Journal of Experimental & Clinical Cancer Research最新文献

筛选
英文 中文
UBASH3B-mediated MRPL12 Y60 dephosphorylation inhibits LUAD development by driving mitochondrial metabolism reprogramming. UBASH3B 介导的 MRPL12 Y60 去磷酸化可通过线粒体代谢重编程抑制 LUAD 的发育。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2024-09-30 DOI: 10.1186/s13046-024-03181-x
Xingzhao Ji, Tianyi Zhang, Jian Sun, Xiaojia Song, Guoyuan Ma, Li Xu, Xueru Cao, Yongjian Jing, Fuyuan Xue, Weiying Zhang, Shengnan Sun, Qiang Wan, Yi Liu
{"title":"UBASH3B-mediated MRPL12 Y60 dephosphorylation inhibits LUAD development by driving mitochondrial metabolism reprogramming.","authors":"Xingzhao Ji, Tianyi Zhang, Jian Sun, Xiaojia Song, Guoyuan Ma, Li Xu, Xueru Cao, Yongjian Jing, Fuyuan Xue, Weiying Zhang, Shengnan Sun, Qiang Wan, Yi Liu","doi":"10.1186/s13046-024-03181-x","DOIUrl":"10.1186/s13046-024-03181-x","url":null,"abstract":"<p><strong>Background: </strong>Metabolic reprogramming plays a pivotal role in tumorigenesis and development of lung adenocarcinoma (LUAD). However, the precise mechanisms and potential targets for metabolic reprogramming in LUAD remain elusive. Our prior investigations revealed that the mitochondrial ribosomal protein MRPL12, identified as a novel mitochondrial transcriptional regulatory gene, exerts a critical influence on mitochondrial metabolism. Despite this, the role and regulatory mechanisms underlying MRPL12's transcriptional activity in cancers remain unexplored.</p><p><strong>Methods: </strong>Human LUAD tissues, Tp53<sup>fl/fl</sup>;Kras<sup>G12D</sup>-driven LUAD mouse models, LUAD patient-derived organoids (PDO), and LUAD cell lines were used to explored the expression and function of MRPL12. The posttranslational modification of MRPL12 was analyzed by mass spectrometry, and the oncogenic role of key phosphorylation sites of MRPL12 in LUAD development was verified in vivo and in vitro.</p><p><strong>Results: </strong>MRPL12 was upregulated in human LUAD tissues, Tp53<sup>fl/fl</sup>;Kras<sup>G12D</sup>-driven LUAD tissues in mice, LUAD PDO, and LUAD cell lines, correlating with poor patient survival. Overexpression of MRPL12 significantly promoted LUAD tumorigenesis, metastasis, and PDO formation, while MRPL12 knockdown elicited the opposite phenotype. Additionally, MRPL12 deletion in a Tp53<sup>fl/fl</sup>;Kras<sup>G12D</sup>-driven mouse LUAD model conferred a notable survival advantage, delaying tumor onset and reducing malignant progression. Mechanistically, we discovered that MRPL12 promotes tumor progression by upregulating mitochondrial oxidative phosphorylation. Furthermore, we identified UBASH3B as a specific binder of MRPL12, dephosphorylating tyrosine 60 in MRPL12 (MRPL12 Y60) and inhibiting its oncogenic functions. The decrease in MRPL12 Y60 phosphorylation impeded the binding of MRPL12 to POLRMT, downregulating mitochondrial metabolism in LUAD cells. In-depth in vivo, in vitro, and organoid models validated the inhibitory effect of MRPL12 Y60 mutation on LUAD.</p><p><strong>Conclusion: </strong>This study establishes MRPL12 as a novel oncogene in LUAD, contributing to LUAD pathogenesis by orchestrating mitochondrial metabolism reprogramming towards oxidative phosphorylation (OXPHOS). Furthermore, it confirms Y60 as a specific phosphorylation modification site regulating MRPL12's oncogenic functions, offering insights for the development of LUAD-specific targeted drugs and clinical interventions.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"268"},"PeriodicalIF":11.4,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11441236/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142331521","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
LncOCMRL1 promotes oral squamous cell carcinoma growth and metastasis via the RRM2/EMT pathway. LncOCMRL1 通过 RRM2/EMT 通路促进口腔鳞状细胞癌的生长和转移。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2024-09-30 DOI: 10.1186/s13046-024-03190-w
Nan Lu, Qiming Jiang, Tianshu Xu, Qiyuan Gao, Yuepeng Wang, Zixian Huang, Zhiquan Huang, Xiaoding Xu
{"title":"LncOCMRL1 promotes oral squamous cell carcinoma growth and metastasis via the RRM2/EMT pathway.","authors":"Nan Lu, Qiming Jiang, Tianshu Xu, Qiyuan Gao, Yuepeng Wang, Zixian Huang, Zhiquan Huang, Xiaoding Xu","doi":"10.1186/s13046-024-03190-w","DOIUrl":"10.1186/s13046-024-03190-w","url":null,"abstract":"<p><strong>Background: </strong>Long noncoding RNAs (lncRNAs) are widely involved in cancer development and progression, but the functions of most lncRNAs have not yet been elucidated. Metastasis is the main factor restricting the therapeutic outcomes of various cancer types, including oral squamous cell carcinoma (OSCC). Therefore, exploring the key lncRNAs that regulate OSCC metastasis and elucidating their molecular mechanisms will facilitate the development of new strategies for effective OSCC therapy.</p><p><strong>Methods: </strong>We analyzed the lncRNA expression profiles of tumor tissues from OSCC patients with and without cervical lymph node metastasis, and OSCC cell lines. We revealed high expression of oral squamous cell carcinoma metastasis-related lncRNA 1 (lncOCMRL1) in OSCC patient tumor tissues with lymph node metastasis and highly metastatic OSCC cell lines. The effects of lncOCMRL1 knockdown on the invasion, migration and proliferation abilities of OSCC cells were explored through qRT-PCR, Transwell, colony formation, and cell proliferation experiments. The mechanism by which lncOCMRL1 promotes OSCC metastasis and proliferation was explored through RNA pull-down, silver staining, mass spectrometry, RIP, and WB experiments. To increase its translational potential, we developed a reduction-responsive nanodelivery system to deliver siRNA for antitumor therapy.</p><p><strong>Results: </strong>We determined that lncOCMRL1 is highly expressed in OSCC metastatic tumor tissues and cells. Functional studies have shown that high lncOCMRL1 expression can promote the growth and metastasis of OSCC cells both in vivo and in vitro. Mechanistically, lncOCMRL1 could induce epithelial-mesenchymal transition (EMT) via the suppression of RRM2 ubiquitination and thereby promote the proliferation, invasion, and migration of OSCC cells. We further constructed reduction-responsive nanoparticles (NPs) for the systemic delivery of siRNAs targeting lncOCMRL1 and demonstrated their high efficacy in silencing lncOCMRL1 expression in vivo and significantly inhibited OSCC tumor growth and metastasis.</p><p><strong>Conclusions: </strong>Our results suggest that lncOCMRL1 is a reliable target for blocking lymph node metastasis in OSCC.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"267"},"PeriodicalIF":11.4,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11441159/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142331517","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
MBD2 regulates the progression and chemoresistance of cholangiocarcinoma through interaction with WDR5. MBD2 通过与 WDR5 相互作用调控胆管癌的进展和化疗耐药性。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2024-09-30 DOI: 10.1186/s13046-024-03188-4
Da Wang, Junsheng Chen, Guanhua Wu, Fei Xiong, Wenzheng Liu, Qi Wang, Yiyang Kuai, Wenhua Huang, Yongqiang Qi, Bing Wang, Ruizhi He, Yongjun Chen
{"title":"MBD2 regulates the progression and chemoresistance of cholangiocarcinoma through interaction with WDR5.","authors":"Da Wang, Junsheng Chen, Guanhua Wu, Fei Xiong, Wenzheng Liu, Qi Wang, Yiyang Kuai, Wenhua Huang, Yongqiang Qi, Bing Wang, Ruizhi He, Yongjun Chen","doi":"10.1186/s13046-024-03188-4","DOIUrl":"10.1186/s13046-024-03188-4","url":null,"abstract":"<p><strong>Background: </strong>Cholangiocarcinoma (CCA) is a highly malignant, rapidly progressing tumor of the bile duct. Owing to its chemoresistance, it always has an extremely poor prognosis. Therefore, detailed elucidation of the mechanisms of chemoresistance and identification of therapeutic targets are still needed.</p><p><strong>Methods: </strong>We analyzed the expression of MBD2 (Methyl-CpG-binding domain 2) in CCA and normal bile duct tissues using the public database and immunohistochemistry (IHC). The roles of MBD2 in CCA cell proliferation, migration, and chemoresistance ability were validated through CCK-8, plate cloning assay, wound healing assays and xenograft mouse model. In addition, we constructed a primary CCA mouse model to further confirm the effect of MBD2. RNA-seq and co-IP-MS were used to identify the mechanisms by how MBD2 leads to chemoresistance.</p><p><strong>Results: </strong>MBD2 was upregulated in CCA. It promoted the proliferation, migration and chemoresistance of CCA cells. Mechanistically, MBD2 directly interacted with WDR5, bound to the promoter of ABCB1, promoted the trimethylation of H3K4 in this region through KMT2A, and activated the expression of ABCB1. Knocking down WDR5 or KMT2A blocked the transcriptional activation of ABCB1 by MBD2. The molecular inhibitor MM-102 targeted the interaction of WDR5 with KMT2A. MM-102 inhibited the expression of ABCB1 in CCA cells and decreased the chemoresistance of CCA to cisplatin.</p><p><strong>Conclusion: </strong>MBD2 promotes the progression and chemoresistance of CCA through interactions with WDR5. MM-102 can effectively block this process and increase the sensitivity of CCA to cisplatin.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"272"},"PeriodicalIF":11.4,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11440836/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142331518","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
Targeting DNM1L/DRP1-FIS1 axis inhibits high-grade glioma progression by impeding mitochondrial respiratory cristae remodeling. 靶向 DNM1L/DRP1-FIS1 轴可通过阻碍线粒体呼吸嵴重塑抑制高级别胶质瘤的进展。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2024-09-30 DOI: 10.1186/s13046-024-03194-6
Xiaodong Li, Jingjing Tie, Yuze Sun, Chengrong Gong, Shizhou Deng, Xiyu Chen, Shujiao Li, Yaoliang Wang, Zhenhua Wang, Feifei Wu, Hui Liu, Yousheng Wu, Guopeng Zhang, Qingdong Guo, Yanling Yang, Yayun Wang
{"title":"Targeting DNM1L/DRP1-FIS1 axis inhibits high-grade glioma progression by impeding mitochondrial respiratory cristae remodeling.","authors":"Xiaodong Li, Jingjing Tie, Yuze Sun, Chengrong Gong, Shizhou Deng, Xiyu Chen, Shujiao Li, Yaoliang Wang, Zhenhua Wang, Feifei Wu, Hui Liu, Yousheng Wu, Guopeng Zhang, Qingdong Guo, Yanling Yang, Yayun Wang","doi":"10.1186/s13046-024-03194-6","DOIUrl":"10.1186/s13046-024-03194-6","url":null,"abstract":"<p><strong>Background: </strong>The dynamics of mitochondrial respiratory cristae (MRC) and its impact on oxidative phosphorylation (OXPHOS) play a crucial role in driving the progression of high-grade glioma (HGG). However, the underlying mechanism remains unclear.</p><p><strong>Methods: </strong>In the present study, we employed machine learning-based transmission electron microscopy analysis of 7141 mitochondria from 54 resected glioma patients. Additionally, we conducted bioinformatics analysis and multiplex immunohistochemical (mIHC) staining of clinical glioma microarrays to identify key molecules involved in glioma. Subsequently, we modulated the expression levels of mitochondrial dynamic-1-like protein (DNM1L/DRP1), and its two receptors, mitochondrial fission protein 1 (FIS1) and mitochondrial fission factor (MFF), via lentiviral transfection to further investigate the central role of these molecules in the dynamics of glioblastoma (GBM) cells and glioma stem cells (GSCs). We then evaluated the potential impact of DNM1L/DRP1, FIS1, and MFF on the proliferation and progression of GBM cells and GSCs using a combination of CCK-8 assay, Transwell assay, Wound Healing assay, tumor spheroid formation assay and cell derived xenograft assay employing NOD/ShiLtJGpt-Prkdc<sup>em26Cd52</sup>Il2rg<sup>em26Cd22</sup>/Gpt (NCG) mouse model. Subsequently, we validated the ability of the DNM1L/DRP1-FIS1 axis to remodel MRC structure through mitophagy by utilizing Seahorse XF analysis technology, mitochondrial function detection, MRC abundance detection and monitoring dynamic changes in mitophagy.</p><p><strong>Results: </strong>Our findings revealed that compared to low-grade glioma (LGG), HGG exhibited more integrated MRC structures. Further research revealed that DNM1L/DRP1, FIS1, and MFF played pivotal roles in governing mitochondrial fission and remodeling MRC in HGG. The subsequent validation demonstrated that DNM1L/DRP1 exerts a positive regulatory effect on FIS1, whereas the interaction between MFF and FIS1 demonstrates a competitive inhibition relationship. The down-regulation of the DNM1L/DRP1-FIS1 axis significantly impaired mitophagy, thereby hindering the remodeling of MRC and inhibiting OXPHOS function in glioma, ultimately leading to the inhibition of its aggressive progression. In contrast, MFF exerts a contrasting effect on MRC integrity, OXPHOS activity, and glioma progression.</p><p><strong>Conclusions: </strong>This study highlights that the DNM1L/DRP1-FIS1 axis stabilizes MRC structures through mitophagy in HGG cells while driving their OXPHOS activity ultimately leading to robust disease progression. The inhibition of the DNM1L/DRP1-FIS1 axis hinders MRC remodeling and suppresses GBM progression. We propose that down-regulation of the DNM1L/DRP1-FIS1 axis could be a potential therapeutic strategy for treating HGG.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"273"},"PeriodicalIF":11.4,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11440692/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142331520","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
Dynamic glycolytic reprogramming effects on dendritic cells in pancreatic ductal adenocarcinoma. 动态糖酵解重编程对胰腺导管腺癌树突状细胞的影响
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2024-09-30 DOI: 10.1186/s13046-024-03192-8
Bo Zhang, Kenoki Ohuchida, Chikanori Tsutsumi, Yuki Shimada, Yuki Mochida, Koki Oyama, Chika Iwamoto, Nan Sheng, Shuang Fei, Koji Shindo, Naoki Ikenaga, Kohei Nakata, Yoshinao Oda, Masafumi Nakamura
{"title":"Dynamic glycolytic reprogramming effects on dendritic cells in pancreatic ductal adenocarcinoma.","authors":"Bo Zhang, Kenoki Ohuchida, Chikanori Tsutsumi, Yuki Shimada, Yuki Mochida, Koki Oyama, Chika Iwamoto, Nan Sheng, Shuang Fei, Koji Shindo, Naoki Ikenaga, Kohei Nakata, Yoshinao Oda, Masafumi Nakamura","doi":"10.1186/s13046-024-03192-8","DOIUrl":"10.1186/s13046-024-03192-8","url":null,"abstract":"<p><strong>Background: </strong>Pancreatic ductal adenocarcinoma tumors exhibit resistance to chemotherapy, targeted therapies, and even immunotherapy. Dendritic cells use glucose to support their effector functions and play a key role in anti-tumor immunity by promoting cytotoxic CD8<sup>+</sup> T cell activity. However, the effects of glucose and lactate levels on dendritic cells in pancreatic ductal adenocarcinoma are unclear. In this study, we aimed to clarify how glucose and lactate can impact the dendritic cell antigen-presenting function and elucidate the relevant mechanisms.</p><p><strong>Methods: </strong>Glycolytic activity and immune cell infiltration in pancreatic ductal adenocarcinoma were evaluated using patient-derived organoids and resected specimens. Cell lines with increased or decreased glycolysis were established from KPC mice. Flow cytometry and single-cell RNA sequencing were used to evaluate the impacts on the tumor microenvironment. The effects of glucose and lactate on the bone marrow-derived dendritic cell antigen-presenting function were detected by flow cytometry.</p><p><strong>Results: </strong>The pancreatic ductal adenocarcinoma tumor microenvironment exhibited low glucose and high lactate concentrations from varying levels of glycolytic activity in cancer cells. In mouse transplantation models, tumors with increased glycolysis showed enhanced myeloid-derived suppressor cell infiltration and reduced dendritic cell and CD8<sup>+</sup> T cell infiltration, whereas tumors with decreased glycolysis displayed the opposite trends. In three-dimensional co-culture, increased glycolysis in cancer cells suppressed the antigen-presenting function of bone marrow-derived dendritic cells. In addition, low-glucose and high-lactate media inhibited the antigen-presenting and mitochondrial functions of bone marrow-derived dendritic cells.</p><p><strong>Conclusions: </strong>Our study demonstrates the impact of dynamic glycolytic reprogramming on the composition of immune cells in the tumor microenvironment of pancreatic ductal adenocarcinoma, especially on the antigen-presenting function of dendritic cells.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"271"},"PeriodicalIF":11.4,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11441259/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142331501","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
BAP1 regulates HSF1 activity and cancer immunity in pancreatic cancer. BAP1 可调节胰腺癌中 HSF1 的活性和癌症免疫。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2024-09-30 DOI: 10.1186/s13046-024-03196-4
Weiwei Yuan, Qiyue Zhang, Yuhan Zhao, Wentao Xia, Shilin Yin, Xueyi Liang, Taoyu Chen, Gaofeng Li, Yanshen Liu, Zhiqiang Liu, Jinxi Huang
{"title":"BAP1 regulates HSF1 activity and cancer immunity in pancreatic cancer.","authors":"Weiwei Yuan, Qiyue Zhang, Yuhan Zhao, Wentao Xia, Shilin Yin, Xueyi Liang, Taoyu Chen, Gaofeng Li, Yanshen Liu, Zhiqiang Liu, Jinxi Huang","doi":"10.1186/s13046-024-03196-4","DOIUrl":"10.1186/s13046-024-03196-4","url":null,"abstract":"<p><strong>Background: </strong>The vast majority of pancreatic cancers have been shown to be insensitive to single-agent immunotherapy. Exploring the mechanisms of immune resistance and implementing combination therapeutic strategies are crucial for PDAC patients to derive benefits from immunotherapy. Deletion of BAP1 occurs in approximately 27% of PDAC patients and is significantly correlated with poor prognosis, but the mechanism how BAP1-deletion compromises survival of patients with PDAC remain a puzzle.</p><p><strong>Methods: </strong>Bap1 knock-out KPC (KrasG12D/+; LSLTrp53R172H/+; Pdx-1-Cre) mice and control KPC mice, syngeneic xenograft models were applied to analysis the correlation between BAP1 and immune therapy response in PDAC. Immunoprecipitation, RT-qPCR, luciferase and transcriptome analysis were combined to revealing potential mechanisms. Syngeneic xenograft models and flow cytometry were constructed to examine the efficacy of the inhibitor of SIRT1 and its synergistic effect with anti-PD-1 therapy.</p><p><strong>Result: </strong>The deletion of BAP1 contributes to the resistance to immunotherapy in PDAC, which is attributable to BAP1's suppression of the transcriptional activity of HSF1. Specifically, BAP1 competes with SIRT1 for binding to the K80 acetylated HSF1. The BAP1-HSF1 interaction preserves the acetylation of HSF1-K80 and promotes HSF1-HSP70 interaction, facilitating HSF1 oligomerization and detachment from the chromatin. Furthermore, we demonstrate that the targeted inhibition of SIRT1 reverses the immune insensitivity in BAP1 deficient PDAC mouse model.</p><p><strong>Conclusion: </strong>Our study elucidates an unrevealed mechanism by which BAP1 regulates immune therapy response in PDAC via HSF1 inhibition, and providing promising therapeutic strategies to address immune insensitivity in BAP1-deficient PDAC.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"275"},"PeriodicalIF":11.4,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11441124/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142331499","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
Mediator complex subunit 1 promotes oral squamous cell carcinoma progression by activating MMP9 transcription and suppressing CD8+ T cell antitumor immunity. 介导复合体亚基 1 通过激活 MMP9 转录和抑制 CD8+ T 细胞抗肿瘤免疫促进口腔鳞状细胞癌的发展。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2024-09-30 DOI: 10.1186/s13046-024-03191-9
Zhe Li, Mengke Sun, Ruimeng Yang, Zheng Wang, Qianyu Zhu, Yue Zhang, Haosun Yang, Zhaosong Meng, Lizhi Hu, Lei Sui
{"title":"Mediator complex subunit 1 promotes oral squamous cell carcinoma progression by activating MMP9 transcription and suppressing CD8<sup>+</sup> T cell antitumor immunity.","authors":"Zhe Li, Mengke Sun, Ruimeng Yang, Zheng Wang, Qianyu Zhu, Yue Zhang, Haosun Yang, Zhaosong Meng, Lizhi Hu, Lei Sui","doi":"10.1186/s13046-024-03191-9","DOIUrl":"10.1186/s13046-024-03191-9","url":null,"abstract":"<p><strong>Background: </strong>The role of Mediator complex subunit 1 (MED1), a pivotal transcriptional coactivator implicated in diverse biological pathways, remains unexplored in the context of oral squamous cell carcinoma (OSCC). This study aims to elucidate the contributory mechanisms and potential impact of MED1 on the progression of OSCC.</p><p><strong>Methods: </strong>The expression and clinical significance of MED1 in OSCC tissues were evaluated through the bioinformatics analyses. The effects of MED1 on the biological behavior of OSCC cancer cells were assessed both in vitro and in vivo. Dual-luciferase reporter assay, chromatin immunoprecipitation (ChIP) assay, bioinformatic analysis, CD8<sup>+</sup> T cell isolation experiment, coculture experiment, enzyme-linked immunosorbent assay (ELISA), and flow cytometric analysis were employed to elucidate the underlying mechanism through which MED1 operates in the progression of OSCC.</p><p><strong>Results: </strong>MED1 exhibited upregulation in both OSCC tissues and multiple OSCC cell lines, which correlated with decreased overall survival in patients. In vitro experiments demonstrated that knockdown of MED1 in metastatic OSCC cell lines SCC-9 and UPCI-SCC-154 hindered cell migration and invasion, while overexpression of MED1 promoted these processes. Whereas, MED1 knockdown had no impact on proliferation of cell lines mentioned above. In vivo studies further revealed that downregulation of MED1 effectively suppressed distant metastasis in OSCC. Mechanistically, MED1 enhanced the binding of transcription factors c-Jun and c-Fos to the matrix metalloprotein 9 (MMP9) promoters, resulting in a significant upregulation of MMP9 transcription. This process contributes to the migration and invasion of SCC-9 and UPCI-SCC-154 cells. Furthermore, MED1 modulated the expression of programmed death-ligand 1 (PD-L1) through the Notch signaling pathway, consequently impacting the tumor-killing capacity of CD8<sup>+</sup> T cells in the tumor microenvironment.</p><p><strong>Conclusions: </strong>Our findings indicate that MED1 plays a pivotal role in OSCC progression through the activation of MMP9 transcription and suppression of CD8<sup>+</sup> T cell antitumor immunity, suggesting that MED1 may serve as a novel prognostic marker and therapeutic target in OSCC.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"270"},"PeriodicalIF":11.4,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11440895/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142331519","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
HIF-1α-HPRT1 axis promotes tumorigenesis and gefitinib resistance by enhancing purine metabolism in EGFR-mutant lung adenocarcinoma. HIF-1α-HPRT1轴通过增强表皮生长因子受体突变型肺腺癌的嘌呤代谢,促进肿瘤发生和吉非替尼耐药。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2024-09-30 DOI: 10.1186/s13046-024-03184-8
Pengyu Geng, Fei Ye, Peng Dou, Chunxiu Hu, Jiarui He, Jinhui Zhao, Qi Li, Miao Bao, Xiangnan Li, Xinyu Liu, Guowang Xu
{"title":"HIF-1α-HPRT1 axis promotes tumorigenesis and gefitinib resistance by enhancing purine metabolism in EGFR-mutant lung adenocarcinoma.","authors":"Pengyu Geng, Fei Ye, Peng Dou, Chunxiu Hu, Jiarui He, Jinhui Zhao, Qi Li, Miao Bao, Xiangnan Li, Xinyu Liu, Guowang Xu","doi":"10.1186/s13046-024-03184-8","DOIUrl":"10.1186/s13046-024-03184-8","url":null,"abstract":"<p><strong>Background: </strong>The mutations of oncogenic epidermal growth factor receptor (EGFR) is an important cause of lung adenocarcinoma (LUAD) malignance. It has been knowm that metabolic reprogramming is an important hallmark of malignant tumors, and purine metabolism is a key metabolic pathway for tumor progression and drug resistance, but its relationship with the EGFR-mutant LUAD is unclear.</p><p><strong>Methods: </strong>Metabolic reprogramming was studied through capillary electrophoresis-time of flight mass spectrometry (CE-TOF/MS)-based metabolic profiling analysis. Cell proliferation in vitro was evaluated by EdU staining and cell cycle assay. Tumorigenicity in vivo was tested by subcutaneous tumor formation experiment in nude mice. The binding of hypoxia-inducible factor-1 alpha (HIF-1α) and hypoxanthine phosphoribosyltransferase 1 (HPRT1) was detected by DNA pull‑down assay and Chromatin immunoprecipitation (ChIP) assays. HIF-1α, HPRT1, DNA damage and cell apoptosis related genes were examined by western blot. In addition, RNA sequencing, mass spectrometry and bioinformatics analysis were performed.</p><p><strong>Results: </strong>We found that mutated EGFR (muEGFR) upregulates HPRT1 to promote purine metabolism and tumorigenesis of EGFR-mutant LUAD. Mechanistically, muEGFR increases HIF-1α expression through protein stability. Meanwhile, up-regulated HIF-1α bound to the promoter of HPRT1 and transcriptionally activates HPRT1 expression, enhancing purine metabolism to maintain rapid tumor cell proliferation in EGFR-mutant LUAD. Further, gefitinib inhibited the synthesis of purine nucleotides, and HPRT1 inhibition increased the sensitivity of gefitinib to EGFR-mutant LUAD.</p><p><strong>Conclusions: </strong>Our study reveals that muEGFR-HIF-1α-HPRT1 axis plays a key role in EGFR-mutant LUAD and provides a new strategy-inhibiting purine metabolism for treating EGFR-mutant LUAD.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"269"},"PeriodicalIF":11.4,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11441087/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142331515","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
Copy number amplification-induced overexpression of lncRNA LOC101927668 facilitates colorectal cancer progression by recruiting hnRNPD to disrupt RBM47/p53/p21 signaling. 拷贝数扩增诱导的lncRNA LOC101927668过表达通过招募hnRNPD破坏RBM47/p53/p21信号转导而促进结直肠癌的进展。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2024-09-30 DOI: 10.1186/s13046-024-03193-7
Zaozao Wang, Haibo Han, Chenghai Zhang, Chenxin Wu, Jiabo Di, Pu Xing, Xiaowen Qiao, Kai Weng, Hao Hao, Xinying Yang, Yifan Hou, Beihai Jiang, Xiangqian Su
{"title":"Copy number amplification-induced overexpression of lncRNA LOC101927668 facilitates colorectal cancer progression by recruiting hnRNPD to disrupt RBM47/p53/p21 signaling.","authors":"Zaozao Wang, Haibo Han, Chenghai Zhang, Chenxin Wu, Jiabo Di, Pu Xing, Xiaowen Qiao, Kai Weng, Hao Hao, Xinying Yang, Yifan Hou, Beihai Jiang, Xiangqian Su","doi":"10.1186/s13046-024-03193-7","DOIUrl":"10.1186/s13046-024-03193-7","url":null,"abstract":"<p><strong>Background: </strong>Somatic copy number alterations (SCNAs) are pivotal in cancer progression and patient prognosis. Dysregulated long non-coding RNAs (lncRNAs), modulated by SCNAs, significantly impact tumorigenesis, including colorectal cancer (CRC). Nonetheless, the functional significance of lncRNAs induced by SCNAs in CRC remains largely unexplored.</p><p><strong>Methods: </strong>The dysregulated lncRNA LOC101927668, induced by copy number amplification, was identified through comprehensive bioinformatic analyses utilizing multidimensional data. Subsequent in situ hybridization was employed to ascertain the subcellular localization of LOC101927668, and gain- and loss-of-function experiments were conducted to elucidate its role in CRC progression. The downstream targets and signaling pathway influenced by LOC101927668 were identified and validated through a comprehensive approach, encompassing RNA sequencing, RT-qPCR, Western blot analysis, dual-luciferase reporter assay, evaluation of mRNA and protein degradation, and rescue experiments. Analysis of AU-rich elements (AREs) within the mRNA 3' untranslated region (UTR) of the downstream target, along with exploration of putative ARE-binding proteins, was conducted. RNA pull-down, mass spectrometry, RNA immunoprecipitation, and dual-luciferase reporter assays were employed to elucidate potential interacting proteins of LOC101927668 and further delineate the regulatory mechanism between LOC101927668 and its downstream target. Moreover, subcutaneous xenograft and orthotopic liver xenograft tumor models were utilized to evaluate the in vivo impact of LOC101927668 on CRC cells and investigate its correlation with downstream targets.</p><p><strong>Results: </strong>Significantly overexpressed LOC101927668, driven by chr7p22.3-p14.3 amplification, was markedly correlated with unfavorable clinical outcomes in our CRC patient cohort, as well as in TCGA and GEO datasets. Moreover, we demonstrated that enforced expression of LOC101927668 significantly enhanced cell proliferation, migration, and invasion, while its depletion impeded these processes in a p53-dependent manner. Mechanistically, nucleus-localized LOC101927668 recruited hnRNPD and translocated to the cytoplasm, accelerating the destabilization of RBM47 mRNA, a transcription factor of p53. As a nucleocytoplasmic shuttling protein, hnRNPD mediated RBM47 destabilization by binding to the ARE motif within RBM47 3'UTR, thereby suppressing the p53 signaling pathway and facilitating CRC progression.</p><p><strong>Conclusions: </strong>The overexpression of LOC101927668, driven by SCNAs, facilitates CRC proliferation and metastasis by recruiting hnRNPD, thus perturbing the RBM47/p53/p21 signaling pathway. These findings underscore the pivotal roles of LOC101927668 and highlight its therapeutic potential in anti-CRC interventions.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"274"},"PeriodicalIF":11.4,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11440719/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142331500","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
Focusing on CD8+ T-cell phenotypes: improving solid tumor therapy. 关注 CD8+ T 细胞表型:改善实体瘤治疗。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2024-09-28 DOI: 10.1186/s13046-024-03195-5
Zhouchi Yao, Yayun Zeng, Cheng Liu, Huimin Jin, Hong Wang, Yue Zhang, Chengming Ding, Guodong Chen, Daichao Wu
{"title":"Focusing on CD8<sup>+</sup> T-cell phenotypes: improving solid tumor therapy.","authors":"Zhouchi Yao, Yayun Zeng, Cheng Liu, Huimin Jin, Hong Wang, Yue Zhang, Chengming Ding, Guodong Chen, Daichao Wu","doi":"10.1186/s13046-024-03195-5","DOIUrl":"https://doi.org/10.1186/s13046-024-03195-5","url":null,"abstract":"<p><p>Vigorous CD8<sup>+</sup> T cells play a crucial role in recognizing tumor cells and combating solid tumors. How T cells efficiently recognize and target tumor antigens, and how they maintain the activity in the \"rejection\" of solid tumor microenvironment, are major concerns. Recent advances in understanding of the immunological trajectory and lifespan of CD8<sup>+</sup> T cells have provided guidance for the design of more optimal anti-tumor immunotherapy regimens. Here, we review the newly discovered methods to enhance the function of CD8<sup>+</sup> T cells against solid tumors, focusing on optimizing T cell receptor (TCR) expression, improving antigen recognition by engineered T cells, enhancing signal transduction of the TCR-CD3 complex, inducing the homing of polyclonal functional T cells to tumors, reversing T cell exhaustion under chronic antigen stimulation, and reprogramming the energy and metabolic pathways of T cells. We also discuss how to participate in the epigenetic changes of CD8<sup>+</sup> T cells to regulate two key indicators of anti-tumor responses, namely effectiveness and persistence.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"266"},"PeriodicalIF":11.4,"publicationDate":"2024-09-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11437975/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142331514","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
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