首页 > 最新文献

Journal of Experimental & Clinical Cancer Research最新文献

英文 中文
CALB2 drives pancreatic cancer metastasis through inflammatory reprogramming of the tumor microenvironment. CALB2通过肿瘤微环境的炎症重编程驱动胰腺癌转移。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-10-03 DOI: 10.1186/s13046-024-03201-w
Jinxin Tao, Yani Gu, Zeyu Zhang, Guihu Weng, Yueze Liu, Jie Ren, Yanan Shi, Jiangdong Qiu, Yuanyang Wang, Dan Su, Ruobing Wang, Yifan Fu, Tao Liu, Liyuan Ye, Wenhao Luo, Hao Chen, Gang Yang, Zhe Cao, Hua Huang, Jianchun Xiao, Bo Ren, Lei You, Taiping Zhang, Yupei Zhao

Background: Early dissemination to distant organs accounts for the dismal prognosis of patients with pancreatic ductal adenocarcinoma (PDAC). Chronic, dysregulated, persistent and unresolved inflammation provides a preferred tumor microenvironment (TME) for tumorigenesis, development, and metastasis. A better understanding of the key regulators that maintain inflammatory TME and the development of predictive biomarkers to identify patients who are most likely to benefit from specific inflammatory-targeted therapies is crucial for advancing personalized cancer treatment.

Methods: This study identified cell-specific expression of CALB2 in human PDAC through single-cell RNA sequencing analysis and assessed its clinicopathological correlations in tissue microarray using multi-color immunofluorescence. Co-culture systems containing cancer-associated fibroblasts (CAFs) and patient-derived organoids (PDOs) in vitro and in vivo were employed to elucidate the effects of CALB2-activated CAFs on PDAC malignancy. Furthermore, CUT&RUN assays, luciferase reporter assays, RNA sequencing, and gain- or loss-of-function assays were used to unravel the molecular mechanisms of CALB2-mediated inflammatory reprogramming and metastasis. Additionally, immunocompetent KPC organoid allograft models were constructed to evaluate CALB2-induced immunosuppression and PDAC metastasis, as well as the efficacy of inflammation-targeted therapy.

Results: CALB2 was highly expressed both in CAFs and cancer cells and correlated with an unfavorable prognosis and immunosuppressive TME in PDAC patients. CALB2 collaborated with hypoxia to activate an inflammatory fibroblast phenotype, which promoted PDAC cell migration and PDO growth in vitro and in vivo. In turn, CALB2-activated CAFs upregulated CALB2 expression in cancer cells through IL6-STAT3 signaling-mediated direct transcription. In cancer cells, CALB2 further activated Ca2+-CXCL14 inflammatory axis to facilitate PDAC metastatic outgrowth and immunosuppression. Genetic or pharmaceutical inhibition of CXCL14 significantly suppressed CALB2-mediated metastatic colonization of PDAC cells in vivo and extended mouse survival.

Conclusions: These findings identify CALB2 as a key regulator of inflammatory reprogramming to promote PDAC metastatic progression. Combination therapy with αCXCL14 monoclonal antibody and gemcitabine emerges as a promising strategy to suppress distant metastasis and improve survival outcomes in PDAC with CALB2 overexpression.

背景:胰腺导管腺癌(PDAC)患者预后不佳的原因是肿瘤早期向远处器官播散。慢性、失调、持续和未解决的炎症为肿瘤发生、发展和转移提供了有利的肿瘤微环境(TME)。更好地了解维持炎症TME的关键调控因子以及开发预测性生物标记物来识别最有可能从特定炎症靶向疗法中获益的患者,对于推进个性化癌症治疗至关重要:本研究通过单细胞RNA测序分析确定了CALB2在人PDAC中的细胞特异性表达,并使用多色免疫荧光评估了其在组织芯片中的临床病理相关性。为了阐明CALB2激活的CAFs对PDAC恶性肿瘤的影响,研究人员采用了包含癌相关成纤维细胞(CAFs)和患者衍生器官组织(PDOs)的体外和体内共培养系统。此外,研究还采用了CUT&RUN测定、荧光素酶报告实验、RNA测序以及功能增益或缺失实验来揭示CALB2介导的炎症重编程和转移的分子机制。此外,还构建了免疫功能正常的KPC类器官异体移植模型,以评估CALB2诱导的免疫抑制和PDAC转移以及炎症靶向治疗的疗效:结果:CALB2在CAFs和癌细胞中均高表达,并与PDAC患者的不良预后和免疫抑制TME相关。CALB2与缺氧共同激活了炎性成纤维细胞表型,从而促进了PDAC细胞的体外和体内迁移和PDO的生长。反过来,CALB2激活的CAF通过IL6-STAT3信号介导的直接转录上调癌细胞中CALB2的表达。在癌细胞中,CALB2 进一步激活 Ca2+-CXCL14 炎症轴,促进 PDAC 转移生长和免疫抑制。基因或药物抑制 CXCL14 能显著抑制 CALB2 介导的 PDAC 细胞体内转移定植,并延长小鼠存活期:这些研究结果表明,CALB2是炎症重编程促进PDAC转移进展的关键调节因子。αCXCL14单克隆抗体与吉西他滨的联合疗法是抑制远处转移和改善CALB2过表达的PDAC患者生存预后的一种有前途的策略。
{"title":"CALB2 drives pancreatic cancer metastasis through inflammatory reprogramming of the tumor microenvironment.","authors":"Jinxin Tao, Yani Gu, Zeyu Zhang, Guihu Weng, Yueze Liu, Jie Ren, Yanan Shi, Jiangdong Qiu, Yuanyang Wang, Dan Su, Ruobing Wang, Yifan Fu, Tao Liu, Liyuan Ye, Wenhao Luo, Hao Chen, Gang Yang, Zhe Cao, Hua Huang, Jianchun Xiao, Bo Ren, Lei You, Taiping Zhang, Yupei Zhao","doi":"10.1186/s13046-024-03201-w","DOIUrl":"10.1186/s13046-024-03201-w","url":null,"abstract":"<p><strong>Background: </strong>Early dissemination to distant organs accounts for the dismal prognosis of patients with pancreatic ductal adenocarcinoma (PDAC). Chronic, dysregulated, persistent and unresolved inflammation provides a preferred tumor microenvironment (TME) for tumorigenesis, development, and metastasis. A better understanding of the key regulators that maintain inflammatory TME and the development of predictive biomarkers to identify patients who are most likely to benefit from specific inflammatory-targeted therapies is crucial for advancing personalized cancer treatment.</p><p><strong>Methods: </strong>This study identified cell-specific expression of CALB2 in human PDAC through single-cell RNA sequencing analysis and assessed its clinicopathological correlations in tissue microarray using multi-color immunofluorescence. Co-culture systems containing cancer-associated fibroblasts (CAFs) and patient-derived organoids (PDOs) in vitro and in vivo were employed to elucidate the effects of CALB2-activated CAFs on PDAC malignancy. Furthermore, CUT&RUN assays, luciferase reporter assays, RNA sequencing, and gain- or loss-of-function assays were used to unravel the molecular mechanisms of CALB2-mediated inflammatory reprogramming and metastasis. Additionally, immunocompetent KPC organoid allograft models were constructed to evaluate CALB2-induced immunosuppression and PDAC metastasis, as well as the efficacy of inflammation-targeted therapy.</p><p><strong>Results: </strong>CALB2 was highly expressed both in CAFs and cancer cells and correlated with an unfavorable prognosis and immunosuppressive TME in PDAC patients. CALB2 collaborated with hypoxia to activate an inflammatory fibroblast phenotype, which promoted PDAC cell migration and PDO growth in vitro and in vivo. In turn, CALB2-activated CAFs upregulated CALB2 expression in cancer cells through IL6-STAT3 signaling-mediated direct transcription. In cancer cells, CALB2 further activated Ca<sup>2+</sup>-CXCL14 inflammatory axis to facilitate PDAC metastatic outgrowth and immunosuppression. Genetic or pharmaceutical inhibition of CXCL14 significantly suppressed CALB2-mediated metastatic colonization of PDAC cells in vivo and extended mouse survival.</p><p><strong>Conclusions: </strong>These findings identify CALB2 as a key regulator of inflammatory reprogramming to promote PDAC metastatic progression. Combination therapy with αCXCL14 monoclonal antibody and gemcitabine emerges as a promising strategy to suppress distant metastasis and improve survival outcomes in PDAC with CALB2 overexpression.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"277"},"PeriodicalIF":11.4,"publicationDate":"2024-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11448066/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142367268","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
IRF2 loss is associated with reduced MHC I pathway transcripts in subsets of most human cancers and causes resistance to checkpoint immunotherapy in human and mouse melanomas. IRF2 的缺失与大多数人类癌症亚群中 MHC I 通路转录物的减少有关,并导致人类和小鼠黑色素瘤对检查点免疫疗法产生抗药性。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-10-02 DOI: 10.1186/s13046-024-03187-5
G Sari, K Dhatchinamoorthy, L Orellano-Ariza, L M Ferreira, M A Brehm, K Rock

Background: In order for cancers to progress, they must evade elimination by CD8 T cells or other immune mechanisms. CD8 T cells recognize and kill tumor cells that display immunogenic tumor peptides bound to MHC I molecules. One of the ways that cancers can escape such killing is by reducing expression of MHC I molecules, and loss of MHC I is frequently observed in tumors. There are multiple different mechanisms that can underly the loss of MHC I complexes on tumor and it is currently unclear whether there are particular mechanisms that occur frequently and, if so, in what types of cancers. Also of importance to know is whether the loss of MHC I is reversible and how such loss and/or its restoration would impact responses to immunotherapy. Here, we investigate these issues for loss of IRF1 and IRF2, which are transcription factors that drive expression of MHC I pathway genes and some killing mechanisms.

Methods: Bioinformatics analyses of IRF2 and IRF2-dependent gene transcripts were performed for all human cancers in the TCGA RNAseq database. IRF2 protein-DNA-binding was analyzed in ChIPseq databases. CRISRPcas9 was used to knock out IRF1 and IRF2 genes in human and mouse melanoma cells and the resulting phenotypes were analyzed in vitro and in vivo.

Results: Transcriptomic analysis revealed that IRF2 expression was reduced in a substantial subset of cases in almost all types of human cancers. When this occurred there was a corresponding reduction in the expression of IRF2-regulated genes that were needed for CD8 T cell recognition. To test cause and effect for these IRF2 correlations and the consequences of IRF2 loss, we gene-edited IRF2 in a patient-derived melanoma and a mouse melanoma. The IRF2 gene-edited melanomas had reduced expression of transcripts for genes in the MHC I pathway and decreased levels of MHC I complexes on the cell surface. Levels of Caspase 7, an IRF2 target gene involved in CD8 T cell killing of tumors, were also reduced. This loss of IRF2 caused both human and mouse melanomas to become resistant to immunotherapy with a checkpoint inhibitor. Importantly, these effects were reversible. Stimulation of the IRF2-deficient melanomas with interferon induced the expression of a functionally homologous transcription factor, IRF1, which then restored the MHC I pathway and responsiveness to CPI.

Conclusions: Our study shows that a subset of cases within most types of cancers downregulates IRF2 and that this can allow cancers to escape immune control. This can cause resistance to checkpoint blockade immunotherapy and is reversible with currently available biologics.

背景:癌症要取得进展,必须避开 CD8 T 细胞或其他免疫机制的清除。CD8 T 细胞能识别并杀死显示与 MHC I 分子结合的免疫原性肿瘤肽的肿瘤细胞。癌症逃避这种杀伤的方法之一是减少 MHC I 分子的表达,而 MHC I 的缺失在肿瘤中经常出现。MHC I 复合物在肿瘤中的缺失有多种不同的机制,目前还不清楚是否存在经常发生的特定机制,如果存在,是在哪种类型的癌症中。同样重要的是,MHC I 的缺失是否可逆,以及这种缺失和/或其恢复将如何影响对免疫疗法的反应。IRF1和IRF2是驱动MHC I通路基因表达和一些杀伤机制的转录因子:方法:对 TCGA RNAseq 数据库中所有人类癌症的 IRF2 和 IRF2 依赖性基因转录本进行生物信息学分析。在 ChIPseq 数据库中分析了 IRF2 蛋白-DNA 结合。利用 CRISRPcas9 基因敲除人和小鼠黑色素瘤细胞中的 IRF1 和 IRF2 基因,并对由此产生的表型进行体外和体内分析:结果:转录组分析表明,在几乎所有类型的人类癌症中,IRF2的表达在相当一部分病例中都有所减少。在这种情况下,CD8 T 细胞识别所需的 IRF2 调控基因的表达也相应减少。为了检验这些IRF2相关性的因果关系以及IRF2缺失的后果,我们在患者衍生的黑色素瘤和小鼠黑色素瘤中对IRF2进行了基因编辑。经 IRF2 基因编辑的黑色素瘤的 MHC I 通路基因转录本表达量减少,细胞表面的 MHC I 复合物水平降低。参与CD8 T细胞杀伤肿瘤的IRF2靶基因Caspase 7的水平也有所降低。IRF2的缺失导致人类和小鼠黑色素瘤对使用检查点抑制剂的免疫疗法产生抗药性。重要的是,这些影响是可逆的。用干扰素刺激IRF2缺失的黑色素瘤可诱导功能同源的转录因子IRF1的表达,从而恢复MHC I通路和对CPI的反应性:我们的研究表明,在大多数类型的癌症中,有一部分病例会下调IRF2,这可能会使癌症逃避免疫控制。这可能导致对检查点阻断免疫疗法产生抗药性,而目前可用的生物制剂可逆转这种抗药性。
{"title":"IRF2 loss is associated with reduced MHC I pathway transcripts in subsets of most human cancers and causes resistance to checkpoint immunotherapy in human and mouse melanomas.","authors":"G Sari, K Dhatchinamoorthy, L Orellano-Ariza, L M Ferreira, M A Brehm, K Rock","doi":"10.1186/s13046-024-03187-5","DOIUrl":"10.1186/s13046-024-03187-5","url":null,"abstract":"<p><strong>Background: </strong>In order for cancers to progress, they must evade elimination by CD8 T cells or other immune mechanisms. CD8 T cells recognize and kill tumor cells that display immunogenic tumor peptides bound to MHC I molecules. One of the ways that cancers can escape such killing is by reducing expression of MHC I molecules, and loss of MHC I is frequently observed in tumors. There are multiple different mechanisms that can underly the loss of MHC I complexes on tumor and it is currently unclear whether there are particular mechanisms that occur frequently and, if so, in what types of cancers. Also of importance to know is whether the loss of MHC I is reversible and how such loss and/or its restoration would impact responses to immunotherapy. Here, we investigate these issues for loss of IRF1 and IRF2, which are transcription factors that drive expression of MHC I pathway genes and some killing mechanisms.</p><p><strong>Methods: </strong>Bioinformatics analyses of IRF2 and IRF2-dependent gene transcripts were performed for all human cancers in the TCGA RNAseq database. IRF2 protein-DNA-binding was analyzed in ChIPseq databases. CRISRPcas9 was used to knock out IRF1 and IRF2 genes in human and mouse melanoma cells and the resulting phenotypes were analyzed in vitro and in vivo.</p><p><strong>Results: </strong>Transcriptomic analysis revealed that IRF2 expression was reduced in a substantial subset of cases in almost all types of human cancers. When this occurred there was a corresponding reduction in the expression of IRF2-regulated genes that were needed for CD8 T cell recognition. To test cause and effect for these IRF2 correlations and the consequences of IRF2 loss, we gene-edited IRF2 in a patient-derived melanoma and a mouse melanoma. The IRF2 gene-edited melanomas had reduced expression of transcripts for genes in the MHC I pathway and decreased levels of MHC I complexes on the cell surface. Levels of Caspase 7, an IRF2 target gene involved in CD8 T cell killing of tumors, were also reduced. This loss of IRF2 caused both human and mouse melanomas to become resistant to immunotherapy with a checkpoint inhibitor. Importantly, these effects were reversible. Stimulation of the IRF2-deficient melanomas with interferon induced the expression of a functionally homologous transcription factor, IRF1, which then restored the MHC I pathway and responsiveness to CPI.</p><p><strong>Conclusions: </strong>Our study shows that a subset of cases within most types of cancers downregulates IRF2 and that this can allow cancers to escape immune control. This can cause resistance to checkpoint blockade immunotherapy and is reversible with currently available biologics.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"276"},"PeriodicalIF":11.4,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11446056/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142362436","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
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 Q1 ONCOLOGY 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

Background: 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.

Methods: Human LUAD tissues, Tp53fl/fl;KrasG12D-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.

Results: MRPL12 was upregulated in human LUAD tissues, Tp53fl/fl;KrasG12D-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 Tp53fl/fl;KrasG12D-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.

Conclusion: 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.

背景:代谢重编程在肺腺癌(LUAD)的肿瘤发生和发展中起着关键作用。然而,LUAD 中代谢重编程的确切机制和潜在靶点仍未确定。我们之前的研究发现,线粒体核糖体蛋白 MRPL12 是一种新型的线粒体转录调控基因,对线粒体代谢具有关键影响。方法:利用人类 LUAD 组织、Tp53fl/fl;KrasG12D 驱动的 LUAD 小鼠模型、LUAD 患者衍生的器官组织(PDO)和 LUAD 细胞系探讨 MRPL12 的表达和功能。通过质谱分析了MRPL12的翻译后修饰,并在体内和体外验证了MRPL12关键磷酸化位点在LUAD发展中的致癌作用:结果:MRPL12在人类LUAD组织、Tp53fl/fl;KrasG12D驱动的小鼠LUAD组织、LUAD PDO和LUAD细胞系中上调,与患者生存率低相关。MRPL12的过表达能显著促进LUAD肿瘤发生、转移和PDO的形成,而MRPL12的敲除则会引起相反的表型。此外,在Tp53fl/fl;KrasG12D驱动的小鼠LUAD模型中,MRPL12缺失会带来明显的生存优势,延缓肿瘤的发生并减少恶性进展。从机理上讲,我们发现 MRPL12 通过上调线粒体氧化磷酸化促进肿瘤进展。此外,我们还发现 UBASH3B 是 MRPL12 的特异性粘合剂,能使 MRPL12 中的酪氨酸 60(MRPL12 Y60)去磷酸化,抑制其致癌功能。MRPL12 Y60磷酸化的减少阻碍了MRPL12与POLRMT的结合,从而下调了LUAD细胞的线粒体代谢。深入的体内、体外和类器官模型验证了MRPL12 Y60突变对LUAD的抑制作用:这项研究证实,MRPL12是LUAD中的一种新型癌基因,它通过协调线粒体代谢重编程,使其向氧化磷酸化(OXPHOS)方向发展,从而促进了LUAD的发病机制。此外,研究还证实Y60是调节MRPL12致癌功能的特异性磷酸化修饰位点,为开发LUAD特异性靶向药物和临床干预提供了启示。
{"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 Q1 ONCOLOGY 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

Background: 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.

Methods: 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.

Results: 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.

Conclusions: Our results suggest that lncOCMRL1 is a reliable target for blocking lymph node metastasis in OSCC.

背景:长非编码RNA(lncRNA)广泛参与癌症的发生和发展,但大多数lncRNA的功能尚未阐明。转移是制约包括口腔鳞状细胞癌(OSCC)在内的各种癌症治疗效果的主要因素。因此,探索调控OSCC转移的关键lncRNA并阐明其分子机制将有助于开发有效治疗OSCC的新策略:我们分析了有和无颈淋巴结转移的OSCC患者肿瘤组织以及OSCC细胞系的lncRNA表达谱。我们发现,在有淋巴结转移的OSCC患者肿瘤组织和高度转移的OSCC细胞系中,口腔鳞状细胞癌转移相关lncRNA 1(lncOCMRL1)的表达量很高。通过qRT-PCR、Transwell、集落形成和细胞增殖实验探讨了敲除lncOCMRL1对OSCC细胞侵袭、迁移和增殖能力的影响。通过 RNA pull-down、银染色、质谱分析、RIP 和 WB 实验探讨了 lncOCMRL1 促进 OSCC 转移和增殖的机制。为了提高其转化潜力,我们开发了一种还原反应纳米递送系统来递送 siRNA 用于抗肿瘤治疗:我们发现lncOCMRL1在OSCC转移性肿瘤组织和细胞中高表达。功能研究表明,lncOCMRL1的高表达可促进OSCC细胞在体内和体外的生长和转移。从机理上讲,lncOCMRL1可通过抑制RRM2泛素化诱导上皮-间质转化(EMT),从而促进OSCC细胞的增殖、侵袭和迁移。我们进一步构建了还原反应纳米颗粒(NPs),用于全身性递送靶向lncOCMRL1的siRNAs,并证明它们能高效抑制lncOCMRL1在体内的表达,显著抑制OSCC肿瘤的生长和转移:我们的研究结果表明,lncOCMRL1是阻断OSCC淋巴结转移的可靠靶点。
{"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 Q1 ONCOLOGY 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

Background: 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.

Methods: 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.

Results: 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.

Conclusion: 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.

背景:胆管癌(CCA)是一种高度恶性、进展迅速的胆管肿瘤。由于具有化疗耐药性,它的预后一直极差。因此,仍需详细阐明化疗耐药机制并确定治疗靶点:方法:我们利用公共数据库和免疫组织化学(IHC)分析了 MBD2(甲基-CpG 结合域 2)在 CCA 和正常胆管组织中的表达。我们通过 CCK-8、平板克隆试验、伤口愈合试验和异种移植小鼠模型验证了 MBD2 在 CCA 细胞增殖、迁移和化疗耐受能力中的作用。此外,我们还构建了原代 CCA 小鼠模型,以进一步证实 MBD2 的作用。RNA-seq和co-IP-MS用于鉴定MBD2导致化疗耐药性的机制:结果:MBD2在CCA中上调。结果:MBD2在CCA中上调,促进了CCA细胞的增殖、迁移和化疗耐受性。从机理上讲,MBD2与WDR5直接相互作用,结合到ABCB1的启动子上,通过KMT2A促进该区域H3K4的三甲基化,激活ABCB1的表达。敲除 WDR5 或 KMT2A 会阻止 MBD2 对 ABCB1 的转录激活。分子抑制剂 MM-102 针对的是 WDR5 与 KMT2A 的相互作用。MM-102抑制了CCA细胞中ABCB1的表达,降低了CCA对顺铂的化疗耐药性:结论:MBD2通过与WDR5相互作用促进CCA的进展和化疗耐药性。结论:MBD2通过与WDR5的相互作用促进CCA的进展和化疗耐受性,MM-102能有效阻断这一过程,提高CCA对顺铂的敏感性。
{"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 Q1 ONCOLOGY 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

Background: 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.

Methods: 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-Prkdcem26Cd52Il2rgem26Cd22/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.

Results: 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.

Conclusions: 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.

背景:线粒体呼吸嵴(MRC)的动态及其对氧化磷酸化(OXPHOS)的影响在推动高级别胶质瘤(HGG)的进展中起着至关重要的作用。然而,其潜在机制仍不清楚:在本研究中,我们采用基于机器学习的透射电子显微镜分析了 54 例切除胶质瘤患者的 7141 个线粒体。此外,我们还对临床胶质瘤芯片进行了生物信息学分析和多重免疫组化(mIHC)染色,以确定参与胶质瘤的关键分子。随后,我们通过慢病毒转染调节了线粒体动态-1样蛋白(DNM1L/DRP1)及其两种受体线粒体裂变蛋白1(FIS1)和线粒体裂变因子(MFF)的表达水平,进一步研究了这些分子在胶质母细胞瘤(GBM)细胞和胶质瘤干细胞(GSCs)动态变化中的核心作用。然后,我们采用 CCK-8 试验、Transwell 试验、伤口愈合试验、肿瘤球形成试验和细胞衍生异种移植试验(NOD/ShiLtJGpt-Prkdcem26Cd52Il2rgem26Cd22/Gpt (NCG)小鼠模型),评估了 DNM1L/DRP1、FIS1 和 MFF 对 GBM 细胞和 GSC 增殖和进展的潜在影响。随后,我们利用海马 XF 分析技术、线粒体功能检测、MRC 丰度检测和有丝分裂动态变化监测,验证了 DNM1L/DRP1-FIS1 轴通过有丝分裂重塑 MRC 结构的能力:我们的研究结果表明,与低级别胶质瘤(LGG)相比,HGG表现出更完整的MRC结构。进一步的研究发现,DNM1L/DRP1、FIS1和MFF在HGG的线粒体分裂和MRC重塑中起着关键作用。随后的验证表明,DNM1L/DRP1 对 FIS1 具有正向调控作用,而 MFF 与 FIS1 之间的相互作用则表现出竞争性抑制关系。DNM1L/DRP1-FIS1轴的下调会显著损害有丝分裂,从而阻碍MRC的重塑,抑制胶质瘤的OXPHOS功能,最终导致抑制其侵袭性进展。与此相反,MFF对MRC的完整性、OXPHOS活性和胶质瘤的进展产生了相反的影响:本研究强调,DNM1L/DRP1-FIS1 轴通过 HGG 细胞中的有丝分裂来稳定 MRC 结构,同时驱动其 OXPHOS 活性,最终导致疾病的强劲进展。抑制 DNM1L/DRP1-FIS1 轴会阻碍 MRC 重塑并抑制 GBM 的进展。我们认为,下调 DNM1L/DRP1-FIS1 轴可能是治疗 HGG 的一种潜在治疗策略。
{"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 Q1 ONCOLOGY 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

Background: 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+ 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.

Methods: 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.

Results: 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+ 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.

Conclusions: 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.

背景:胰腺导管腺癌肿瘤对化疗、靶向疗法甚至免疫疗法均表现出抗药性。树突状细胞利用葡萄糖支持其效应功能,并通过促进细胞毒性 CD8+ T 细胞活性在抗肿瘤免疫中发挥关键作用。然而,葡萄糖和乳酸盐水平对胰腺导管腺癌树突状细胞的影响尚不清楚。本研究旨在阐明葡萄糖和乳酸盐如何影响树突状细胞的抗原递呈功能,并阐明相关机制:方法:使用患者衍生的器官组织和切除标本评估了胰腺导管腺癌中的糖代谢活性和免疫细胞浸润。从 KPC 小鼠身上建立了糖酵解增加或减少的细胞系。流式细胞术和单细胞 RNA 测序被用来评估对肿瘤微环境的影响。流式细胞术检测了葡萄糖和乳酸盐对骨髓树突状细胞抗原递呈功能的影响:结果:胰腺导管腺癌肿瘤微环境表现出低葡萄糖和高乳酸盐浓度,这与癌细胞不同程度的糖酵解活性有关。在小鼠移植模型中,糖酵解增加的肿瘤显示髓源性抑制细胞浸润增强,树突状细胞和 CD8+ T 细胞浸润减少,而糖酵解减少的肿瘤则显示相反的趋势。在三维共培养中,癌细胞中糖酵解增加会抑制骨髓树突状细胞的抗原递呈功能。此外,低葡萄糖和高乳酸盐培养基抑制了骨髓树突状细胞的抗原递呈和线粒体功能:我们的研究证明了动态糖酵解重编程对胰腺导管腺癌肿瘤微环境中免疫细胞组成的影响,尤其是对树突状细胞抗原递呈功能的影响。
{"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 Q1 ONCOLOGY 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

Background: 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.

Methods: 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.

Result: 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.

Conclusion: 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.

背景:绝大多数胰腺癌已被证明对单药剂免疫疗法不敏感。探索免疫耐受的机制并实施联合治疗策略对于PDAC患者从免疫疗法中获益至关重要。约27%的PDAC患者存在BAP1缺失,且与预后不良显著相关,但BAP1缺失如何影响PDAC患者生存的机制仍是一个谜:方法:应用BAP1基因敲除KPC(KrasG12D/+; LSLTrp53R172H/+; Pdx-1-Cre)小鼠和对照KPC小鼠、合成异种移植模型分析BAP1与PDAC免疫治疗反应的相关性。免疫沉淀、RT-qPCR、荧光素酶和转录组分析相结合,揭示了潜在的机制。研究人员构建了合成异种移植模型和流式细胞术,以检测SIRT1抑制剂的疗效及其与抗PD-1疗法的协同作用:结果:BAP1的缺失导致了PDAC对免疫疗法的耐药性,这是由于BAP1抑制了HSF1的转录活性。具体来说,BAP1与SIRT1竞争结合到K80乙酰化的HSF1上。BAP1 与 HSF1 的相互作用保留了 HSF1-K80 的乙酰化,并促进了 HSF1-HSP70 的相互作用,从而促进了 HSF1 的寡聚化和与染色质的分离。此外,我们还证明了靶向抑制 SIRT1 可逆转 BAP1 缺乏的 PDAC 小鼠模型的免疫不敏感性:我们的研究阐明了 BAP1 通过抑制 HSF1 调节 PDAC 免疫治疗反应的一种尚未揭示的机制,并为解决 BAP1 缺乏型 PDAC 的免疫不敏感问题提供了有前景的治疗策略。
{"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 Q1 ONCOLOGY 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

Background: 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.

Methods: 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+ 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.

Results: 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+ T cells in the tumor microenvironment.

Conclusions: Our findings indicate that MED1 plays a pivotal role in OSCC progression through the activation of MMP9 transcription and suppression of CD8+ T cell antitumor immunity, suggesting that MED1 may serve as a novel prognostic marker and therapeutic target in OSCC.

背景:Mediator complex subunit 1 (MED1)是一种关键的转录辅激活因子,与多种生物通路有关,但它在口腔鳞状细胞癌(OSCC)中的作用仍有待探索。本研究旨在阐明 MED1 对 OSCC 进展的作用机制和潜在影响:方法:通过生物信息学分析评估MED1在OSCC组织中的表达和临床意义。方法:通过生物信息学分析评估了MED1在OSCC组织中的表达和临床意义,并在体外和体内评估了MED1对OSCC癌细胞生物学行为的影响。研究采用了双荧光素酶报告实验、染色质免疫沉淀(ChIP)实验、生物信息学分析、CD8+ T细胞分离实验、共培养实验、酶联免疫吸附实验(ELISA)和流式细胞分析等方法,以阐明MED1在OSCC进展过程中的潜在作用机制:结果:MED1在OSCC组织和多种OSCC细胞系中均表现出上调,这与患者总生存率的下降有关。体外实验表明,在转移性 OSCC 细胞系 SCC-9 和 UPCI-SCC-154 中敲除 MED1 会阻碍细胞迁移和侵袭,而过表达 MED1 则会促进这些过程。而敲除 MED1 对上述细胞株的增殖没有影响。体内研究进一步发现,下调 MED1 能有效抑制 OSCC 的远处转移。从机理上讲,MED1增强了转录因子c-Jun和c-Fos与基质金属蛋白9(MMP9)启动子的结合,导致MMP9转录显著上调。这一过程有助于 SCC-9 和 UPCI-SCC-154 细胞的迁移和侵袭。此外,MED1还通过Notch信号通路调节程序性死亡配体1(PD-L1)的表达,从而影响肿瘤微环境中CD8+ T细胞的肿瘤杀伤能力:我们的研究结果表明,MED1通过激活MMP9转录和抑制CD8+ T细胞抗肿瘤免疫在OSCC的进展中起着关键作用,这表明MED1可作为OSCC的新型预后标志物和治疗靶点。
{"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 Q1 ONCOLOGY 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

Background: 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.

Methods: 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.

Results: 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.

Conclusions: 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.

背景:致癌表皮生长因子受体(EGFR)突变是肺腺癌(LUAD)恶性化的重要原因。人们已经知道,代谢重编程是恶性肿瘤的一个重要标志,而嘌呤代谢是肿瘤进展和耐药性的一个关键代谢途径,但其与表皮生长因子受体突变型肺腺癌(LUAD)的关系尚不清楚:方法:通过基于毛细管电泳-飞行时间质谱(CE-TOF/MS)的代谢谱分析研究了代谢重编程。体外细胞增殖通过EdU染色和细胞周期测定进行评估。体内致瘤性通过裸鼠皮下肿瘤形成实验进行检测。缺氧诱导因子-1α(HIF-1α)和次黄嘌呤磷酸核糖基转移酶1(HPRT1)的结合通过DNA牵引试验和染色质免疫沉淀(ChIP)试验进行检测。通过 Western 印迹检测了 HIF-1α、HPRT1、DNA 损伤和细胞凋亡相关基因。此外,还进行了 RNA 测序、质谱分析和生物信息学分析:结果:我们发现,突变的表皮生长因子受体(muEGFR)上调HPRT1,促进表皮生长因子受体突变的LUAD的嘌呤代谢和肿瘤发生。从机理上讲,muEGFR通过蛋白稳定性增加HIF-1α的表达。同时,上调的HIF-1α与HPRT1的启动子结合,转录激活HPRT1的表达,促进嘌呤代谢,从而维持EGFR突变型LUAD肿瘤细胞的快速增殖。此外,吉非替尼抑制了嘌呤核苷酸的合成,HPRT1抑制增加了吉非替尼对表皮生长因子受体突变型LUAD的敏感性:我们的研究揭示了muEGFR-HIF-1α-HPRT1轴在表皮生长因子受体突变型LUAD中的关键作用,并为抑制嘌呤代谢治疗表皮生长因子受体突变型LUAD提供了新策略。
{"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
期刊
Journal of Experimental & Clinical Cancer Research
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1