首页 > 最新文献

Molecular Cancer最新文献

英文 中文
Serum small RNAs in metastatic colorectal cancer predict response to chemotherapy and characterize high-risk patients 转移性结直肠癌血清小 RNA 预测化疗反应并描述高危患者特征
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-06-27 DOI: 10.1186/s12943-024-02042-7
Robin Mjelle, Are K. Kristensen, Tora S. Solheim, Ganna S. Westvik, Hege Elvebakken, Eva Hofsli
Metastatic colorectal cancer (mCRC) presents significant challenges in clinical management due to its heterogeneity and variable response to treatment. In this study, we conducted comprehensive small RNA (sRNA) sequencing analyses to identify sRNA biomarkers associated with survival and treatment response in mCRC patients. We measured serum sRNAs before and after chemotherapy treatment in a discovery cohort of 189 mCRC patients. Our analysis revealed 25 microRNAs (miRNA) as significantly associated with overall survival at baseline. We found that 11 of the 25 significant miRNAs were also significant in an independent validation cohort of 20 mCRC patients, including the top five miRNAs from the discovery cohort. Importantly, all but four of the 25 significant miRNAs from the discovery cohort had hazard ratios in the same direction in the validation cohort. Among the 25 significant miRNAs, we identified the miR-320 family of miRNAs as the strongest independent prognostic marker, with high baseline levels correlating with poor survival outcomes. Furthermore, post-treatment levels of the same miRNAs were even more predictive of overall survival, emphasizing the prognostic value of serum changes in miRNA levels before and after treatment. Moreover, we observed significant changes in serum miRNAs and other sRNAs when comparing samples before and after chemotherapy, with distinct expression patterns between responders and non-responders. Leveraging these differential expression patterns, we established a serum sRNA signature that accurately predicts response to chemotherapy with an area under the curve (AUC) of 0.8. In summary, our study highlights the prognostic and predictive potential of sRNA biomarkers in mCRC, offering valuable insights into patient stratification and personalized treatment approaches.
转移性结直肠癌(mCRC)由于其异质性和对治疗的不同反应,给临床治疗带来了巨大挑战。在这项研究中,我们进行了全面的小 RNA(sRNA)测序分析,以确定与 mCRC 患者生存和治疗反应相关的 sRNA 生物标志物。我们测量了 189 名 mCRC 患者化疗前后的血清 sRNA。我们的分析发现,25 种微小 RNA(miRNA)与基线总生存期显著相关。我们发现,在由 20 名 mCRC 患者组成的独立验证队列中,25 个重要 miRNA 中的 11 个也具有重要意义,其中包括发现队列中的前 5 个 miRNA。重要的是,在发现队列的 25 个重要 miRNA 中,除了 4 个之外,其他所有 miRNA 在验证队列中的危险比都是相同的。在这25个重要的miRNA中,我们发现miR-320家族miRNA是最强的独立预后标志物,高基线水平与不良生存结果相关。此外,治疗后相同 miRNA 的水平对总生存期的预测作用更大,这强调了治疗前后血清中 miRNA 水平变化的预后价值。此外,在比较化疗前后的样本时,我们观察到血清中的 miRNA 和其他 sRNA 发生了显著变化,应答者和非应答者的表达模式截然不同。利用这些差异表达模式,我们建立了一个血清 sRNA 特征,它能准确预测化疗反应,曲线下面积 (AUC) 为 0.8。总之,我们的研究强调了 sRNA 生物标志物在 mCRC 中的预后和预测潜力,为患者分层和个性化治疗方法提供了宝贵的见解。
{"title":"Serum small RNAs in metastatic colorectal cancer predict response to chemotherapy and characterize high-risk patients","authors":"Robin Mjelle, Are K. Kristensen, Tora S. Solheim, Ganna S. Westvik, Hege Elvebakken, Eva Hofsli","doi":"10.1186/s12943-024-02042-7","DOIUrl":"https://doi.org/10.1186/s12943-024-02042-7","url":null,"abstract":"Metastatic colorectal cancer (mCRC) presents significant challenges in clinical management due to its heterogeneity and variable response to treatment. In this study, we conducted comprehensive small RNA (sRNA) sequencing analyses to identify sRNA biomarkers associated with survival and treatment response in mCRC patients. We measured serum sRNAs before and after chemotherapy treatment in a discovery cohort of 189 mCRC patients. Our analysis revealed 25 microRNAs (miRNA) as significantly associated with overall survival at baseline. We found that 11 of the 25 significant miRNAs were also significant in an independent validation cohort of 20 mCRC patients, including the top five miRNAs from the discovery cohort. Importantly, all but four of the 25 significant miRNAs from the discovery cohort had hazard ratios in the same direction in the validation cohort. Among the 25 significant miRNAs, we identified the miR-320 family of miRNAs as the strongest independent prognostic marker, with high baseline levels correlating with poor survival outcomes. Furthermore, post-treatment levels of the same miRNAs were even more predictive of overall survival, emphasizing the prognostic value of serum changes in miRNA levels before and after treatment. Moreover, we observed significant changes in serum miRNAs and other sRNAs when comparing samples before and after chemotherapy, with distinct expression patterns between responders and non-responders. Leveraging these differential expression patterns, we established a serum sRNA signature that accurately predicts response to chemotherapy with an area under the curve (AUC) of 0.8. In summary, our study highlights the prognostic and predictive potential of sRNA biomarkers in mCRC, offering valuable insights into patient stratification and personalized treatment approaches.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-06-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141461990","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Deciphering the tumor immune microenvironment from a multidimensional omics perspective: insight into next-generation CAR-T cell immunotherapy and beyond. 从多维 omics 角度解读肿瘤免疫微环境:洞察下一代 CAR-T 细胞免疫疗法及其他疗法。
IF 27.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-06-26 DOI: 10.1186/s12943-024-02047-2
Zhaokai Zhou, Jiahui Wang, Jiaojiao Wang, Shuai Yang, Ruizhi Wang, Ge Zhang, Zhengrui Li, Run Shi, Zhan Wang, Qiong Lu

Tumor immune microenvironment (TIME) consists of intra-tumor immunological components and plays a significant role in tumor initiation, progression, metastasis, and response to therapy. Chimeric antigen receptor (CAR)-T cell immunotherapy has revolutionized the cancer treatment paradigm. Although CAR-T cell immunotherapy has emerged as a successful treatment for hematologic malignancies, it remains a conundrum for solid tumors. The heterogeneity of TIME is responsible for poor outcomes in CAR-T cell immunotherapy against solid tumors. The advancement of highly sophisticated technology enhances our exploration in TIME from a multi-omics perspective. In the era of machine learning, multi-omics studies could reveal the characteristics of TIME and its immune resistance mechanism. Therefore, the clinical efficacy of CAR-T cell immunotherapy in solid tumors could be further improved with strategies that target unfavorable conditions in TIME. Herein, this review seeks to investigate the factors influencing TIME formation and propose strategies for improving the effectiveness of CAR-T cell immunotherapy through a multi-omics perspective, with the ultimate goal of developing personalized therapeutic approaches.

肿瘤免疫微环境(TIME)由肿瘤内免疫成分组成,在肿瘤的发生、发展、转移和治疗反应中发挥着重要作用。嵌合抗原受体(CAR)-T 细胞免疫疗法彻底改变了癌症治疗模式。虽然 CAR-T 细胞免疫疗法已成功治疗了血液系统恶性肿瘤,但对于实体瘤来说,它仍然是一个难题。TIME的异质性是CAR-T细胞免疫疗法治疗实体瘤疗效不佳的原因。高精尖技术的进步加强了我们从多组学角度对 TIME 的探索。在机器学习时代,多组学研究可以揭示 TIME 的特征及其免疫耐受机制。因此,针对TIME中的不利条件采取相应策略,可进一步提高CAR-T细胞免疫疗法在实体瘤中的临床疗效。在此,本综述旨在通过多组学视角研究影响TIME形成的因素,并提出提高CAR-T细胞免疫疗法疗效的策略,最终实现开发个性化治疗方法的目标。
{"title":"Deciphering the tumor immune microenvironment from a multidimensional omics perspective: insight into next-generation CAR-T cell immunotherapy and beyond.","authors":"Zhaokai Zhou, Jiahui Wang, Jiaojiao Wang, Shuai Yang, Ruizhi Wang, Ge Zhang, Zhengrui Li, Run Shi, Zhan Wang, Qiong Lu","doi":"10.1186/s12943-024-02047-2","DOIUrl":"10.1186/s12943-024-02047-2","url":null,"abstract":"<p><p>Tumor immune microenvironment (TIME) consists of intra-tumor immunological components and plays a significant role in tumor initiation, progression, metastasis, and response to therapy. Chimeric antigen receptor (CAR)-T cell immunotherapy has revolutionized the cancer treatment paradigm. Although CAR-T cell immunotherapy has emerged as a successful treatment for hematologic malignancies, it remains a conundrum for solid tumors. The heterogeneity of TIME is responsible for poor outcomes in CAR-T cell immunotherapy against solid tumors. The advancement of highly sophisticated technology enhances our exploration in TIME from a multi-omics perspective. In the era of machine learning, multi-omics studies could reveal the characteristics of TIME and its immune resistance mechanism. Therefore, the clinical efficacy of CAR-T cell immunotherapy in solid tumors could be further improved with strategies that target unfavorable conditions in TIME. Herein, this review seeks to investigate the factors influencing TIME formation and propose strategies for improving the effectiveness of CAR-T cell immunotherapy through a multi-omics perspective, with the ultimate goal of developing personalized therapeutic approaches.</p>","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":27.7,"publicationDate":"2024-06-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11201788/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141450976","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
Patients with ASPSCR1-TFE3 fusion achieve better response to ICI based combination therapy among TFE3-rearranged renal cell carcinoma 在TFE3重组肾细胞癌中,ASPSCR1-TFE3融合患者对基于ICI的联合疗法反应更好
IF 37.3 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-06-26 DOI: 10.1186/s12943-024-02044-5
Junjie Zhao, Yanfeng Tang, Xu Hu, Xiaoxue Yin, Yuntian Chen, Junru Chen, Haoyang Liu, Haolin Liu, Jiayu Liang, Xingming Zhang, Jinge Zhao, Sha Zhu, Yuchao Ni, Zhipeng Wang, Jindong Dai, Zilin Wang, Yaowen Zhang, Jin Yao, Ni Chen, Pengfei Shen, Zhenhua H. Liu, Hao Zeng, Guangxi X. Sun
TFE3-rearranged renal cell carcinoma (TFE3-rRCC) is a rare but highly heterogeneous renal cell carcinoma (RCC) entity, of which the clinical treatment landscape is largely undefined. This study aims to evaluate and compare the efficacy of different systemic treatments and further explore the molecular correlates. Thirty-eight patients with metastatic TFE3-rRCC were enrolled. Main outcomes included progression-free survival (PFS), overall survival, objective response rate (ORR) and disease control rate. RNA sequencing was performed on 32 tumors. Patients receiving first-line immune checkpoint inhibitor (ICI) based combination therapy achieved longer PFS than those treated without ICI (median PFS: 11.5 vs. 5.1 months, P = 0.098). After stratification of fusion partners, the superior efficacy of first-line ICI based combination therapy was predominantly observed in ASPSCR1-TFE3 rRCC (median PFS: not reached vs. 6.5 months, P = 0.01; ORR: 67.5% vs. 10.0%, P = 0.019), but almost not in non-ASPSCR1-TFE3 rRCC. Transcriptomic data revealed enrichment of ECM and collagen-related signaling in ASPSCR1-TFE3 rRCC, which might interfere with the potential efficacy of anti-angiogenic monotherapy. Whereas angiogenesis and immune activities were exclusively enriched in ASPSCR1-TFE3 rRCC and promised the better clinical outcomes with ICI plus tyrosine kinase inhibitor combination therapy. The current study represents the largest cohort comparing treatment outcomes and investigating molecular correlates of metastatic TFE3-rRCC based on fusion partner stratification. ICI based combination therapy could serve as an effective first-line treatment option for metastatic ASPSCR1-TFE3 rRCC patients. Regarding with other fusion subtypes, further investigations should be performed to explore the molecular mechanisms to propose pointed therapeutic strategy accordingly.
TFE3重排肾细胞癌(TFE3-rRCC)是一种罕见但高度异质性的肾细胞癌(RCC)实体,其临床治疗前景在很大程度上尚未明确。本研究旨在评估和比较不同系统治疗方法的疗效,并进一步探讨其分子相关性。研究共纳入38例转移性TFE3-rRCC患者。主要结果包括无进展生存期(PFS)、总生存期、客观反应率(ORR)和疾病控制率。对32个肿瘤进行了RNA测序。接受基于免疫检查点抑制剂(ICI)的一线联合疗法的患者比未接受ICI治疗的患者获得了更长的PFS(中位PFS:11.5个月 vs. 5.1个月,P = 0.098)。在对融合伙伴进行分层后,基于 ICI 的一线联合疗法的卓越疗效主要在 ASPSCR1-TFE3 rRCC 中观察到(中位 PFS:未达到 vs. 6.5 个月,P = 0.01;ORR:67.5% vs. 10.0%,P = 0.019),但在非 ASPSCR1-TFE3 rRCC 中几乎没有观察到。转录组数据显示,ASPSCR1-TFE3 rRCC中与ECM和胶原蛋白相关的信号转导丰富,这可能会干扰抗血管生成单药的潜在疗效。而血管生成和免疫活动在ASPSCR1-TFE3型rRCC中完全富集,这就保证了ICI加酪氨酸激酶抑制剂联合疗法能取得更好的临床疗效。目前的研究是基于融合伙伴分层比较转移性TFE3-rRCC治疗效果并研究其分子相关性的最大队列。基于 ICI 的联合疗法可作为转移性 ASPSCR1-TFE3 rRCC 患者的有效一线治疗方案。对于其他融合亚型,应进一步研究其分子机制,从而提出相应的治疗策略。
{"title":"Patients with ASPSCR1-TFE3 fusion achieve better response to ICI based combination therapy among TFE3-rearranged renal cell carcinoma","authors":"Junjie Zhao, Yanfeng Tang, Xu Hu, Xiaoxue Yin, Yuntian Chen, Junru Chen, Haoyang Liu, Haolin Liu, Jiayu Liang, Xingming Zhang, Jinge Zhao, Sha Zhu, Yuchao Ni, Zhipeng Wang, Jindong Dai, Zilin Wang, Yaowen Zhang, Jin Yao, Ni Chen, Pengfei Shen, Zhenhua H. Liu, Hao Zeng, Guangxi X. Sun","doi":"10.1186/s12943-024-02044-5","DOIUrl":"https://doi.org/10.1186/s12943-024-02044-5","url":null,"abstract":"TFE3-rearranged renal cell carcinoma (TFE3-rRCC) is a rare but highly heterogeneous renal cell carcinoma (RCC) entity, of which the clinical treatment landscape is largely undefined. This study aims to evaluate and compare the efficacy of different systemic treatments and further explore the molecular correlates. Thirty-eight patients with metastatic TFE3-rRCC were enrolled. Main outcomes included progression-free survival (PFS), overall survival, objective response rate (ORR) and disease control rate. RNA sequencing was performed on 32 tumors. Patients receiving first-line immune checkpoint inhibitor (ICI) based combination therapy achieved longer PFS than those treated without ICI (median PFS: 11.5 vs. 5.1 months, P = 0.098). After stratification of fusion partners, the superior efficacy of first-line ICI based combination therapy was predominantly observed in ASPSCR1-TFE3 rRCC (median PFS: not reached vs. 6.5 months, P = 0.01; ORR: 67.5% vs. 10.0%, P = 0.019), but almost not in non-ASPSCR1-TFE3 rRCC. Transcriptomic data revealed enrichment of ECM and collagen-related signaling in ASPSCR1-TFE3 rRCC, which might interfere with the potential efficacy of anti-angiogenic monotherapy. Whereas angiogenesis and immune activities were exclusively enriched in ASPSCR1-TFE3 rRCC and promised the better clinical outcomes with ICI plus tyrosine kinase inhibitor combination therapy. The current study represents the largest cohort comparing treatment outcomes and investigating molecular correlates of metastatic TFE3-rRCC based on fusion partner stratification. ICI based combination therapy could serve as an effective first-line treatment option for metastatic ASPSCR1-TFE3 rRCC patients. Regarding with other fusion subtypes, further investigations should be performed to explore the molecular mechanisms to propose pointed therapeutic strategy accordingly.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-06-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141452979","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Advances in spatial transcriptomics and its applications in cancer research 空间转录组学的进展及其在癌症研究中的应用
IF 37.3 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-06-20 DOI: 10.1186/s12943-024-02040-9
Yang Jin, Yuanli Zuo, Gang Li, Wenrong Liu, Yitong Pan, Ting Fan, Xin Fu, Xiaojun Yao, Yong Peng
Malignant tumors have increasing morbidity and high mortality, and their occurrence and development is a complicate process. The development of sequencing technologies enabled us to gain a better understanding of the underlying genetic and molecular mechanisms in tumors. In recent years, the spatial transcriptomics sequencing technologies have been developed rapidly and allow the quantification and illustration of gene expression in the spatial context of tissues. Compared with the traditional transcriptomics technologies, spatial transcriptomics technologies not only detect gene expression levels in cells, but also inform the spatial location of genes within tissues, cell composition of biological tissues, and interaction between cells. Here we summarize the development of spatial transcriptomics technologies, spatial transcriptomics tools and its application in cancer research. We also discuss the limitations and challenges of current spatial transcriptomics approaches, as well as future development and prospects.
恶性肿瘤的发病率和死亡率越来越高,其发生和发展是一个复杂的过程。测序技术的发展使我们能够更好地了解肿瘤的潜在遗传和分子机制。近年来,空间转录组学测序技术发展迅速,可对组织空间背景下的基因表达进行量化和图解。与传统的转录组学技术相比,空间转录组学技术不仅能检测细胞内的基因表达水平,还能告知基因在组织内的空间位置、生物组织的细胞组成以及细胞之间的相互作用。在此,我们总结了空间转录组学技术的发展、空间转录组学工具及其在癌症研究中的应用。我们还讨论了当前空间转录组学方法的局限性和挑战,以及未来的发展和前景。
{"title":"Advances in spatial transcriptomics and its applications in cancer research","authors":"Yang Jin, Yuanli Zuo, Gang Li, Wenrong Liu, Yitong Pan, Ting Fan, Xin Fu, Xiaojun Yao, Yong Peng","doi":"10.1186/s12943-024-02040-9","DOIUrl":"https://doi.org/10.1186/s12943-024-02040-9","url":null,"abstract":"Malignant tumors have increasing morbidity and high mortality, and their occurrence and development is a complicate process. The development of sequencing technologies enabled us to gain a better understanding of the underlying genetic and molecular mechanisms in tumors. In recent years, the spatial transcriptomics sequencing technologies have been developed rapidly and allow the quantification and illustration of gene expression in the spatial context of tissues. Compared with the traditional transcriptomics technologies, spatial transcriptomics technologies not only detect gene expression levels in cells, but also inform the spatial location of genes within tissues, cell composition of biological tissues, and interaction between cells. Here we summarize the development of spatial transcriptomics technologies, spatial transcriptomics tools and its application in cancer research. We also discuss the limitations and challenges of current spatial transcriptomics approaches, as well as future development and prospects.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-06-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141430417","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
The role of RNA methylation in tumor immunity and its potential in immunotherapy RNA 甲基化在肿瘤免疫中的作用及其在免疫疗法中的潜力
IF 37.3 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-06-20 DOI: 10.1186/s12943-024-02041-8
Yan Li, Haoer Jin, Qingling Li, Liangrong Shi, Yitao Mao, Luqing Zhao
RNA methylation, a prevalent post-transcriptional modification, has garnered considerable attention in research circles. It exerts regulatory control over diverse biological functions by modulating RNA splicing, translation, transport, and stability. Notably, studies have illuminated the substantial impact of RNA methylation on tumor immunity. The primary types of RNA methylation encompass N6-methyladenosine (m6A), 5-methylcytosine (m5C), N1-methyladenosine (m1A), and N7-methylguanosine (m7G), and 3-methylcytidine (m3C). Compelling evidence underscores the involvement of RNA methylation in regulating the tumor microenvironment (TME). By affecting RNA translation and stability through the "writers", "erasers" and "readers", RNA methylation exerts influence over the dysregulation of immune cells and immune factors. Consequently, RNA methylation plays a pivotal role in modulating tumor immunity and mediating various biological behaviors, encompassing proliferation, invasion, metastasis, etc. In this review, we discussed the mechanisms and functions of several RNA methylations, providing a comprehensive overview of their biological roles and underlying mechanisms within the tumor microenvironment and among immunocytes. By exploring how these RNA modifications mediate tumor immune evasion, we also examine their potential applications in immunotherapy. This review aims to provide novel insights and strategies for identifying novel targets in RNA methylation and advancing cancer immunotherapy efficacy.
RNA 甲基化是一种普遍的转录后修饰,在研究领域引起了广泛关注。它通过调节 RNA 的剪接、翻译、转运和稳定性,对多种生物功能进行调控。值得注意的是,研究揭示了 RNA 甲基化对肿瘤免疫的重大影响。RNA 甲基化的主要类型包括 N6-甲基腺苷(m6A)、5-甲基胞嘧啶(m5C)、N1-甲基腺苷(m1A)、N7-甲基鸟苷(m7G)和 3-甲基胞嘧啶(m3C)。有确凿证据表明,RNA 甲基化参与了肿瘤微环境(TME)的调控。通过 "书写者"、"擦除者 "和 "阅读者 "对 RNA 翻译和稳定性的影响,RNA 甲基化对免疫细胞和免疫因子的失调产生了影响。因此,RNA 甲基化在调节肿瘤免疫和介导各种生物学行为(包括增殖、侵袭、转移等)方面发挥着关键作用。在这篇综述中,我们讨论了几种 RNA 甲基化的机制和功能,全面概述了它们在肿瘤微环境和免疫细胞中的生物学作用和内在机制。通过探讨这些 RNA 修饰如何介导肿瘤免疫逃避,我们还研究了它们在免疫疗法中的潜在应用。本综述旨在为确定 RNA 甲基化的新靶点和提高癌症免疫疗法疗效提供新的见解和策略。
{"title":"The role of RNA methylation in tumor immunity and its potential in immunotherapy","authors":"Yan Li, Haoer Jin, Qingling Li, Liangrong Shi, Yitao Mao, Luqing Zhao","doi":"10.1186/s12943-024-02041-8","DOIUrl":"https://doi.org/10.1186/s12943-024-02041-8","url":null,"abstract":"RNA methylation, a prevalent post-transcriptional modification, has garnered considerable attention in research circles. It exerts regulatory control over diverse biological functions by modulating RNA splicing, translation, transport, and stability. Notably, studies have illuminated the substantial impact of RNA methylation on tumor immunity. The primary types of RNA methylation encompass N6-methyladenosine (m6A), 5-methylcytosine (m5C), N1-methyladenosine (m1A), and N7-methylguanosine (m7G), and 3-methylcytidine (m3C). Compelling evidence underscores the involvement of RNA methylation in regulating the tumor microenvironment (TME). By affecting RNA translation and stability through the \"writers\", \"erasers\" and \"readers\", RNA methylation exerts influence over the dysregulation of immune cells and immune factors. Consequently, RNA methylation plays a pivotal role in modulating tumor immunity and mediating various biological behaviors, encompassing proliferation, invasion, metastasis, etc. In this review, we discussed the mechanisms and functions of several RNA methylations, providing a comprehensive overview of their biological roles and underlying mechanisms within the tumor microenvironment and among immunocytes. By exploring how these RNA modifications mediate tumor immune evasion, we also examine their potential applications in immunotherapy. This review aims to provide novel insights and strategies for identifying novel targets in RNA methylation and advancing cancer immunotherapy efficacy.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-06-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141430416","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Circ6834 suppresses non-small cell lung cancer progression by destabilizing ANHAK and regulating miR-873-5p/TXNIP axis Circ6834通过破坏ANHAK的稳定性和调节miR-873-5p/TXNIP轴抑制非小细胞肺癌的进展
IF 37.3 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-06-18 DOI: 10.1186/s12943-024-02038-3
Maoye Wang, Xiaoge Ding, Xinjian Fang, Jing Xu, Yanke Chen, Yu Qian, Jiahui Zhang, Dan Yu, Xiaoxin Zhang, Xiuqin Ma, Taofeng Zhu, Jianmei Gu, Xu Zhang
Circular RNAs (circRNAs) play important roles in cancer progression and metastasis. However, the expression profiles and biological roles of circRNAs in non-small cell lung cancer (NSCLC) remain unclear. In this study, we identified a novel circRNA, hsa_circ_0006834 (termed circ6834), in NSCLC by RNA-seq and investigated the biological role of circ6834 in NSCLC progression in vitro and in vivo. Finally, the molecular mechanism of circ6834 was revealed by tagged RNA affinity purification (TRAP), western blot, RNA immunoprecipitation, dual luciferase reporter gene assays and rescue experiments. Our results showed that circ6834 was downregulated in NSCLC tumor tissues and cell lines. Circ6834 overexpression inhibited NSCLC cell growth and metastasis both in vitro and in vivo, while circ6834 knockdown had the opposite effect. We found that TGF-β treatment decreased circ6834 expression, which was associated with the QKI reduction in NSCLC cells and circ6834 antagonized TGF-β-induced EMT and metastasis in NSCLC cells. Mechanistically, circ6834 bound to AHNAK protein, a key regulator of TGF-β/Smad signaling, and inhibited its stability by enhancing TRIM25-mediated ubiquitination and degradation. In addition, circ6834 acted as a miRNA sponge for miR-873-5p and upregulated TXNIP gene expression, which together inactivated the TGF-β/Smad signaling pathway in NSCLC cells. In conclusion, circ6834 is a tumor-suppressive circRNA that inhibits NSCLC progression by forming a negative regulatory feedback loop with the TGF-β/Smad signaling pathway and represents a novel therapeutic target for NSCLC.
环状 RNA(circRNA)在癌症进展和转移中发挥着重要作用。然而,circRNAs 在非小细胞肺癌(NSCLC)中的表达谱和生物学作用仍不清楚。在这项研究中,我们通过RNA-seq鉴定了NSCLC中的一种新型circRNA--hsa_circ_0006834(称为circ6834),并研究了circ6834在NSCLC体外和体内进展中的生物学作用。最后,通过标记 RNA 亲和纯化(TRAP)、Western 印迹、RNA 免疫沉淀、双荧光素酶报告基因实验和拯救实验揭示了 circ6834 的分子机制。结果表明,circ6834在NSCLC肿瘤组织和细胞系中被下调。circ6834过表达可抑制NSCLC细胞在体外和体内的生长和转移,而circ6834敲除则有相反的效果。我们发现,TGF-β处理会降低circ6834的表达,这与NSCLC细胞中QKI的降低有关,而且circ6834能拮抗TGF-β诱导的NSCLC细胞的EMT和转移。从机理上讲,circ6834与TGF-β/Smad信号转导的关键调控因子AHNAK蛋白结合,并通过增强TRIM25介导的泛素化和降解抑制其稳定性。此外,circ6834还是miR-873-5p的miRNA海绵,并上调TXNIP基因的表达,这些作用共同使NSCLC细胞中的TGF-β/Smad信号通路失活。总之,circ6834是一种抑制肿瘤的circRNA,它通过与TGF-β/Smad信号通路形成负向调节反馈环来抑制NSCLC的进展,是NSCLC的一个新的治疗靶点。
{"title":"Circ6834 suppresses non-small cell lung cancer progression by destabilizing ANHAK and regulating miR-873-5p/TXNIP axis","authors":"Maoye Wang, Xiaoge Ding, Xinjian Fang, Jing Xu, Yanke Chen, Yu Qian, Jiahui Zhang, Dan Yu, Xiaoxin Zhang, Xiuqin Ma, Taofeng Zhu, Jianmei Gu, Xu Zhang","doi":"10.1186/s12943-024-02038-3","DOIUrl":"https://doi.org/10.1186/s12943-024-02038-3","url":null,"abstract":"Circular RNAs (circRNAs) play important roles in cancer progression and metastasis. However, the expression profiles and biological roles of circRNAs in non-small cell lung cancer (NSCLC) remain unclear. In this study, we identified a novel circRNA, hsa_circ_0006834 (termed circ6834), in NSCLC by RNA-seq and investigated the biological role of circ6834 in NSCLC progression in vitro and in vivo. Finally, the molecular mechanism of circ6834 was revealed by tagged RNA affinity purification (TRAP), western blot, RNA immunoprecipitation, dual luciferase reporter gene assays and rescue experiments. Our results showed that circ6834 was downregulated in NSCLC tumor tissues and cell lines. Circ6834 overexpression inhibited NSCLC cell growth and metastasis both in vitro and in vivo, while circ6834 knockdown had the opposite effect. We found that TGF-β treatment decreased circ6834 expression, which was associated with the QKI reduction in NSCLC cells and circ6834 antagonized TGF-β-induced EMT and metastasis in NSCLC cells. Mechanistically, circ6834 bound to AHNAK protein, a key regulator of TGF-β/Smad signaling, and inhibited its stability by enhancing TRIM25-mediated ubiquitination and degradation. In addition, circ6834 acted as a miRNA sponge for miR-873-5p and upregulated TXNIP gene expression, which together inactivated the TGF-β/Smad signaling pathway in NSCLC cells. In conclusion, circ6834 is a tumor-suppressive circRNA that inhibits NSCLC progression by forming a negative regulatory feedback loop with the TGF-β/Smad signaling pathway and represents a novel therapeutic target for NSCLC.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-06-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141334131","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Multi-omics profiling reveal cells with novel oncogenic cluster, TRAP1low/CAMSAP3low, emerge more aggressive behavior and poor-prognosis in early-stage endometrial cancer. 多组学分析表明,在早期子宫内膜癌中,具有新型致癌基因簇 TRAP1low/CAMSAP3low 的细胞更具侵袭性,预后更差。
IF 37.3 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-06-17 DOI: 10.1186/s12943-024-02039-2
Xiaodan Mao, Xiaoyue Tang, Jingxuan Ye, Shuxia Xu, Yue Wang, Xianhua Liu, Qibin Wu, Xite Lin, Maotong Zhang, Jiangfeng Liu, Juntao Yang, Pengming Sun

The clinical heterogeneity of early-stage endometrial cancer (EC) is worthy of further study to identify high-quality prognostic markers and their potential role in aggressive tumor behavior. Mutation of TP53 was considered as an important primary triage in modified molecular typing for EC, it still cannot precisely predict the prognosis of EC. After proteomic analysis of cancer and para-cancerous tissues from 24 early-stage endometrioid EC patients with different survival outcomes, 13 differentially expressed proteins were screen out while 2 proteins enriched in p53 signaling pathway were further identified by single-cell transcriptome (scRNA-seq). Interestingly, tumor necrosis factor type-1 receptor-associated protein (TRAP1) and calmodulin-regulated spectrin-associated protein family member 3 (CAMSAP3) were found to be significantly downregulated in the specific cell cluster. Expectedly, the signature genes of TRAP1low/CAMSAP3low cluster included classical oncogenes. Moreover, close cellular interactions were observed between myeloid cells and the TRAP1low/CAMSAP3low cluster after systematically elucidating their relationship with tumor microenvironment (TME). The expression of TRAP1 and CAMSAP3 was verified by immunohistochemistry. Thus, a novel prediction model combining TRAP1, CAMSAP3 and TP53 was construct by multi-omics. Compared with the area under the curve, it demonstrated a significantly improvemrnt in the diagnostic efficacy in EC patients from TCGA bank. In conclusion, this work improved the current knowledge regarding the prognosis of early-stage EC through proteomics and scRNA-seq. These findings may lead to improvements in precise risk stratification of early-stage EC patients.

早期子宫内膜癌(EC)的临床异质性值得进一步研究,以确定高质量的预后标志物及其在侵袭性肿瘤行为中的潜在作用。TP53突变被认为是子宫内膜癌改良分子分型中重要的主要分型指标,但它仍不能准确预测子宫内膜癌的预后。对24例早期子宫内膜异位症患者的癌组织和癌旁组织进行蛋白质组学分析后,筛选出13种不同表达的蛋白质,并通过单细胞转录组(scRNA-seq)进一步鉴定出2种富含p53信号通路的蛋白质。有趣的是,在特定细胞群中,肿瘤坏死因子1型受体相关蛋白(TRAP1)和钙调素调控谱蛋白相关蛋白家族成员3(CAMSAP3)被显著下调。TRAP1-low/CAMSAP3-low细胞簇的特征基因包括经典的癌基因,这在意料之中。此外,在系统阐明髓系细胞与肿瘤微环境(TME)的关系后,发现髓系细胞与TRAP1low/CAMSAP3low细胞群之间存在密切的细胞相互作用。TRAP1和CAMSAP3的表达通过免疫组化得到了验证。因此,多组学构建了一个结合TRAP1、CAMSAP3和TP53的新型预测模型。与曲线下面积相比,该模型明显提高了对 TCGA 库中 EC 患者的诊断效率。总之,这项研究通过蛋白质组学和 scRNA-seq,提高了人们对早期心肌梗死预后的认识。这些发现可能会改善早期EC患者的精确风险分层。
{"title":"Multi-omics profiling reveal cells with novel oncogenic cluster, TRAP1<sup>low</sup>/CAMSAP3<sup>low</sup>, emerge more aggressive behavior and poor-prognosis in early-stage endometrial cancer.","authors":"Xiaodan Mao, Xiaoyue Tang, Jingxuan Ye, Shuxia Xu, Yue Wang, Xianhua Liu, Qibin Wu, Xite Lin, Maotong Zhang, Jiangfeng Liu, Juntao Yang, Pengming Sun","doi":"10.1186/s12943-024-02039-2","DOIUrl":"10.1186/s12943-024-02039-2","url":null,"abstract":"<p><p>The clinical heterogeneity of early-stage endometrial cancer (EC) is worthy of further study to identify high-quality prognostic markers and their potential role in aggressive tumor behavior. Mutation of TP53 was considered as an important primary triage in modified molecular typing for EC, it still cannot precisely predict the prognosis of EC. After proteomic analysis of cancer and para-cancerous tissues from 24 early-stage endometrioid EC patients with different survival outcomes, 13 differentially expressed proteins were screen out while 2 proteins enriched in p53 signaling pathway were further identified by single-cell transcriptome (scRNA-seq). Interestingly, tumor necrosis factor type-1 receptor-associated protein (TRAP1) and calmodulin-regulated spectrin-associated protein family member 3 (CAMSAP3) were found to be significantly downregulated in the specific cell cluster. Expectedly, the signature genes of TRAP1<sup>low</sup>/CAMSAP3<sup>low</sup> cluster included classical oncogenes. Moreover, close cellular interactions were observed between myeloid cells and the TRAP1<sup>low</sup>/CAMSAP3<sup>low</sup> cluster after systematically elucidating their relationship with tumor microenvironment (TME). The expression of TRAP1 and CAMSAP3 was verified by immunohistochemistry. Thus, a novel prediction model combining TRAP1, CAMSAP3 and TP53 was construct by multi-omics. Compared with the area under the curve, it demonstrated a significantly improvemrnt in the diagnostic efficacy in EC patients from TCGA bank. In conclusion, this work improved the current knowledge regarding the prognosis of early-stage EC through proteomics and scRNA-seq. These findings may lead to improvements in precise risk stratification of early-stage EC patients.</p>","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11181528/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141331477","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
Mechanism of ERBB2 gene overexpression by the formation of super-enhancer with genomic structural abnormalities in lung adenocarcinoma without clinically actionable genetic alterations 在无临床可操作基因改变的肺腺癌中,通过形成基因组结构异常的超级增强子实现 ERBB2 基因过表达的机制
IF 37.3 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-06-11 DOI: 10.1186/s12943-024-02035-6
Syuzo Kaneko, Ken Takasawa, Ken Asada, Kouya Shiraishi, Noriko Ikawa, Hidenori Machino, Norio Shinkai, Maiko Matsuda, Mari Masuda, Shungo Adachi, Satoshi Takahashi, Kazuma Kobayashi, Nobuji Kouno, Amina Bolatkan, Masaaki Komatsu, Masayoshi Yamada, Mototaka Miyake, Hirokazu Watanabe, Akiko Tateishi, Takaaki Mizuno, Yu Okubo, Masami Mukai, Tatsuya Yoshida, Yukihiro Yoshida, Hidehito Horinouchi, Shun-Ichi Watanabe, Yuichiro Ohe, Yasushi Yatabe, Vassiliki Saloura, Takashi Kohno, Ryuji Hamamoto
In an extensive genomic analysis of lung adenocarcinomas (LUADs), driver mutations have been recognized as potential targets for molecular therapy. However, there remain cases where target genes are not identified. Super-enhancers and structural variants are frequently identified in several hundred loci per case. Despite this, most cancer research has approached the analysis of these data sets separately, without merging and comparing the data, and there are no examples of integrated analysis in LUAD. We performed an integrated analysis of super-enhancers and structural variants in a cohort of 174 LUAD cases that lacked clinically actionable genetic alterations. To achieve this, we conducted both WGS and H3K27Ac ChIP-seq analyses using samples with driver gene mutations and those without, allowing for a comprehensive investigation of the potential roles of super-enhancer in LUAD cases. We demonstrate that most genes situated in these overlapped regions were associated with known and previously unknown driver genes and aberrant expression resulting from the formation of super-enhancers accompanied by genomic structural abnormalities. Hi-C and long-read sequencing data further corroborated this insight. When we employed CRISPR-Cas9 to induce structural abnormalities that mimicked cases with outlier ERBB2 gene expression, we observed an elevation in ERBB2 expression. These abnormalities are associated with a higher risk of recurrence after surgery, irrespective of the presence or absence of driver mutations. Our findings suggest that aberrant gene expression linked to structural polymorphisms can significantly impact personalized cancer treatment by facilitating the identification of driver mutations and prognostic factors, contributing to a more comprehensive understanding of LUAD pathogenesis.
在对肺腺癌(LUADs)进行的广泛基因组分析中,驱动突变被认为是分子疗法的潜在靶点。然而,仍有一些病例的靶基因未被确定。超级增强子和结构变异经常在每个病例的几百个位点中被发现。尽管如此,大多数癌症研究都是单独分析这些数据集,而没有对数据进行合并和比较,也没有对 LUAD 进行综合分析的实例。我们对一组 174 例缺乏临床可操作基因改变的 LUAD 病例中的超级增强子和结构变异进行了综合分析。为此,我们利用有驱动基因突变和无驱动基因突变的样本进行了 WGS 和 H3K27Ac ChIP-seq 分析,从而对超级增强子在 LUAD 病例中的潜在作用进行了全面研究。我们发现,位于这些重叠区域的大多数基因都与已知和以前未知的驱动基因有关,超级增强子的形成导致异常表达,并伴有基因组结构异常。Hi-C和长线程测序数据进一步证实了这一观点。当我们使用 CRISPR-Cas9 诱导结构异常,模拟 ERBB2 基因表达异常的病例时,我们观察到 ERBB2 表达的升高。无论是否存在驱动基因突变,这些异常都与术后复发风险较高有关。我们的研究结果表明,与结构多态性相关的异常基因表达可以促进驱动基因突变和预后因素的识别,从而对个性化癌症治疗产生重大影响,有助于更全面地了解 LUAD 的发病机制。
{"title":"Mechanism of ERBB2 gene overexpression by the formation of super-enhancer with genomic structural abnormalities in lung adenocarcinoma without clinically actionable genetic alterations","authors":"Syuzo Kaneko, Ken Takasawa, Ken Asada, Kouya Shiraishi, Noriko Ikawa, Hidenori Machino, Norio Shinkai, Maiko Matsuda, Mari Masuda, Shungo Adachi, Satoshi Takahashi, Kazuma Kobayashi, Nobuji Kouno, Amina Bolatkan, Masaaki Komatsu, Masayoshi Yamada, Mototaka Miyake, Hirokazu Watanabe, Akiko Tateishi, Takaaki Mizuno, Yu Okubo, Masami Mukai, Tatsuya Yoshida, Yukihiro Yoshida, Hidehito Horinouchi, Shun-Ichi Watanabe, Yuichiro Ohe, Yasushi Yatabe, Vassiliki Saloura, Takashi Kohno, Ryuji Hamamoto","doi":"10.1186/s12943-024-02035-6","DOIUrl":"https://doi.org/10.1186/s12943-024-02035-6","url":null,"abstract":"In an extensive genomic analysis of lung adenocarcinomas (LUADs), driver mutations have been recognized as potential targets for molecular therapy. However, there remain cases where target genes are not identified. Super-enhancers and structural variants are frequently identified in several hundred loci per case. Despite this, most cancer research has approached the analysis of these data sets separately, without merging and comparing the data, and there are no examples of integrated analysis in LUAD. We performed an integrated analysis of super-enhancers and structural variants in a cohort of 174 LUAD cases that lacked clinically actionable genetic alterations. To achieve this, we conducted both WGS and H3K27Ac ChIP-seq analyses using samples with driver gene mutations and those without, allowing for a comprehensive investigation of the potential roles of super-enhancer in LUAD cases. We demonstrate that most genes situated in these overlapped regions were associated with known and previously unknown driver genes and aberrant expression resulting from the formation of super-enhancers accompanied by genomic structural abnormalities. Hi-C and long-read sequencing data further corroborated this insight. When we employed CRISPR-Cas9 to induce structural abnormalities that mimicked cases with outlier ERBB2 gene expression, we observed an elevation in ERBB2 expression. These abnormalities are associated with a higher risk of recurrence after surgery, irrespective of the presence or absence of driver mutations. Our findings suggest that aberrant gene expression linked to structural polymorphisms can significantly impact personalized cancer treatment by facilitating the identification of driver mutations and prognostic factors, contributing to a more comprehensive understanding of LUAD pathogenesis.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-06-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141304328","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Cell surface CD55 traffics to the nucleus leading to cisplatin resistance and stemness by inducing PRC2 and H3K27 trimethylation on chromatin in ovarian cancer. 细胞表面CD55通过诱导PRC2和染色质上的H3K27三甲基化迁移到细胞核,导致卵巢癌的顺铂耐药性和干性。
IF 37.3 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-06-10 DOI: 10.1186/s12943-024-02028-5
Rashmi Bharti, Goutam Dey, Debjit Khan, Alex Myers, Olivia G Huffman, Caner Saygin, Chad Braley, Elliott Richards, Naseer Sangwan, Belinda Willard, Justin D Lathia, Paul L Fox, Feng Lin, Babal Kant Jha, J Mark Brown, Jennifer S Yu, Mohammed Dwidar, Amy Joehlin-Price, Roberto Vargas, Chad M Michener, Michelle S Longworth, Ofer Reizes

Background: Platinum resistance is the primary cause of poor survival in ovarian cancer (OC) patients. Targeted therapies and biomarkers of chemoresistance are critical for the treatment of OC patients. Our previous studies identified cell surface CD55, a member of the complement regulatory proteins, drives chemoresistance and maintenance of cancer stem cells (CSCs). CSCs are implicated in tumor recurrence and metastasis in multiple cancers.

Methods: Protein localization assays including immunofluorescence and subcellular fractionation were used to identify CD55 at the cell surface and nucleus of cancer cells. Protein half-life determinations were used to compare cell surface and nuclear CD55 stability. CD55 deletion mutants were generated and introduced into cancer cells to identify the nuclear trafficking code, cisplatin sensitivity, and stem cell frequency that were assayed using in vitro and in vivo models. Detection of CD55 binding proteins was analyzed by immunoprecipitation followed by mass spectrometry. Target pathways activated by CD55 were identified by RNA sequencing.

Results: CD55 localizes to the nucleus of a subset of OC specimens, ascites from chemoresistant patients, and enriched in chemoresistant OC cells. We determined that nuclear CD55 is glycosylated and derived from the cell surface pool of CD55. Nuclear localization is driven by a trafficking code containing the serine/threonine (S/T) domain of CD55. Nuclear CD55 is necessary for cisplatin resistance, stemness, and cell proliferation in OC cells. CD55 S/T domain is necessary for nuclear entry and inducing chemoresistance to cisplatin in both in vitro and in vivo models. Deletion of the CD55 S/T domain is sufficient to sensitize chemoresistant OC cells to cisplatin. In the nucleus, CD55 binds and attenuates the epigenetic regulator and tumor suppressor ZMYND8 with a parallel increase in H3K27 trimethylation and members of the Polycomb Repressive Complex 2.

Conclusions: For the first time, we show CD55 localizes to the nucleus in OC and promotes CSC and chemoresistance. Our studies identify a therapeutic mechanism for treating platinum resistant ovarian cancer by blocking CD55 nuclear entry.

背景:铂类抗药性是卵巢癌(OC)患者生存率低下的主要原因。靶向疗法和化疗耐药性生物标志物对治疗卵巢癌患者至关重要。我们之前的研究发现,细胞表面 CD55(补体调节蛋白的成员之一)可驱动化疗耐药性和癌症干细胞(CSCs)的维持。CSCs与多种癌症的肿瘤复发和转移有关:方法:采用免疫荧光和亚细胞分馏等蛋白质定位检测方法来鉴定癌细胞细胞表面和细胞核中的 CD55。蛋白质半衰期测定用于比较细胞表面和细胞核 CD55 的稳定性。生成 CD55 基因缺失突变体并将其导入癌细胞,以确定核转运密码、顺铂敏感性和干细胞频率,并使用体外和体内模型进行检测。CD55结合蛋白的检测是通过免疫沉淀法和质谱法进行分析的。通过 RNA 测序确定了 CD55 激活的靶途径:结果:CD55定位于一部分OC标本和化疗耐药患者腹水的细胞核中,并富集于化疗耐药的OC细胞中。我们确定核CD55是糖基化的,来自细胞表面的CD55池。核定位由包含 CD55 的丝氨酸/苏氨酸(S/T)结构域的转运代码驱动。核 CD55 是 OC 细胞耐顺铂性、干性和细胞增殖的必要条件。在体外和体内模型中,CD55 S/T 结构域是核进入和诱导顺铂化疗耐药性的必要条件。CD55 S/T结构域的缺失足以使对顺铂具有化疗耐药性的OC细胞对顺铂敏感。在细胞核中,CD55与表观遗传调节因子和肿瘤抑制因子ZMYND8结合并减弱其作用,同时增加H3K27三甲基化和多角体抑制复合体2的成员:我们首次发现 CD55 在 OC 中定位到细胞核,并促进 CSC 和化疗耐受性。我们的研究发现了一种通过阻断 CD55 进入细胞核来治疗铂类耐药卵巢癌的治疗机制。
{"title":"Cell surface CD55 traffics to the nucleus leading to cisplatin resistance and stemness by inducing PRC2 and H3K27 trimethylation on chromatin in ovarian cancer.","authors":"Rashmi Bharti, Goutam Dey, Debjit Khan, Alex Myers, Olivia G Huffman, Caner Saygin, Chad Braley, Elliott Richards, Naseer Sangwan, Belinda Willard, Justin D Lathia, Paul L Fox, Feng Lin, Babal Kant Jha, J Mark Brown, Jennifer S Yu, Mohammed Dwidar, Amy Joehlin-Price, Roberto Vargas, Chad M Michener, Michelle S Longworth, Ofer Reizes","doi":"10.1186/s12943-024-02028-5","DOIUrl":"10.1186/s12943-024-02028-5","url":null,"abstract":"<p><strong>Background: </strong>Platinum resistance is the primary cause of poor survival in ovarian cancer (OC) patients. Targeted therapies and biomarkers of chemoresistance are critical for the treatment of OC patients. Our previous studies identified cell surface CD55, a member of the complement regulatory proteins, drives chemoresistance and maintenance of cancer stem cells (CSCs). CSCs are implicated in tumor recurrence and metastasis in multiple cancers.</p><p><strong>Methods: </strong>Protein localization assays including immunofluorescence and subcellular fractionation were used to identify CD55 at the cell surface and nucleus of cancer cells. Protein half-life determinations were used to compare cell surface and nuclear CD55 stability. CD55 deletion mutants were generated and introduced into cancer cells to identify the nuclear trafficking code, cisplatin sensitivity, and stem cell frequency that were assayed using in vitro and in vivo models. Detection of CD55 binding proteins was analyzed by immunoprecipitation followed by mass spectrometry. Target pathways activated by CD55 were identified by RNA sequencing.</p><p><strong>Results: </strong>CD55 localizes to the nucleus of a subset of OC specimens, ascites from chemoresistant patients, and enriched in chemoresistant OC cells. We determined that nuclear CD55 is glycosylated and derived from the cell surface pool of CD55. Nuclear localization is driven by a trafficking code containing the serine/threonine (S/T) domain of CD55. Nuclear CD55 is necessary for cisplatin resistance, stemness, and cell proliferation in OC cells. CD55 S/T domain is necessary for nuclear entry and inducing chemoresistance to cisplatin in both in vitro and in vivo models. Deletion of the CD55 S/T domain is sufficient to sensitize chemoresistant OC cells to cisplatin. In the nucleus, CD55 binds and attenuates the epigenetic regulator and tumor suppressor ZMYND8 with a parallel increase in H3K27 trimethylation and members of the Polycomb Repressive Complex 2.</p><p><strong>Conclusions: </strong>For the first time, we show CD55 localizes to the nucleus in OC and promotes CSC and chemoresistance. Our studies identify a therapeutic mechanism for treating platinum resistant ovarian cancer by blocking CD55 nuclear entry.</p>","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-06-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11163727/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141296463","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
Exosomal circSIPA1L3-mediated intercellular communication contributes to glucose metabolic reprogramming and progression of triple negative breast cancer. 外泌体circSIPA1L3介导的细胞间通讯有助于葡萄糖代谢重编程和三阴性乳腺癌的进展。
IF 37.3 1区 医学 Q1 Biochemistry, Genetics and Molecular Biology Pub Date : 2024-06-08 DOI: 10.1186/s12943-024-02037-4
Yiran Liang, Fangzhou Ye, Dan Luo, Li Long, Yajie Wang, Yuhan Jin, Lei Wang, Yaming Li, Dianwen Han, Bing Chen, Wenjing Zhao, Lijuan Wang, Qifeng Yang

Background: Breast cancer is the most common malignant tumor, and metastasis remains the major cause of poor prognosis. Glucose metabolic reprogramming is one of the prominent hallmarks in cancer, providing nutrients and energy to support dramatically elevated tumor growth and metastasis. Nevertheless, the potential mechanistic links between glycolysis and breast cancer progression have not been thoroughly elucidated.

Methods: RNA-seq analysis was used to identify glucose metabolism-related circRNAs. The expression of circSIPA1L3 in breast cancer tissues and serum was examined by qRT-PCR, and further assessed its diagnostic value. We also evaluated the prognostic potential of circSIPA1L3 by analyzing a cohort of 238 breast cancer patients. Gain- and loss-of-function experiments, transcriptomic analysis, and molecular biology experiments were conducted to explore the biological function and regulatory mechanism of circSIPA1L3.

Results: Using RNA-seq analysis, circSIPA1L3 was identified as the critical mediator responsible for metabolic adaption upon energy stress. Gain- and loss-of-function experiments revealed that circSIPA1L3 exerted a stimulative effect on breast cancer progression and glycolysis, which could also be transported by exosomes and facilitated malignant behaviors among breast cancer cells. Significantly, the elevated lactate secretion caused by circSIPA1L3-mediated glycolysis enhancement promoted the recruitment of tumor associated macrophage and their tumor-promoting roles. Mechanistically, EIF4A3 induced the cyclization and cytoplasmic export of circSIPA1L3, which inhibited ubiquitin-mediated IGF2BP3 degradation through enhancing the UPS7-IGF2BP3 interaction. Furthermore, circSIPA1L3 increased mRNA stability of the lactate export carrier SLC16A1 and the glucose intake enhancer RAB11A through either strengthening their interaction with IGF2BP3 or sponging miR-665, leading to enhanced glycolytic metabolism. Clinically, elevated circSIPA1L3 expression indicated unfavorable prognosis base on the cohort of 238 breast cancer patients. Moreover, circSIPA1L3 was highly expressed in the serum of breast cancer patients and exhibited high diagnostic value for breast cancer patients.

Conclusions: Our study highlights the oncogenic role of circSIPA1L3 through mediating glucose metabolism, which might serve as a promising diagnostic and prognostic biomarker and potential therapeutic target for breast cancer.

背景:乳腺癌是最常见的恶性肿瘤,转移仍然是预后不良的主要原因。糖代谢重编程是癌症的显著特征之一,它为急剧增加的肿瘤生长和转移提供营养和能量。然而,糖酵解与乳腺癌进展之间的潜在机理联系尚未得到彻底阐明:方法:利用 RNA-seq 分析鉴定与糖代谢相关的 circRNA。方法:我们利用 RNA-seq 分析鉴定了葡萄糖代谢相关的 circRNAs,并通过 qRT-PCR 检测了 circSIPA1L3 在乳腺癌组织和血清中的表达,进一步评估了其诊断价值。我们还通过分析一组 238 例乳腺癌患者评估了 circSIPA1L3 的预后潜力。为了探索circSIPA1L3的生物学功能和调控机制,我们进行了功能增益和功能缺失实验、转录组分析和分子生物学实验:结果:通过RNA-seq分析,circSIPA1L3被确定为能量应激时负责代谢适应的关键介质。功能增益和功能缺失实验显示,circSIPA1L3对乳腺癌的进展和糖酵解有刺激作用,它还能通过外泌体运输,促进乳腺癌细胞的恶性行为。值得注意的是,circSIPA1L3介导的糖酵解增强所引起的乳酸分泌增加促进了肿瘤相关巨噬细胞的招募,并使其发挥促瘤作用。从机制上看,EIF4A3诱导circSIPA1L3环化和胞质输出,通过增强UPS7-IGF2BP3相互作用,抑制泛素介导的IGF2BP3降解。此外,circSIPA1L3 还通过加强乳酸输出载体 SLC16A1 和葡萄糖摄入增强子 RAB11A 与 IGF2BP3 的相互作用或疏导 miR-665 来增加它们的 mRNA 稳定性,从而导致糖酵解代谢的增强。在临床上,基于 238 例乳腺癌患者,circSIPA1L3 表达升高表明预后不良。此外,circSIPA1L3在乳腺癌患者血清中高表达,对乳腺癌患者具有很高的诊断价值:我们的研究强调了circSIPA1L3通过介导葡萄糖代谢所起的致癌作用,它可能成为一种有前途的乳腺癌诊断和预后生物标志物以及潜在的治疗靶点。
{"title":"Exosomal circSIPA1L3-mediated intercellular communication contributes to glucose metabolic reprogramming and progression of triple negative breast cancer.","authors":"Yiran Liang, Fangzhou Ye, Dan Luo, Li Long, Yajie Wang, Yuhan Jin, Lei Wang, Yaming Li, Dianwen Han, Bing Chen, Wenjing Zhao, Lijuan Wang, Qifeng Yang","doi":"10.1186/s12943-024-02037-4","DOIUrl":"10.1186/s12943-024-02037-4","url":null,"abstract":"<p><strong>Background: </strong>Breast cancer is the most common malignant tumor, and metastasis remains the major cause of poor prognosis. Glucose metabolic reprogramming is one of the prominent hallmarks in cancer, providing nutrients and energy to support dramatically elevated tumor growth and metastasis. Nevertheless, the potential mechanistic links between glycolysis and breast cancer progression have not been thoroughly elucidated.</p><p><strong>Methods: </strong>RNA-seq analysis was used to identify glucose metabolism-related circRNAs. The expression of circSIPA1L3 in breast cancer tissues and serum was examined by qRT-PCR, and further assessed its diagnostic value. We also evaluated the prognostic potential of circSIPA1L3 by analyzing a cohort of 238 breast cancer patients. Gain- and loss-of-function experiments, transcriptomic analysis, and molecular biology experiments were conducted to explore the biological function and regulatory mechanism of circSIPA1L3.</p><p><strong>Results: </strong>Using RNA-seq analysis, circSIPA1L3 was identified as the critical mediator responsible for metabolic adaption upon energy stress. Gain- and loss-of-function experiments revealed that circSIPA1L3 exerted a stimulative effect on breast cancer progression and glycolysis, which could also be transported by exosomes and facilitated malignant behaviors among breast cancer cells. Significantly, the elevated lactate secretion caused by circSIPA1L3-mediated glycolysis enhancement promoted the recruitment of tumor associated macrophage and their tumor-promoting roles. Mechanistically, EIF4A3 induced the cyclization and cytoplasmic export of circSIPA1L3, which inhibited ubiquitin-mediated IGF2BP3 degradation through enhancing the UPS7-IGF2BP3 interaction. Furthermore, circSIPA1L3 increased mRNA stability of the lactate export carrier SLC16A1 and the glucose intake enhancer RAB11A through either strengthening their interaction with IGF2BP3 or sponging miR-665, leading to enhanced glycolytic metabolism. Clinically, elevated circSIPA1L3 expression indicated unfavorable prognosis base on the cohort of 238 breast cancer patients. Moreover, circSIPA1L3 was highly expressed in the serum of breast cancer patients and exhibited high diagnostic value for breast cancer patients.</p><p><strong>Conclusions: </strong>Our study highlights the oncogenic role of circSIPA1L3 through mediating glucose metabolism, which might serve as a promising diagnostic and prognostic biomarker and potential therapeutic target for breast cancer.</p>","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-06-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11161950/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141288356","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
Molecular Cancer
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