首页 > 最新文献

Oncogenesis最新文献

英文 中文
Transcriptomic analysis identifies B-lymphocyte kinase as a therapeutic target for desmoplastic small round cell tumor cancer stem cell-like cells. 转录组分析发现B淋巴细胞激酶是去瘤小圆细胞瘤干细胞样细胞的治疗靶点。
IF 6.2 2区 医学 Q1 ONCOLOGY Pub Date : 2024-01-04 DOI: 10.1038/s41389-023-00504-z
Justin W Magrath, Dane A Flinchum, Alifiani B Hartono, Shruthi Sanjitha Sampath, Tina M O'Grady, Melody Baddoo, Liang Haoyang, Xiaojiang Xu, Erik K Flemington, Sean B Lee

Desmoplastic small round cell tumor (DSRCT) is an aggressive pediatric cancer caused by the EWSR1-WT1 fusion oncoprotein. The tumor is refractory to treatment with a 5-year survival rate of only 15-25%, necessitating the development of novel therapeutics, especially those able to target chemoresistant subpopulations. Novel in vitro cancer stem cell-like (CSC-like) culture conditions increase the expression of stemness markers (SOX2, NANOG) and reduce DSRCT cell line susceptibility to chemotherapy while maintaining the ability of DSRCT cells to form xenografts. To gain insights into this chemoresistant model, RNA-seq was performed to elucidate transcriptional alterations between DSRCT cells grown in CSC-like spheres and normal 2-dimensional adherent state. Commonly upregulated and downregulated genes were identified and utilized in pathway analysis revealing upregulation of pathways related to chromatin assembly and disassembly and downregulation of pathways including cell junction assembly and extracellular matrix organization. Alterations in chromatin assembly suggest a role for epigenetics in the DSRCT CSC-like state, which was further investigated with ATAC-seq, identifying over 10,000 differentially accessible peaks, including 4444 sphere accessible peaks and 6,120 adherent accessible peaks. Accessible regions were associated with higher gene expression, including increased accessibility of the CSC marker SOX2 in CSC-like culture conditions. These analyses were further utilized to identify potential CSC therapeutic targets, leading to the identification of B-lymphocyte kinase (BLK) as a CSC-enriched, EWSR1-WT1-regulated, druggable target. BLK inhibition and knockdown reduced CSC-like properties, including abrogation of tumorsphere formation and stemness marker expression. Importantly, BLK knockdown reduced DSRCT CSC-like cell chemoresistance, making its inhibition a promising target for future combination therapy.

脱屑性小圆形细胞瘤(DSRCT)是一种由 EWSR1-WT1 融合肿瘤蛋白引起的侵袭性小儿癌症。这种肿瘤难治,5年生存率仅为15%-25%,因此有必要开发新型疗法,尤其是针对化疗耐药亚群的疗法。新型体外癌症干细胞样(CSC-like)培养条件增加了干性标志物(SOX2、NANOG)的表达,降低了DSRCT细胞系对化疗的敏感性,同时保持了DSRCT细胞形成异种移植的能力。为了深入了解这种化疗耐药模型,我们进行了 RNA-seq 研究,以阐明 DSRCT 细胞在 CSC 样球状生长和正常二维粘附状态下的转录变化。研究人员确定了常见的上调和下调基因,并将其用于通路分析,结果显示与染色质组装和分解相关的通路上调,而包括细胞连接组装和细胞外基质组织在内的通路下调。染色质组装的改变表明表观遗传学在DSRCT CSC样状态中的作用,ATAC-seq对此进行了进一步研究,确定了超过10,000个不同的可访问峰,包括4444个球状可访问峰和6,120个粘附可访问峰。可访问区域与较高的基因表达相关,包括在 CSC 样培养条件下 CSC 标记 SOX2 的可访问性增加。我们进一步利用这些分析来确定潜在的 CSC 治疗靶点,最终确定了 B 淋巴细胞激酶(BLK)作为 CSC 富集的、受 EWSR1-WT1 调节的药物靶点。抑制和敲除BLK可减少类似CSC的特性,包括肿瘤球的形成和干性标志物的表达。重要的是,BLK基因敲除降低了DSRCT CSC样细胞的化疗耐药性,使其抑制成为未来有希望的联合疗法靶点。
{"title":"Transcriptomic analysis identifies B-lymphocyte kinase as a therapeutic target for desmoplastic small round cell tumor cancer stem cell-like cells.","authors":"Justin W Magrath, Dane A Flinchum, Alifiani B Hartono, Shruthi Sanjitha Sampath, Tina M O'Grady, Melody Baddoo, Liang Haoyang, Xiaojiang Xu, Erik K Flemington, Sean B Lee","doi":"10.1038/s41389-023-00504-z","DOIUrl":"10.1038/s41389-023-00504-z","url":null,"abstract":"<p><p>Desmoplastic small round cell tumor (DSRCT) is an aggressive pediatric cancer caused by the EWSR1-WT1 fusion oncoprotein. The tumor is refractory to treatment with a 5-year survival rate of only 15-25%, necessitating the development of novel therapeutics, especially those able to target chemoresistant subpopulations. Novel in vitro cancer stem cell-like (CSC-like) culture conditions increase the expression of stemness markers (SOX2, NANOG) and reduce DSRCT cell line susceptibility to chemotherapy while maintaining the ability of DSRCT cells to form xenografts. To gain insights into this chemoresistant model, RNA-seq was performed to elucidate transcriptional alterations between DSRCT cells grown in CSC-like spheres and normal 2-dimensional adherent state. Commonly upregulated and downregulated genes were identified and utilized in pathway analysis revealing upregulation of pathways related to chromatin assembly and disassembly and downregulation of pathways including cell junction assembly and extracellular matrix organization. Alterations in chromatin assembly suggest a role for epigenetics in the DSRCT CSC-like state, which was further investigated with ATAC-seq, identifying over 10,000 differentially accessible peaks, including 4444 sphere accessible peaks and 6,120 adherent accessible peaks. Accessible regions were associated with higher gene expression, including increased accessibility of the CSC marker SOX2 in CSC-like culture conditions. These analyses were further utilized to identify potential CSC therapeutic targets, leading to the identification of B-lymphocyte kinase (BLK) as a CSC-enriched, EWSR1-WT1-regulated, druggable target. BLK inhibition and knockdown reduced CSC-like properties, including abrogation of tumorsphere formation and stemness marker expression. Importantly, BLK knockdown reduced DSRCT CSC-like cell chemoresistance, making its inhibition a promising target for future combination therapy.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":"13 1","pages":"2"},"PeriodicalIF":6.2,"publicationDate":"2024-01-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10767073/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139098374","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cell competition and cancer from Drosophila to mammals. 从果蝇到哺乳动物的细胞竞争与癌症
IF 6.2 2区 医学 Q1 ONCOLOGY Pub Date : 2024-01-03 DOI: 10.1038/s41389-023-00505-y
Bojie Cong, Ross L Cagan

Throughout an individual's life, somatic cells acquire cancer-associated mutations. A fraction of these mutations trigger tumour formation, a phenomenon partly driven by the interplay of mutant and wild-type cell clones competing for dominance; conversely, other mutations function against tumour initiation. This mechanism of 'cell competition', can shift clone dynamics by evaluating the relative status of clonal populations, promoting 'winners' and eliminating 'losers'. This review examines the role of cell competition in the context of tumorigenesis, tumour progression and therapeutic intervention.

在人的一生中,体细胞会发生与癌症相关的突变。其中一部分突变会诱发肿瘤的形成,这种现象部分是由突变型细胞克隆和野生型细胞克隆争夺优势地位的相互作用所驱动的;反之,其他突变则对肿瘤的形成起抑制作用。这种 "细胞竞争 "机制可以通过评估克隆群体的相对地位来改变克隆动态,促进 "赢家",淘汰 "输家"。本综述探讨了细胞竞争在肿瘤发生、肿瘤进展和治疗干预中的作用。
{"title":"Cell competition and cancer from Drosophila to mammals.","authors":"Bojie Cong, Ross L Cagan","doi":"10.1038/s41389-023-00505-y","DOIUrl":"10.1038/s41389-023-00505-y","url":null,"abstract":"<p><p>Throughout an individual's life, somatic cells acquire cancer-associated mutations. A fraction of these mutations trigger tumour formation, a phenomenon partly driven by the interplay of mutant and wild-type cell clones competing for dominance; conversely, other mutations function against tumour initiation. This mechanism of 'cell competition', can shift clone dynamics by evaluating the relative status of clonal populations, promoting 'winners' and eliminating 'losers'. This review examines the role of cell competition in the context of tumorigenesis, tumour progression and therapeutic intervention.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":"13 1","pages":"1"},"PeriodicalIF":6.2,"publicationDate":"2024-01-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10764339/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139087937","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Programmed cell death 11 modulates but not entirely relies on p53-HDM2 loop to facilitate G2/M transition in colorectal cancer cells 程序性细胞死亡 11 可调节但并非完全依赖 p53-HDM2 环路来促进结直肠癌细胞的 G2/M 转变
IF 6.2 2区 医学 Q1 ONCOLOGY Pub Date : 2023-12-07 DOI: 10.1038/s41389-023-00501-2
Li Ding, Yujie Xu, Lin Xu, Chenhong Zhao, Zhiping Zhang, Jie Zhang, Kai Liao, Yuerou Chen, Jingwen Li, Xinyu Mei, Xinyue Zhang

We previously described a nucleolar protein RSL1D1 but distributed throughout the nucleus in HCT116 colorectal cancer (CRC) cells to facilitate G1/S transition by inhibiting p53 signaling. Here, we found another nucleolar protein, programmed cell death 11 (PDCD11), also with an “Extra-nucleolar” localization in CRC cells but to regulate G2/M checkpoint. This protein directly interacts with p53 and HDM2 in the nucleoplasm, thereby recruiting p53 to HDM2 for ubiquitination and degradation. The ensuing downregulation of p53 increases the CDK1 level to help the cells pass G2/M checkpoint. Upon DNA damage stress, PDCD11 gains the power to upregulate CDK1 independently of p53. Beyond these, PDCD11 also upregulates CDC25C in a p53-independent manner to dephosphorylate CDK1 to facilitate G2/M transition. Downregulation of PDCD11 greatly reduced cancer cell growth in vitro and in vivo, additionally sensitized cells to DNA damage signals, highlighting that PDCD11 is a crucial driving factor of CRC and a potential target for cancer treatment.

我们以前曾描述过一种核极蛋白 RSL1D1,它分布在 HCT116 大肠癌(CRC)细胞的整个细胞核中,通过抑制 p53 信号传导促进 G1/S 转换。在这里,我们发现了另一种核极蛋白--程序性细胞死亡 11(PDCD11),它在 CRC 细胞中也有 "核外 "定位,但却能调节 G2/M 检查点。该蛋白直接与核质中的 p53 和 HDM2 相互作用,从而将 p53 募集到 HDM2 上进行泛素化和降解。随之而来的 p53 下调会增加 CDK1 的水平,从而帮助细胞通过 G2/M 检查点。当出现 DNA 损伤应激时,PDCD11 能够独立于 p53 上调 CDK1。除此之外,PDCD11 还能以不依赖 p53 的方式上调 CDC25C,使 CDK1 去磷酸化,从而促进 G2/M 过渡。下调PDCD11可大大降低癌细胞在体外和体内的生长,并使细胞对DNA损伤信号敏感,这表明PDCD11是CRC的一个重要驱动因子,也是癌症治疗的一个潜在靶点。
{"title":"Programmed cell death 11 modulates but not entirely relies on p53-HDM2 loop to facilitate G2/M transition in colorectal cancer cells","authors":"Li Ding, Yujie Xu, Lin Xu, Chenhong Zhao, Zhiping Zhang, Jie Zhang, Kai Liao, Yuerou Chen, Jingwen Li, Xinyu Mei, Xinyue Zhang","doi":"10.1038/s41389-023-00501-2","DOIUrl":"https://doi.org/10.1038/s41389-023-00501-2","url":null,"abstract":"<p>We previously described a nucleolar protein RSL1D1 but distributed throughout the nucleus in HCT116 colorectal cancer (CRC) cells to facilitate G1/S transition by inhibiting p53 signaling. Here, we found another nucleolar protein, programmed cell death 11 (PDCD11), also with an “Extra-nucleolar” localization in CRC cells but to regulate G2/M checkpoint. This protein directly interacts with p53 and HDM2 in the nucleoplasm, thereby recruiting p53 to HDM2 for ubiquitination and degradation. The ensuing downregulation of p53 increases the CDK1 level to help the cells pass G2/M checkpoint. Upon DNA damage stress, PDCD11 gains the power to upregulate CDK1 independently of p53. Beyond these, PDCD11 also upregulates CDC25C in a p53-independent manner to dephosphorylate CDK1 to facilitate G2/M transition. Downregulation of PDCD11 greatly reduced cancer cell growth in vitro and in vivo, additionally sensitized cells to DNA damage signals, highlighting that PDCD11 is a crucial driving factor of CRC and a potential target for cancer treatment.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":"195 1","pages":""},"PeriodicalIF":6.2,"publicationDate":"2023-12-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138557084","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Identifying a locus in super-enhancer and its resident NFE2L1/MAFG as transcriptional factors that drive PD-L1 expression and immune evasion. 鉴定超级增强子中的一个位点及其驻留的NFE2L1/MAFG作为驱动PD-L1表达和免疫逃避的转录因子。
IF 6.2 2区 医学 Q1 ONCOLOGY Pub Date : 2023-11-20 DOI: 10.1038/s41389-023-00500-3
Conglin Shi, Liuting Chen, Hui Pi, Henglu Cui, Chenyang Fan, Fangzheng Tan, Xuanhao Qu, Rong Sun, Fengbo Zhao, Yihua Song, Yuanyuan Wu, Miaomiao Chen, Wenkai Ni, Lishuai Qu, Renfang Mao, Yihui Fan

Although the transcriptional regulation of the programmed death ligand 1 (PD-L1) promoter has been extensively studied, the transcription factor residing in the PD-L1 super-enhancer has not been comprehensively explored. Through saturated CRISPR-Cas9 screening of the core region of the PD-L1 super-enhancer, we have identified a crucial genetic locus, referred to as locus 22, which is essential for PD-L1 expression. Locus 22 is a potential binding site for NFE2:MAF transcription factors. Although genetic silencing of NRF2 (NFE2L2) did not result in a reduction of PD-L1 expression, further analysis reveals that MAFG and NFE2L1 (NRF1) play a critical role in the expression of PD-L1. Importantly, lipopolysaccharides (LPS) as the major component of intratumoral bacteria could greatly induce PD-L1 expression, which is dependent on the PD-L1 super-enhancer, locus 22, and NFE2L1/MAFG. Mechanistically, genetic modification of locus 22 and silencing of MAFG greatly reduce BRD4 binding and loop formation but have minimal effects on H3K27Ac modification. Unlike control cells, cells with genetic modification of locus 22 and silencing of NFE2L1/MAFG failed to escape T cell-mediated killing. In breast cancer, the expression of MAFG is positively correlated with the expression of PD-L1. Taken together, our findings demonstrate the critical role of locus 22 and its associated transcription factor NFE2L1/MAFG in super-enhancer- and LPS-induced PD-L1 expression. Our findings provide new insight into understanding the regulation of PD-L1 transcription and intratumoral bacteria-mediated immune evasion.

尽管程序性死亡配体1 (PD-L1)启动子的转录调控已被广泛研究,但驻留在PD-L1超增强子中的转录因子尚未被全面探索。通过饱和CRISPR-Cas9筛选PD-L1超增强子的核心区域,我们已经确定了一个至关重要的遗传位点,称为位点22,它对PD-L1的表达至关重要。基因座22是NFE2:MAF转录因子的潜在结合位点。虽然NRF2 (NFE2L2)的基因沉默不会导致PD-L1表达的降低,但进一步的分析表明,MAFG和NFE2L1 (NRF1)在PD-L1的表达中起着关键作用。重要的是,脂多糖(LPS)作为肿瘤内细菌的主要成分可以极大地诱导PD-L1的表达,而PD-L1的表达依赖于PD-L1超增强子、位点22和NFE2L1/MAFG。从机制上看,22号位点的遗传修饰和MAFG的沉默极大地减少了BRD4的结合和环的形成,但对H3K27Ac修饰的影响很小。与对照细胞不同,基因修改22位点并沉默NFE2L1/MAFG的细胞无法逃脱T细胞介导的杀伤。在乳腺癌中,MAFG的表达与PD-L1的表达呈正相关。综上所述,我们的研究结果证明了基因座22及其相关转录因子NFE2L1/MAFG在超增强子和脂多糖诱导的PD-L1表达中的关键作用。我们的发现为理解PD-L1转录调控和肿瘤内细菌介导的免疫逃避提供了新的见解。
{"title":"Identifying a locus in super-enhancer and its resident NFE2L1/MAFG as transcriptional factors that drive PD-L1 expression and immune evasion.","authors":"Conglin Shi, Liuting Chen, Hui Pi, Henglu Cui, Chenyang Fan, Fangzheng Tan, Xuanhao Qu, Rong Sun, Fengbo Zhao, Yihua Song, Yuanyuan Wu, Miaomiao Chen, Wenkai Ni, Lishuai Qu, Renfang Mao, Yihui Fan","doi":"10.1038/s41389-023-00500-3","DOIUrl":"10.1038/s41389-023-00500-3","url":null,"abstract":"<p><p>Although the transcriptional regulation of the programmed death ligand 1 (PD-L1) promoter has been extensively studied, the transcription factor residing in the PD-L1 super-enhancer has not been comprehensively explored. Through saturated CRISPR-Cas9 screening of the core region of the PD-L1 super-enhancer, we have identified a crucial genetic locus, referred to as locus 22, which is essential for PD-L1 expression. Locus 22 is a potential binding site for NFE2:MAF transcription factors. Although genetic silencing of NRF2 (NFE2L2) did not result in a reduction of PD-L1 expression, further analysis reveals that MAFG and NFE2L1 (NRF1) play a critical role in the expression of PD-L1. Importantly, lipopolysaccharides (LPS) as the major component of intratumoral bacteria could greatly induce PD-L1 expression, which is dependent on the PD-L1 super-enhancer, locus 22, and NFE2L1/MAFG. Mechanistically, genetic modification of locus 22 and silencing of MAFG greatly reduce BRD4 binding and loop formation but have minimal effects on H3K27Ac modification. Unlike control cells, cells with genetic modification of locus 22 and silencing of NFE2L1/MAFG failed to escape T cell-mediated killing. In breast cancer, the expression of MAFG is positively correlated with the expression of PD-L1. Taken together, our findings demonstrate the critical role of locus 22 and its associated transcription factor NFE2L1/MAFG in super-enhancer- and LPS-induced PD-L1 expression. Our findings provide new insight into understanding the regulation of PD-L1 transcription and intratumoral bacteria-mediated immune evasion.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":"12 1","pages":"56"},"PeriodicalIF":6.2,"publicationDate":"2023-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10662283/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138176964","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
NF-κB signaling activation and roles in thyroid cancers: implication of MAP3K14/NIK. NF-κB信号激活及其在甲状腺癌中的作用:MAP3K14/NIK的意义。
IF 6.2 2区 医学 Q1 ONCOLOGY Pub Date : 2023-11-16 DOI: 10.1038/s41389-023-00496-w
Françoise Cormier, Selma Housni, Florent Dumont, Mélodie Villard, Béatrix Cochand-Priollet, Françoise Mercier-Nomé, Karine Perlemoine, Jérôme Bertherat, Lionel Groussin

Among follicular-derived thyroid cancers (TC), those with aggressive behavior and resistance to current treatments display poor prognosis. NF-κB signaling pathways are involved in tumor progression of various cancers. Here, we finely characterize the NF-κB pathways and their involvement in TC. By using immunoblot and gel shift assays, we demonstrated that both classical and alternative NF-κB pathways are activated in ten TC-derived cell lines, leading to activated RelA/p50 and RelB/p50 NF-κB dimers. By analyzing the RNAseq data of the large papillary thyroid carcinoma (PTC) cohort from The Cancer Genome Atlas (TCGA) project, we identified a tumor progression-related NF-κB signature in BRAFV600E mutated-PTCs. That corroborated with the role of RelA and RelB in cell migration and invasion processes that we demonstrated specifically in BRAFV600E mutated-cell lines, together with their role in the control of expression of genes implicated in invasiveness (MMP1, PLAU, LCN2 and LGALS3). We also identified NF-κB-inducing kinase (NIK) as a novel actor of the constitutive activation of the NF-κB pathways in TC-derived cell lines. Finally, its implication in invasiveness and its overexpression in PTC samples make NIK a potential therapeutic target for advanced TC treatment.

在滤泡源性甲状腺癌(TC)中,具有侵袭性行为和对当前治疗有抵抗力的患者预后较差。NF-κB信号通路参与多种癌症的肿瘤进展。在这里,我们很好地描述了NF-κB通路及其在TC中的作用。通过免疫印迹和凝胶转移实验,我们证明了经典和替代NF-κB途径在10个tc来源的细胞系中被激活,导致激活RelA/p50和RelB/p50 NF-κB二聚体。通过分析来自癌症基因组图谱(TCGA)项目的大乳头状甲状腺癌(PTC)队列的RNAseq数据,我们在BRAFV600E突变的PTC中发现了肿瘤进展相关的NF-κB特征。这证实了RelA和RelB在BRAFV600E突变细胞系中在细胞迁移和侵袭过程中的作用,以及它们在控制侵袭性相关基因(MMP1、PLAU、LCN2和LGALS3)表达中的作用。我们还发现NF-κB诱导激酶(NIK)是tc来源细胞系中NF-κB通路组成性激活的新参与者。最后,它的侵袭性及其在PTC样品中的过表达使NIK成为晚期TC治疗的潜在治疗靶点。
{"title":"NF-κB signaling activation and roles in thyroid cancers: implication of MAP3K14/NIK.","authors":"Françoise Cormier, Selma Housni, Florent Dumont, Mélodie Villard, Béatrix Cochand-Priollet, Françoise Mercier-Nomé, Karine Perlemoine, Jérôme Bertherat, Lionel Groussin","doi":"10.1038/s41389-023-00496-w","DOIUrl":"10.1038/s41389-023-00496-w","url":null,"abstract":"<p><p>Among follicular-derived thyroid cancers (TC), those with aggressive behavior and resistance to current treatments display poor prognosis. NF-κB signaling pathways are involved in tumor progression of various cancers. Here, we finely characterize the NF-κB pathways and their involvement in TC. By using immunoblot and gel shift assays, we demonstrated that both classical and alternative NF-κB pathways are activated in ten TC-derived cell lines, leading to activated RelA/p50 and RelB/p50 NF-κB dimers. By analyzing the RNAseq data of the large papillary thyroid carcinoma (PTC) cohort from The Cancer Genome Atlas (TCGA) project, we identified a tumor progression-related NF-κB signature in BRAF<sup>V600E</sup> mutated-PTCs. That corroborated with the role of RelA and RelB in cell migration and invasion processes that we demonstrated specifically in BRAF<sup>V600E</sup> mutated-cell lines, together with their role in the control of expression of genes implicated in invasiveness (MMP1, PLAU, LCN2 and LGALS3). We also identified NF-κB-inducing kinase (NIK) as a novel actor of the constitutive activation of the NF-κB pathways in TC-derived cell lines. Finally, its implication in invasiveness and its overexpression in PTC samples make NIK a potential therapeutic target for advanced TC treatment.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":"12 1","pages":"55"},"PeriodicalIF":6.2,"publicationDate":"2023-11-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10654696/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"136398535","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Kinesin family member 18B activates mTORC1 signaling via actin gamma 1 to promote the recurrence of human hepatocellular carcinoma. Kinesin家族成员18B通过肌动蛋白γ 1激活mTORC1信号通路,促进人肝细胞癌的复发。
IF 6.2 2区 医学 Q1 ONCOLOGY Pub Date : 2023-11-13 DOI: 10.1038/s41389-023-00499-7
Qian Li, Mengqing Sun, Yao Meng, Mengqing Feng, Menglan Wang, Cunjie Chang, Heng Dong, Fangtian Bu, Chao Xu, Jing Liu, Qi Ling, Yiting Qiao, Jianxiang Chen

The mechanistic target of rapamycin complex 1 (mTORC1) signaling pathway is frequently reported to be hyperactivated in hepatocellular carcinoma (HCC) and contributes to HCC recurrence. However, the underlying regulatory mechanisms of mTORC1 signaling in HCC are not fully understood. In the present study, we found that the expression of kinesin family member 18B (KIF18B) was positively correlated with mTORC1 signaling in HCC, and the upregulation of KIF18B and p-mTOR was associated with a poor prognosis and HCC recurrence. Utilizing in vitro and in vivo assays, we showed that KIF18B promoted HCC cell proliferation and migration through activating mTORC1 signaling. Mechanistically, we identified Actin gamma 1 (γ-Actin) as a binding partner of KIF18B. KIF18B and γ-Actin synergistically modulated lysosome positioning, promoted mTORC1 translocation to lysosome membrane, and prohibited p70 S6K from entering lysosomes for degradation, which finally led to the enhancement of mTORC1 signaling transduction. Moreover, we found that KIF18B was a direct target of Forkhead box M1, which explains the potential mechanism of KIF18B overexpression in HCC. Our study highlights the potential of KIF18B as a therapeutic target for the treatment of HCC.

雷帕霉素复合物1 (mTORC1)信号通路的机制靶点经常被报道在肝细胞癌(HCC)中过度激活,并导致HCC复发。然而,mTORC1信号在HCC中的潜在调控机制尚不完全清楚。在本研究中,我们发现HCC中激酶家族成员18B (KIF18B)的表达与mTORC1信号通路呈正相关,KIF18B和p-mTOR的上调与预后不良和HCC复发有关。通过体外和体内实验,我们发现KIF18B通过激活mTORC1信号通路促进HCC细胞增殖和迁移。在机制上,我们确定了肌动蛋白γ- 1 (γ-Actin)是KIF18B的结合伙伴。KIF18B和γ-Actin协同调节溶酶体定位,促进mTORC1易位至溶酶体膜,阻止p70 S6K进入溶酶体降解,最终导致mTORC1信号转导增强。此外,我们发现KIF18B是叉头盒M1的直接靶点,这解释了KIF18B在HCC中过表达的潜在机制。我们的研究强调了KIF18B作为HCC治疗靶点的潜力。
{"title":"Kinesin family member 18B activates mTORC1 signaling via actin gamma 1 to promote the recurrence of human hepatocellular carcinoma.","authors":"Qian Li, Mengqing Sun, Yao Meng, Mengqing Feng, Menglan Wang, Cunjie Chang, Heng Dong, Fangtian Bu, Chao Xu, Jing Liu, Qi Ling, Yiting Qiao, Jianxiang Chen","doi":"10.1038/s41389-023-00499-7","DOIUrl":"10.1038/s41389-023-00499-7","url":null,"abstract":"<p><p>The mechanistic target of rapamycin complex 1 (mTORC1) signaling pathway is frequently reported to be hyperactivated in hepatocellular carcinoma (HCC) and contributes to HCC recurrence. However, the underlying regulatory mechanisms of mTORC1 signaling in HCC are not fully understood. In the present study, we found that the expression of kinesin family member 18B (KIF18B) was positively correlated with mTORC1 signaling in HCC, and the upregulation of KIF18B and p-mTOR was associated with a poor prognosis and HCC recurrence. Utilizing in vitro and in vivo assays, we showed that KIF18B promoted HCC cell proliferation and migration through activating mTORC1 signaling. Mechanistically, we identified Actin gamma 1 (γ-Actin) as a binding partner of KIF18B. KIF18B and γ-Actin synergistically modulated lysosome positioning, promoted mTORC1 translocation to lysosome membrane, and prohibited p70 S6K from entering lysosomes for degradation, which finally led to the enhancement of mTORC1 signaling transduction. Moreover, we found that KIF18B was a direct target of Forkhead box M1, which explains the potential mechanism of KIF18B overexpression in HCC. Our study highlights the potential of KIF18B as a therapeutic target for the treatment of HCC.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":"12 1","pages":"54"},"PeriodicalIF":6.2,"publicationDate":"2023-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10643429/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"92155922","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Coordinate transcriptional regulation of ErbB2/3 by C-terminal binding protein 2 signals sensitivity to ErbB2 inhibition in pancreatic adenocarcinoma. 胰腺癌中C端结合蛋白2对ErbB2/3的协同转录调控信号对ErbB2抑制的敏感性。
IF 6.2 2区 医学 Q1 ONCOLOGY Pub Date : 2023-11-10 DOI: 10.1038/s41389-023-00498-8
Kranthi Kumar Chougoni, Haemin Park, Priyadarshan K Damle, Travis Mason, Bo Cheng, Martin M Dcona, Barbara Szomju, Mikhail G Dozmorov, Michael O Idowu, Steven R Grossman

There is a critical need to identify new therapeutic vulnerabilities in pancreatic ductal adenocarcinoma (PDAC). Transcriptional co-regulators C-terminal binding proteins (CtBP) 1 and 2 are highly overexpressed in human PDAC, and CRISPR-based homozygous deletion of Ctbp2 in a mouse PDAC cell line (CKP) dramatically decreased tumor growth, reduced metastasis, and prolonged survival in orthotopic mouse allografts. Transcriptomic profiling of tumors derived from CKP vs. Ctbp2-deleted CKP cells (CKP/KO) revealed significant downregulation of the EGFR-superfamily receptor Erbb3, the heterodimeric signaling partner for both EGFR and ErbB2. Compared with CKP cells, CKP/KO cells also demonstrated reduced Erbb2 expression and did not activate downstream Akt signaling after stimulation of Erbb3 by its ligand neuregulin-1. ErbB3 expression in human PDAC cell lines was similarly dependent on CtBP2 and depletion of ErbB3 in a human PDAC cell line severely attenuated growth, demonstrating the critical role of ErbB3 signaling in maintaining PDAC cell growth. Sensitivity to the ErbB2-targeted tyrosine kinase inhibitor lapatinib, but not the EGFR-targeted agent erlotinib, varied in proportion to the level of ErbB3 expression in mouse and human PDAC cells, suggesting that an ErBb2 inhibitor can effectively leverage CtBP2-driven transcriptional activation of physiologic ErbB2/3 expression and signaling in PDAC cells for therapeutic benefit.

迫切需要确定胰腺导管腺癌(PDAC)新的治疗弱点。转录共调节因子C末端结合蛋白(CtBP)1和2在人PDAC中高度过表达,并且在小鼠PDAC细胞系(CKP)中基于CRISPR的Ctbp2纯合缺失显著降低了原位小鼠同种异体移植物中的肿瘤生长、减少了转移和延长了生存期。来源于CKP与Ctbp2缺失CKP细胞的肿瘤的转录组学分析(CKP/KO)显示EGFR超家族受体Erbb3显著下调,Erbb3是EGFR和ErbB2的异二聚体信号伴侣。与CKP细胞相比,CKP/KO细胞在其配体neuregulin-1刺激Erbb3后,也表现出Erbb2表达减少,并且不激活下游Akt信号传导。ErbB3在人PDAC细胞系中的表达类似地依赖于CtBP2,并且在人PDDC细胞系中ErbB3的缺失严重减弱了生长,证明了ErbB3信号在维持PDAC细胞生长中的关键作用。对ErbB2靶向酪氨酸激酶抑制剂拉帕替尼(而不是EGFR靶向剂埃洛替尼)的敏感性与小鼠和人PDAC细胞中ErbB3的表达水平成比例变化,这表明ErbB2抑制剂可以有效地利用PDAC细胞内生理性ErbB2/3表达和信号传导的CtBP2驱动的转录激活来获得治疗益处。
{"title":"Coordinate transcriptional regulation of ErbB2/3 by C-terminal binding protein 2 signals sensitivity to ErbB2 inhibition in pancreatic adenocarcinoma.","authors":"Kranthi Kumar Chougoni, Haemin Park, Priyadarshan K Damle, Travis Mason, Bo Cheng, Martin M Dcona, Barbara Szomju, Mikhail G Dozmorov, Michael O Idowu, Steven R Grossman","doi":"10.1038/s41389-023-00498-8","DOIUrl":"10.1038/s41389-023-00498-8","url":null,"abstract":"<p><p>There is a critical need to identify new therapeutic vulnerabilities in pancreatic ductal adenocarcinoma (PDAC). Transcriptional co-regulators C-terminal binding proteins (CtBP) 1 and 2 are highly overexpressed in human PDAC, and CRISPR-based homozygous deletion of Ctbp2 in a mouse PDAC cell line (CKP) dramatically decreased tumor growth, reduced metastasis, and prolonged survival in orthotopic mouse allografts. Transcriptomic profiling of tumors derived from CKP vs. Ctbp2-deleted CKP cells (CKP/KO) revealed significant downregulation of the EGFR-superfamily receptor Erbb3, the heterodimeric signaling partner for both EGFR and ErbB2. Compared with CKP cells, CKP/KO cells also demonstrated reduced Erbb2 expression and did not activate downstream Akt signaling after stimulation of Erbb3 by its ligand neuregulin-1. ErbB3 expression in human PDAC cell lines was similarly dependent on CtBP2 and depletion of ErbB3 in a human PDAC cell line severely attenuated growth, demonstrating the critical role of ErbB3 signaling in maintaining PDAC cell growth. Sensitivity to the ErbB2-targeted tyrosine kinase inhibitor lapatinib, but not the EGFR-targeted agent erlotinib, varied in proportion to the level of ErbB3 expression in mouse and human PDAC cells, suggesting that an ErBb2 inhibitor can effectively leverage CtBP2-driven transcriptional activation of physiologic ErbB2/3 expression and signaling in PDAC cells for therapeutic benefit.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":"12 1","pages":"53"},"PeriodicalIF":6.2,"publicationDate":"2023-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10638350/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"72210277","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Correction: PBK phosphorylates MSL1 to elicit epigenetic modulation of CD276 in nasopharyngeal carcinoma. 更正:PBK磷酸化MSL1以引发鼻咽癌中CD276的表观遗传学调节。
IF 6.2 2区 医学 Q1 ONCOLOGY Pub Date : 2023-11-08 DOI: 10.1038/s41389-022-00399-2
Meng-Yao Wang, Bin Qi, Fang Wang, Zhi-Rui Lin, Ming-Yi Li, Wen-Jing Yin, Yan-Yi Zhu, Lu He, Yi Yu, Fang Yang, Jin-Quan Liu, Dong-Ping Chen
{"title":"Correction: PBK phosphorylates MSL1 to elicit epigenetic modulation of CD276 in nasopharyngeal carcinoma.","authors":"Meng-Yao Wang, Bin Qi, Fang Wang, Zhi-Rui Lin, Ming-Yi Li, Wen-Jing Yin, Yan-Yi Zhu, Lu He, Yi Yu, Fang Yang, Jin-Quan Liu, Dong-Ping Chen","doi":"10.1038/s41389-022-00399-2","DOIUrl":"10.1038/s41389-022-00399-2","url":null,"abstract":"","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":"12 1","pages":"52"},"PeriodicalIF":6.2,"publicationDate":"2023-11-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10632349/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"71484538","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Combined in vitro/in vivo genome-wide CRISPR screens in triple negative breast cancer identify cancer stemness regulators in paclitaxel resistance. 三阴性乳腺癌症的体外/体内全基因组CRISPR联合筛选确定了紫杉醇耐药性中的癌症干性调节因子。
IF 6.2 2区 医学 Q1 ONCOLOGY Pub Date : 2023-11-06 DOI: 10.1038/s41389-023-00497-9
Gang Yan, Meiou Dai, Sophie Poulet, Ni Wang, Julien Boudreault, Girija Daliah, Suhad Ali, Jean-Jacques Lebrun

Triple negative breast cancer (TNBC) is defined as lacking the expressions of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). TNBC patients exhibit relatively poor clinical outcomes due to lack of molecular markers for targeted therapies. As such chemotherapy often remains the only systemic treatment option for these patients. While chemotherapy can initially help shrink TNBC tumor size, patients eventually develop resistance to drug, leading to tumor recurrence. We report a combined in vitro/in vivo genome-wide CRISPR synthetic lethality screening approach in a relevant TNBC cell line model to identify several targets responsible for the chemotherapy drug, paclitaxel resistance. Computational analysis integrating in vitro and in vivo data identified a set of genes, for which specific loss-of-function deletion enhanced paclitaxel resistance in TNBC. We found that several of these genes (ATP8B3, FOXR2, FRG2, HIST1H4A) act as cancer stemness negative regulators. Finally, using in vivo orthotopic transplantation TNBC models we showed that FRG2 gene deletion reduced paclitaxel efficacy and promoted tumor metastasis, while increasing FRG2 expression by means of CRISPR activation efficiently sensitized TNBC tumors to paclitaxel treatment and inhibited their metastatic abilities. In summary, the combined in vitro/in vivo genome-wide CRISPR screening approach proved effective as a tool to identify novel regulators of paclitaxel resistance/sensitivity and highlight the FRG2 gene as a potential therapeutical target overcoming paclitaxel resistance in TNBC.

癌症三阴性(TNBC)被定义为缺乏雌激素受体(ER)、孕酮受体(PR)和人表皮生长因子受体2(HER2)的表达。由于缺乏靶向治疗的分子标记物,TNBC患者表现出相对较差的临床结果。因此,化疗通常仍然是这些患者唯一的全身治疗选择。虽然化疗最初可以帮助缩小TNBC肿瘤的大小,但患者最终会产生耐药性,导致肿瘤复发。我们报道了一种在相关TNBC细胞系模型中进行体外/体内全基因组CRISPR合成致死性联合筛选的方法,以确定导致化疗药物紫杉醇耐药性的几个靶点。整合体外和体内数据的计算分析确定了一组基因,特异性功能缺失增强了TNBC中紫杉醇的耐药性。我们发现其中几个基因(ATP8B3、FOXR2、FRG2、HIST14A)作为癌症干性负调控因子。最后,使用体内原位移植TNBC模型,我们发现FRG2基因缺失降低了紫杉醇的疗效并促进了肿瘤转移,同时通过CRISPR激活增加了FRG2的表达,有效地使TNBC肿瘤对紫杉醇治疗敏感并抑制了其转移能力。总之,体外/体内全基因组CRISPR联合筛选方法被证明是一种有效的工具,可以识别紫杉醇耐药性/敏感性的新调节因子,并强调FRG2基因是克服TNBC紫杉醇耐药性的潜在治疗靶点。
{"title":"Combined in vitro/in vivo genome-wide CRISPR screens in triple negative breast cancer identify cancer stemness regulators in paclitaxel resistance.","authors":"Gang Yan, Meiou Dai, Sophie Poulet, Ni Wang, Julien Boudreault, Girija Daliah, Suhad Ali, Jean-Jacques Lebrun","doi":"10.1038/s41389-023-00497-9","DOIUrl":"10.1038/s41389-023-00497-9","url":null,"abstract":"<p><p>Triple negative breast cancer (TNBC) is defined as lacking the expressions of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). TNBC patients exhibit relatively poor clinical outcomes due to lack of molecular markers for targeted therapies. As such chemotherapy often remains the only systemic treatment option for these patients. While chemotherapy can initially help shrink TNBC tumor size, patients eventually develop resistance to drug, leading to tumor recurrence. We report a combined in vitro/in vivo genome-wide CRISPR synthetic lethality screening approach in a relevant TNBC cell line model to identify several targets responsible for the chemotherapy drug, paclitaxel resistance. Computational analysis integrating in vitro and in vivo data identified a set of genes, for which specific loss-of-function deletion enhanced paclitaxel resistance in TNBC. We found that several of these genes (ATP8B3, FOXR2, FRG2, HIST1H4A) act as cancer stemness negative regulators. Finally, using in vivo orthotopic transplantation TNBC models we showed that FRG2 gene deletion reduced paclitaxel efficacy and promoted tumor metastasis, while increasing FRG2 expression by means of CRISPR activation efficiently sensitized TNBC tumors to paclitaxel treatment and inhibited their metastatic abilities. In summary, the combined in vitro/in vivo genome-wide CRISPR screening approach proved effective as a tool to identify novel regulators of paclitaxel resistance/sensitivity and highlight the FRG2 gene as a potential therapeutical target overcoming paclitaxel resistance in TNBC.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":"12 1","pages":"51"},"PeriodicalIF":6.2,"publicationDate":"2023-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10628277/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"71484491","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Fibroblasts in metastatic lymph nodes confer cisplatin resistance to ESCC tumor cells via PI16. 转移性淋巴结中的成纤维细胞通过PI16赋予ESCC肿瘤细胞顺铂耐药性。
IF 6.2 2区 医学 Q1 ONCOLOGY Pub Date : 2023-11-01 DOI: 10.1038/s41389-023-00495-x
Lily Liang, Xu Zhang, Xiaodong Su, Tingting Zeng, Daqin Suo, Jingping Yun, Xin Wang, Xin-Yuan Guan, Yan Li

Although many studies have compared tumor fibroblasts (T-Fbs) and nontumor fibroblasts (N-Fbs), less is understood about the stromal contribution of metastatic lymph node fibroblasts (LN-Fbs) to the evolving microenvironment. Here, we explored the characteristics of LN-Fbs in esophageal squamous cell carcinoma (ESCC) and the interactions between fibroblasts and ESCC tumor cells in metastatic lymph nodes. Fibroblasts were isolated from tumor, nontumor and metastatic lymph node tissues from different patients with ESCC. Transcriptome sequencing was performed on the fibroblasts. Tumor growth and drug-resistance assays were carried out, and characteristics of T-Fbs, N-Fbs and LN-Fbs were determined. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to assay the culture medium of fibroblasts. The results demonstrated that fibroblasts derived from different tissues had different characteristics. Coculture with LN-Fbs conditioned medium inhibited ESCC tumor cell growth and induced chemoresistance in ESCC cells. LN-Fbs induced chemoresistance to cisplatin in ESCC cells by secreting PI16. Coculture with LN-Fbs conditioned medium decreased cisplatin-induced apoptosis in ESCC cells by regulating the p38 and JNK cell signaling pathways. Survival analyses showed that patients with high PI16 expression in Fbs of lymph nodes exhibited worse overall survival. We also examined PI16 expression in interstitial tissues in ESCC tumor samples of patients receiving platinum-based therapy postsurgery and found that high PI16 expression in tumor interstitial tissues was an independent prognostic factor for ESCC patients. In addition, an in vivo assay demonstrated that PI16 knockdown increased the sensitivity of ESCC cells to cisplatin. Our results suggest that fibroblasts in metastatic lymph nodes decrease apoptosis of ESCC cells via PI16, thereby providing a cisplatin-resistance niche and supporting ESCC tumor cells to survive in metastatic lymph nodes. PI16 is also a potential target for effectively blocking the chemoresistance niche signaling circuit in response to cisplatin.

尽管许多研究比较了肿瘤成纤维细胞(T-Fbs)和非肿瘤成纤维纤维细胞(N-Fbs),但对转移性淋巴结成纤维细胞对进化微环境的基质贡献知之甚少。在此,我们探讨了食管鳞状细胞癌(ESCC)中LN Fbs的特征,以及转移淋巴结中成纤维细胞和ESCC肿瘤细胞之间的相互作用。从不同ESCC患者的肿瘤、非肿瘤和转移性淋巴结组织中分离出成纤维细胞。对成纤维细胞进行转录组测序。进行肿瘤生长和耐药性测定,并测定T-Fbs、N-Fbs和LN Fbs的特性。采用液相色谱-串联质谱法(LC-MS/MS)对成纤维细胞培养基进行检测。结果表明,来源于不同组织的成纤维细胞具有不同的特性。与LN-Fbs条件培养基共培养抑制ESCC肿瘤细胞生长并诱导ESCC细胞的化学抗性。LN Fbs通过分泌PI16诱导ESCC细胞对顺铂产生化学耐药性。与LN-Fbs条件培养基共培养通过调节p38和JNK细胞信号通路降低顺铂诱导的ESCC细胞凋亡。生存分析显示,淋巴结Fbs中PI16高表达的患者总体生存率较差。我们还检测了接受铂类药物治疗的ESCC患者术后肿瘤样本中间质组织中PI16的表达,发现肿瘤间质组织的高PI16表达是ESCC患者的独立预后因素。此外,体内测定表明,敲低PI16增加了ESCC细胞对顺铂的敏感性。我们的结果表明,转移性淋巴结中的成纤维细胞通过PI16减少ESCC细胞的凋亡,从而提供顺铂耐药性小生境,并支持ESCC肿瘤细胞在转移性淋巴节中生存。PI16也是有效阻断化疗耐药性小生境信号通路以响应顺铂的潜在靶点。
{"title":"Fibroblasts in metastatic lymph nodes confer cisplatin resistance to ESCC tumor cells via PI16.","authors":"Lily Liang,&nbsp;Xu Zhang,&nbsp;Xiaodong Su,&nbsp;Tingting Zeng,&nbsp;Daqin Suo,&nbsp;Jingping Yun,&nbsp;Xin Wang,&nbsp;Xin-Yuan Guan,&nbsp;Yan Li","doi":"10.1038/s41389-023-00495-x","DOIUrl":"10.1038/s41389-023-00495-x","url":null,"abstract":"<p><p>Although many studies have compared tumor fibroblasts (T-Fbs) and nontumor fibroblasts (N-Fbs), less is understood about the stromal contribution of metastatic lymph node fibroblasts (LN-Fbs) to the evolving microenvironment. Here, we explored the characteristics of LN-Fbs in esophageal squamous cell carcinoma (ESCC) and the interactions between fibroblasts and ESCC tumor cells in metastatic lymph nodes. Fibroblasts were isolated from tumor, nontumor and metastatic lymph node tissues from different patients with ESCC. Transcriptome sequencing was performed on the fibroblasts. Tumor growth and drug-resistance assays were carried out, and characteristics of T-Fbs, N-Fbs and LN-Fbs were determined. Liquid chromatography-tandem mass spectrometry (LC-MS/MS) was used to assay the culture medium of fibroblasts. The results demonstrated that fibroblasts derived from different tissues had different characteristics. Coculture with LN-Fbs conditioned medium inhibited ESCC tumor cell growth and induced chemoresistance in ESCC cells. LN-Fbs induced chemoresistance to cisplatin in ESCC cells by secreting PI16. Coculture with LN-Fbs conditioned medium decreased cisplatin-induced apoptosis in ESCC cells by regulating the p38 and JNK cell signaling pathways. Survival analyses showed that patients with high PI16 expression in Fbs of lymph nodes exhibited worse overall survival. We also examined PI16 expression in interstitial tissues in ESCC tumor samples of patients receiving platinum-based therapy postsurgery and found that high PI16 expression in tumor interstitial tissues was an independent prognostic factor for ESCC patients. In addition, an in vivo assay demonstrated that PI16 knockdown increased the sensitivity of ESCC cells to cisplatin. Our results suggest that fibroblasts in metastatic lymph nodes decrease apoptosis of ESCC cells via PI16, thereby providing a cisplatin-resistance niche and supporting ESCC tumor cells to survive in metastatic lymph nodes. PI16 is also a potential target for effectively blocking the chemoresistance niche signaling circuit in response to cisplatin.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":"12 1","pages":"50"},"PeriodicalIF":6.2,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10620422/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"71425621","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Oncogenesis
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