
Chuanbo Xie, Shuqing Wang, Chi Guo, Yuying Liu, Musheng Zeng
{"title":"Apo10 and TKTL1 in blood macrophages as biomarkers for differentiating lung cancer from benign lung lesions: a comparative study with conventional biomarkers.","authors":"Chuanbo Xie, Shuqing Wang, Chi Guo, Yuying Liu, Musheng Zeng","doi":"10.1007/s13402-023-00838-1","DOIUrl":"10.1007/s13402-023-00838-1","url":null,"abstract":"<p><p>The detection of biomarkers in blood macrophages is a new non-invasive cancer screening method, but its performance in early stage lung cancer screening remains undetermined. We evaluated the Apo10 and TKTL1 levels in blood macrophages of 156 early-stage lung cancer patients and 153 controls. APT (combination of Apo10 and TKTL1) level was significantly higher in the lung cancer group than that in the control group (P < 0.001). AUROC analysis showed that APT has high diagnostic value in differentiating early-stage lung cancer (AUC = 0.9132) and can be considered a biomarker for screening lung cancer patients from individuals with lung nodules.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2023-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9684340","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Clostridium butyricum and its metabolite butyrate promote ferroptosis susceptibility in pancreatic ductal adenocarcinoma.","authors":"Xiaotong Yang, Zhengyan Zhang, Xuqing Shen, Junyi Xu, Yawen Weng, Wei Wang, Jing Xue","doi":"10.1007/s13402-023-00831-8","DOIUrl":"10.1007/s13402-023-00831-8","url":null,"abstract":"<p><strong>Purpose: </strong>Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive disease with limited therapeutic options. The diversity and composition of the intratumoral microbiota are associated with PDAC outcomes, and modulating the tumor microbiota has the potential to influence tumor growth and the host immune response. Here, we explore whether intervention with butyrate-producing probiotics can limit PDAC progression.</p><p><strong>Methods: </strong>Based on the TCGA (PAAD) database, we analyzed the differential communities of intratumoral microbiota in PDAC patients with long survival and short survival and explored the relevant mechanisms of Clostridium butyricum and its metabolite butyrate in the treatment of PDAC. Treatment with Clostridium butyricum or butyrate in combination with the ferroptosis inducer RSL3 in a PDAC mouse model has an inhibitory effect on PDAC progression. The potential molecular mechanisms were verified by flow cytometry, RNA-seq, Western blotting, qRT‒PCR and immunofluorescence.</p><p><strong>Results: </strong>We found that the tumoral butyrate-producing microbiota was linked to a better prognosis and less aggressive features of PDAC. Intervention with Clostridium butyricum or its metabolite butyrate triggered superoxidative stress and intracellular lipid accumulation, which enhanced ferroptosis susceptibility in PDAC.</p><p><strong>Conclusion: </strong>Our study reveals a novel antitumor mechanism of butyrate and suggests the therapeutic potential of butyrate-producing probiotics in PDAC.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2023-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9554560","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Cellular OncologyPub Date : 2023-12-01Epub Date: 2023-07-12DOI: 10.1007/s13402-023-00840-7
Jaewang Lee, Jong-Lyel Roh
{"title":"Epigenetic modulation of ferroptosis in cancer: Identifying epigenetic targets for novel anticancer therapy.","authors":"Jaewang Lee, Jong-Lyel Roh","doi":"10.1007/s13402-023-00840-7","DOIUrl":"10.1007/s13402-023-00840-7","url":null,"abstract":"<p><p>Ferroptosis is a newly recognized form of oxidative-regulated cell death resulting from iron-mediated lipid peroxidation accumulation. Radical-trapping antioxidant systems can eliminate these oxidized lipids and prevent disrupting the integrity of cell membranes. Epigenetic modifications can regulate ferroptosis by altering gene expression or cell phenotype without permanent sequence changes. These mechanisms include DNA methylation, histone modifications, RNA modifications, and noncoding RNAs. Epigenetic alterations in cancer can control the expression of ferroptosis regulators or related pathways, leading to changes in cell sensitivity to ferroptosis inducers or cancer progression. Epigenetic alterations in cancer are influenced by a wide range of cancer hallmarks, contributing to therapeutic resistance. Targeting epigenetic alterations is a promising approach to overcoming cancer resilience. However, the exact mechanisms involved in different types of cancer remain unresolved. Discovering more ferroptosis-associated epigenetic targets and interventions can help overcome current barriers in anticancer therapy. Many papers on epigenetic modifications of ferroptosis have been continuously published, making it essential to summarize the current state-of-the-art in the epigenetic regulation of ferroptosis in human cancer.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2023-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"9774597","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Triggering pyroptosis enhances the antitumor efficacy of PARP inhibitors in prostate cancer.","authors":"Ao Tian, Tingyu Wu, Yanshuang Zhang, Jiachen Chen, Jianjun Sha, Weiliang Xia","doi":"10.1007/s13402-023-00860-3","DOIUrl":"10.1007/s13402-023-00860-3","url":null,"abstract":"<p><strong>Purpose: </strong>PARP inhibitors have revolutionized the treatment landscape for advanced prostate cancer (PCa) patients who harboring mutations in homologous recombination repair (HRR) genes. However, the molecular mechanisms underlying PARP inhibitors function beyond DNA damage repair pathways remain elusive, and identifying novel predictive targets that favorably respond to PARP inhibitors in PCa is an active area of research.</p><p><strong>Methods: </strong>The expression of GSDME in PCa cell lines and human PCa samples was determined by western blotting. Targeted bisulfite sequencing, gene enrichment analysis (GSEA), clone formation, construction of the stably transfected cell lines, lactate dehydrogenase (LDH) assay, western blotting as well as a mouse model of subcutaneous xenografts were used to investigate the role of GSDME in PCa. The combinational therapeutic effect of olaparib and decitabine was determined using both in vitro and in vivo experiments.</p><p><strong>Results: </strong>We have found low expression of GSDME in PCa. Interestingly, we demonstrated that GSDME activity is robustly induced in olaparib-treated cells undergoing pyroptosis, and that high methylation of the GSDME promoter dampens its activity in PCa cells. Intriguingly, genetically overexpressing GSDME does not inhibit tumor cell proliferation but instead confers sensitivity to olaparib. Furthermore, pharmacological treatment with the combination of olaparib and decitabine synergistically induces GSDME expression and cleavage through caspase-3 activation, thus promoting pyroptosis and enhancing anti-tumor response, ultimately resulting in tumor remission.</p><p><strong>Conclusion: </strong>Our findings highlight a novel therapeutic strategy for enhancing the long-term response to olaparib beyond HRR-deficient tumors in PCa, underscoring the critical role of GSDME in regulating tumorigenesis.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2023-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10043015","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}