Background:Cancer cells selectively promote the translation of oncogenic tran-scripts to stimulate cancer progression.Although growing evidence has revealed that tRNA modifications and related genes participate in thi...Background:Cancer cells selectively promote the translation of oncogenic tran-scripts to stimulate cancer progression.Although growing evidence has revealed that tRNA modifications and related genes participate in this process,their roles in head and neck squamous cell carcinoma(HNSCC)remain largely unchar-acterized.Here,we sought to investigate the function and mechanisms of the transfer RNA(tRNA)N7-methylguanosine(m'G)modification in regulating the occurrence and development of HNSCC.Methods:Cell lost of-function and gain-of function assays,xenograft models,conditional knockout and knockin mouse models were used to study the physi-ological functions of tRNA m'G modification in HNSCC tumorigenesis.tRNA modification and expression profiling,mRNA translation profiling and res-cue assays were performed to uncover the underlying molecular mechanisms.Single-cell RNA sequencing(scRNA seq)was conducted to explore the tumor microenvironment changes.Results:The tRNA.m7G methyltransferase complex components Methyltransferase-like 1(METTL1)/WD repeat domain 4(WDR4)were upregulated in HNSCC and associated with a poor prognosis.Functionally,METTL1/WDR4 promoted HNSCC progression and metastasis in cell-based and transgenic mouse models.Mechanistically,ablation of METTL1 reduced the m'G levels of 16 tRNAS,inhibiting the translation of a subset of oncogenic transcripts,including genes related to the phosphatidylinositol-3-kinase/protein kinase B/mammalian target of rapamycin(PI3K/AKT/mTOR)signaling pathway.In addition,chemical modulators of the PI3K/Akt/mTOR signaling pathway reversed the effects of Mettll in mouse HNSCC.Furthermore,scRNA-seq results revealed that Mettll knockout in mouse tumor cells altered the immune landscape and cell-cell interaction between the tumor and stromal compartment.Conclusions:The tRNA m?G methyltransferase METTLI was found to promote the development and malignancy of HNSCC through regulating global mRNA translation,including the PI3K/AKT/mTOR signaling pathway,and found to alter immune landscape.METTLI could be a promising treatment target for HNSCC patients.展开更多
目的探讨m^(7)G相关基因能否作为肝细胞性肝癌预后的生物标志物。方法采用癌症基因组图谱(The Cancer Genome Atlas,TCGA)数据库筛选肝细胞性肝癌组织和癌旁组织中有表达差异的m^(7)G相关基因组,m^(7)G相关基因和临床数据中的生存时间...目的探讨m^(7)G相关基因能否作为肝细胞性肝癌预后的生物标志物。方法采用癌症基因组图谱(The Cancer Genome Atlas,TCGA)数据库筛选肝细胞性肝癌组织和癌旁组织中有表达差异的m^(7)G相关基因组,m^(7)G相关基因和临床数据中的生存时间及生存状态按照样本ID号匹配合并后筛选预后基因组,将两组的交集基因纳入lasso回归,对筛选出来的基因进行风险评分,基于风险评分的中位数值将所有肝细胞性肝癌患者分为高、低两个风险组,并对高分险组和低风险组进行单因素和多因素的Cox回归分析,评价风险评分在预后中的差异。结果经lasso回归筛选出4个模型基因(AGO2、NCBP1、NCBP2、WDR4),高风险组的生存率显著低于低风险组(P=0.027),ROC曲线显示风险模型对患者1,2,3年生存预测的曲线下面积(AUC)分别为0.683,0.604,0.602。肿瘤分期、T分期、M分期和风险评分是肝细胞性肝癌预后的相关因素(P<0.05),其中风险评分是影响肝细胞性肝癌患者生存率的独立预后因素(P=0.044,HR=1.637)。结论m^(7)G相关基因可作为肝细胞性肝癌预后的生物标志物。AGO2、NCBP1、NCBP2和WDR4构建的风险模型对肝细胞性肝癌具有重要的预后价值。展开更多
Background:Methyltransferase 3(METTL3)-mediated N6-methyladenosine(m^(6)A)RNA modification has been demonstrated to be a potential factor in promoting gastric cancer(GC).METTL3 regulates a series of signaling pathways...Background:Methyltransferase 3(METTL3)-mediated N6-methyladenosine(m^(6)A)RNA modification has been demonstrated to be a potential factor in promoting gastric cancer(GC).METTL3 regulates a series of signaling pathways by modifying various mRNAs.This study aimed to identify novel METTL3-mediated signaling pathways and explored possible targets for use in the clinical setting of gastric cancer.Methods:To investigate the proliferation and metastatic capacity ofGCcell lines with METTL3 knockdown,a xenograft,lung metastasis,and popliteal lymph node metastasis model was used.Them^(6)A-modified RNA immunoprecipitation(Me-RIP)sequence was utilized to explore the target mRNAs of METTL3.Cell counting kit 8 and transwell assays were performed to investigate the promoting function of pre-B cell leukemia homeobox 1(PBX1)and GTP cyclohydrolase 1(GCH1).Western blotting and chromatin immunoprecipitation were employed to confirm the involvement of the METTL3-PBX1-GCH1 axis.ELISA and liquid chromatography-mass spectrometry were used to explore the biological function of tetrahydrobiopterin(BH_(4)).Results:Knockdown of METTL3 suppressed xenograft tumor growth and lung/lymph node metastasis in vivo.Mechanistically,we found that METTL3 combined with and stabilized PBX1 mRNAs.Chromatin immunoprecipitation(ChIP)and further experiments suggested that PBX1 acted as a transcription factor inducing GCH1 expression.Moreover,the METTL3-PBX1-GCH1 axis increased BH_(4)levels in GC cells,thereby promoting tumor progression.Conclusions:This study suggested that METTL3 enzymes promote tumor growth and lung/lymph node metastasis via METTL3-PBX1-GCH1 axis increasing BH_(4)levels in GC.展开更多
基金National Natural Science Foundation of China,Grant/Award Numbers:81872409,82173362Natural Science Foundation of Guangdong Province,Grant/Award Numbers:2018A030313610,2019A1515110110,2020A1515010291+1 种基金The Open Funding of the State Key Laboratory of Oral Diseases,Grant/Award Number:SKLOD2021OF02and the use was approved by the Institutional Review Board of the First Affiliated Hospital of Sun Yat-Sen University(2016074).The nude mouse experi-ments performed were approved by the Laboratory Ani-mal Center of Sun Yat-SenUniversity(SYSU-IACUC-2021-000092).The transgenic mouse experiments performed were approved by the Laboratory Animal Center of Sun Yat-Sen University(SYSU-IACUC-2020-000569).
文摘Background:Cancer cells selectively promote the translation of oncogenic tran-scripts to stimulate cancer progression.Although growing evidence has revealed that tRNA modifications and related genes participate in this process,their roles in head and neck squamous cell carcinoma(HNSCC)remain largely unchar-acterized.Here,we sought to investigate the function and mechanisms of the transfer RNA(tRNA)N7-methylguanosine(m'G)modification in regulating the occurrence and development of HNSCC.Methods:Cell lost of-function and gain-of function assays,xenograft models,conditional knockout and knockin mouse models were used to study the physi-ological functions of tRNA m'G modification in HNSCC tumorigenesis.tRNA modification and expression profiling,mRNA translation profiling and res-cue assays were performed to uncover the underlying molecular mechanisms.Single-cell RNA sequencing(scRNA seq)was conducted to explore the tumor microenvironment changes.Results:The tRNA.m7G methyltransferase complex components Methyltransferase-like 1(METTL1)/WD repeat domain 4(WDR4)were upregulated in HNSCC and associated with a poor prognosis.Functionally,METTL1/WDR4 promoted HNSCC progression and metastasis in cell-based and transgenic mouse models.Mechanistically,ablation of METTL1 reduced the m'G levels of 16 tRNAS,inhibiting the translation of a subset of oncogenic transcripts,including genes related to the phosphatidylinositol-3-kinase/protein kinase B/mammalian target of rapamycin(PI3K/AKT/mTOR)signaling pathway.In addition,chemical modulators of the PI3K/Akt/mTOR signaling pathway reversed the effects of Mettll in mouse HNSCC.Furthermore,scRNA-seq results revealed that Mettll knockout in mouse tumor cells altered the immune landscape and cell-cell interaction between the tumor and stromal compartment.Conclusions:The tRNA m?G methyltransferase METTLI was found to promote the development and malignancy of HNSCC through regulating global mRNA translation,including the PI3K/AKT/mTOR signaling pathway,and found to alter immune landscape.METTLI could be a promising treatment target for HNSCC patients.
文摘目的探讨m^(7)G相关基因能否作为肝细胞性肝癌预后的生物标志物。方法采用癌症基因组图谱(The Cancer Genome Atlas,TCGA)数据库筛选肝细胞性肝癌组织和癌旁组织中有表达差异的m^(7)G相关基因组,m^(7)G相关基因和临床数据中的生存时间及生存状态按照样本ID号匹配合并后筛选预后基因组,将两组的交集基因纳入lasso回归,对筛选出来的基因进行风险评分,基于风险评分的中位数值将所有肝细胞性肝癌患者分为高、低两个风险组,并对高分险组和低风险组进行单因素和多因素的Cox回归分析,评价风险评分在预后中的差异。结果经lasso回归筛选出4个模型基因(AGO2、NCBP1、NCBP2、WDR4),高风险组的生存率显著低于低风险组(P=0.027),ROC曲线显示风险模型对患者1,2,3年生存预测的曲线下面积(AUC)分别为0.683,0.604,0.602。肿瘤分期、T分期、M分期和风险评分是肝细胞性肝癌预后的相关因素(P<0.05),其中风险评分是影响肝细胞性肝癌患者生存率的独立预后因素(P=0.044,HR=1.637)。结论m^(7)G相关基因可作为肝细胞性肝癌预后的生物标志物。AGO2、NCBP1、NCBP2和WDR4构建的风险模型对肝细胞性肝癌具有重要的预后价值。
基金Natural Science Foundation of Guangdong Province,Grant/Award Numbers:2018A030313634,2020A1515010214,2021A1515010473Guangdong Basic and Applied Basic Research Foundation,Grant/Award Numbers:2020A1515010214,2018A030313634,2021A1515010473+1 种基金Young Teacher Foundation of Sun Yat-sen University,Grant/Award Number:19ykpy58China Postdoctoral Science Foundation,Grant/Award Number:2020M683087。
文摘Background:Methyltransferase 3(METTL3)-mediated N6-methyladenosine(m^(6)A)RNA modification has been demonstrated to be a potential factor in promoting gastric cancer(GC).METTL3 regulates a series of signaling pathways by modifying various mRNAs.This study aimed to identify novel METTL3-mediated signaling pathways and explored possible targets for use in the clinical setting of gastric cancer.Methods:To investigate the proliferation and metastatic capacity ofGCcell lines with METTL3 knockdown,a xenograft,lung metastasis,and popliteal lymph node metastasis model was used.Them^(6)A-modified RNA immunoprecipitation(Me-RIP)sequence was utilized to explore the target mRNAs of METTL3.Cell counting kit 8 and transwell assays were performed to investigate the promoting function of pre-B cell leukemia homeobox 1(PBX1)and GTP cyclohydrolase 1(GCH1).Western blotting and chromatin immunoprecipitation were employed to confirm the involvement of the METTL3-PBX1-GCH1 axis.ELISA and liquid chromatography-mass spectrometry were used to explore the biological function of tetrahydrobiopterin(BH_(4)).Results:Knockdown of METTL3 suppressed xenograft tumor growth and lung/lymph node metastasis in vivo.Mechanistically,we found that METTL3 combined with and stabilized PBX1 mRNAs.Chromatin immunoprecipitation(ChIP)and further experiments suggested that PBX1 acted as a transcription factor inducing GCH1 expression.Moreover,the METTL3-PBX1-GCH1 axis increased BH_(4)levels in GC cells,thereby promoting tumor progression.Conclusions:This study suggested that METTL3 enzymes promote tumor growth and lung/lymph node metastasis via METTL3-PBX1-GCH1 axis increasing BH_(4)levels in GC.