首页 > 最新文献

Molecular Cancer最新文献

英文 中文
NAC1 promotes stemness and regulates myeloid-derived cell status in triple-negative breast cancer NAC1 在三阴性乳腺癌中促进干性并调节髓源性细胞状态
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-06 DOI: 10.1186/s12943-024-02102-y
Chrispus Ngule, Ruyi Shi, Xingcong Ren, Hongyan Jia, Felix Oyelami, Dong Li, Younhee Park, Jinhwan Kim, Hami Hemati, Yi Zhang, Xiaofang Xiong, Andrew Shinkle, Nathan L. Vanderford, Sara Bachert, Binhua P. Zhou, Jianlong Wang, Jianxun Song, Xia Liu, Jin-Ming Yang
Triple negative breast cancer (TNBC) is a particularly lethal breast cancer (BC) subtype driven by cancer stem cells (CSCs) and an immunosuppressive microenvironment. Our study reveals that nucleus accumbens associated protein 1 (NAC1), a member of the BTB/POZ gene family, plays a crucial role in TNBC by maintaining tumor stemness and influencing myeloid-derived suppressor cells (MDSCs). High NAC1 expression correlates with worse TNBC prognosis. NAC1 knockdown reduced CSC markers and tumor cell proliferation, migration, and invasion. Additionally, NAC1 affects oncogenic pathways such as the CD44-JAK1-STAT3 axis and immunosuppressive signals (TGFβ, IL-6). Intriguingly, the impact of NAC1 on tumor growth varies with the host immune status, showing diminished tumorigenicity in natural killer (NK) cell-competent mice but increased tumorigenicity in NK cell-deficient ones. This highlights the important role of the host immune system in TNBC progression. In addition, high NAC1 level in MDSCs also supports TNBC stemness. Together, this study implies NAC1 as a promising therapeutic target able to simultaneously eradicate CSCs and mitigate immune evasion.
三阴性乳腺癌(TNBC)是一种特别致命的乳腺癌(BC)亚型,由癌症干细胞(CSC)和免疫抑制微环境驱动。我们的研究发现,BTB/POZ基因家族成员之一的核团相关蛋白1(NAC1)通过维持肿瘤干性和影响髓源性抑制细胞(MDSCs),在TNBC中发挥着至关重要的作用。NAC1的高表达与TNBC的不良预后相关。敲除 NAC1 可减少 CSC 标记和肿瘤细胞的增殖、迁移和侵袭。此外,NAC1 还影响致癌通路,如 CD44-JAK1-STAT3 轴和免疫抑制信号(TGFβ、IL-6)。耐人寻味的是,NAC1 对肿瘤生长的影响随宿主免疫状态的不同而变化,在自然杀伤(NK)细胞功能健全的小鼠中,其致瘤性减弱,而在 NK 细胞功能缺陷的小鼠中,其致瘤性增强。这凸显了宿主免疫系统在 TNBC 进展中的重要作用。此外,MDSCs中的高NAC1水平也支持TNBC的干性。总之,这项研究表明,NAC1是一个很有前景的治疗靶点,能够同时消灭癌细胞干细胞和减轻免疫逃避。
{"title":"NAC1 promotes stemness and regulates myeloid-derived cell status in triple-negative breast cancer","authors":"Chrispus Ngule, Ruyi Shi, Xingcong Ren, Hongyan Jia, Felix Oyelami, Dong Li, Younhee Park, Jinhwan Kim, Hami Hemati, Yi Zhang, Xiaofang Xiong, Andrew Shinkle, Nathan L. Vanderford, Sara Bachert, Binhua P. Zhou, Jianlong Wang, Jianxun Song, Xia Liu, Jin-Ming Yang","doi":"10.1186/s12943-024-02102-y","DOIUrl":"https://doi.org/10.1186/s12943-024-02102-y","url":null,"abstract":"Triple negative breast cancer (TNBC) is a particularly lethal breast cancer (BC) subtype driven by cancer stem cells (CSCs) and an immunosuppressive microenvironment. Our study reveals that nucleus accumbens associated protein 1 (NAC1), a member of the BTB/POZ gene family, plays a crucial role in TNBC by maintaining tumor stemness and influencing myeloid-derived suppressor cells (MDSCs). High NAC1 expression correlates with worse TNBC prognosis. NAC1 knockdown reduced CSC markers and tumor cell proliferation, migration, and invasion. Additionally, NAC1 affects oncogenic pathways such as the CD44-JAK1-STAT3 axis and immunosuppressive signals (TGFβ, IL-6). Intriguingly, the impact of NAC1 on tumor growth varies with the host immune status, showing diminished tumorigenicity in natural killer (NK) cell-competent mice but increased tumorigenicity in NK cell-deficient ones. This highlights the important role of the host immune system in TNBC progression. In addition, high NAC1 level in MDSCs also supports TNBC stemness. Together, this study implies NAC1 as a promising therapeutic target able to simultaneously eradicate CSCs and mitigate immune evasion.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-09-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142142519","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
YTHDF2 in peritumoral hepatocytes mediates chemotherapy-induced antitumor immune responses through CX3CL1-mediated CD8+ T cell recruitment. 瘤周肝细胞中的 YTHDF2 通过 CX3CL1 介导的 CD8+ T 细胞募集介导化疗诱导的抗肿瘤免疫反应。
IF 27.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-06 DOI: 10.1186/s12943-024-02097-6
Zhenyun Yang, Xin Wang, Yizhen Fu, Weijie Wu, Zili Hu, Qingyang Lin, Wei Peng, Yangxun Pan, Juncheng Wang, Jinbin Chen, Dandan Hu, Zhongguo Zhou, Li Xu, Yaojun Zhang, Jiajie Hou, Minshan Chen

Peritumoral hepatocytes are critical components of the liver cancer microenvironment, However, the role of peritumoral hepatocytes in the local tumor immune interface and the underlying molecular mechanisms have not been elucidated. YTHDF2, an RNA N6-methyladenosine (m6A) reader, is critical for liver tumor progression. The function and regulatory roles of YTHDF2 in peritumoral hepatocytes are unknown. This study demonstrated that oxaliplatin (OXA) upregulated m6A modification and YTHDF2 expression in hepatocytes. Studies using tumor-bearing liver-specific Ythdf2 knockout mice revealed that hepatocyte YTHDF2 suppresses liver tumor growth through CD8+ T cell recruitment and activation. Additionally, YTHDF2 mediated the response to immunotherapy. Mechanistically, OXA upregulated YTHDF2 expression by activating the cGAS-STING signaling pathway and consequently enhanced the therapeutic outcomes of immunotherapeutic interventions. Ythdf2 stabilized Cx3cl1 transcripts in an m6A-dependent manner, regulating the interplay between CD8+ T cells and the progression of liver malignancies. Thus, this study elucidated the novel role of hepatocyte YTHDF2, which promotes therapy-induced antitumor immune responses in the liver. The findings of this study provide valuable insights into the mechanism underlying the therapeutic benefits of targeting YTHDF2.

瘤周肝细胞是肝癌微环境的重要组成部分,但瘤周肝细胞在局部肿瘤免疫界面中的作用及其潜在的分子机制尚未阐明。YTHDF2是一种RNA N6-甲基腺苷(m6A)阅读器,对肝脏肿瘤的进展至关重要。YTHDF2 在瘤周肝细胞中的功能和调控作用尚不清楚。该研究表明,奥沙利铂(OXA)可上调肝细胞中的m6A修饰和YTHDF2表达。使用肿瘤肝特异性 Ythdf2 基因敲除小鼠进行的研究表明,肝细胞 YTHDF2 可通过 CD8+ T 细胞的募集和活化抑制肝脏肿瘤的生长。此外,YTHDF2还能介导对免疫疗法的反应。从机制上讲,OXA通过激活cGAS-STING信号通路上调了YTHDF2的表达,从而增强了免疫治疗干预的疗效。Ythdf2以m6A依赖的方式稳定Cx3cl1转录本,调节CD8+ T细胞与肝脏恶性肿瘤进展之间的相互作用。因此,本研究阐明了肝细胞YTHDF2的新作用,它能促进治疗诱导的肝脏抗肿瘤免疫反应。本研究的发现为了解靶向YTHDF2的治疗益处的机制提供了宝贵的见解。
{"title":"YTHDF2 in peritumoral hepatocytes mediates chemotherapy-induced antitumor immune responses through CX3CL1-mediated CD8<sup>+</sup> T cell recruitment.","authors":"Zhenyun Yang, Xin Wang, Yizhen Fu, Weijie Wu, Zili Hu, Qingyang Lin, Wei Peng, Yangxun Pan, Juncheng Wang, Jinbin Chen, Dandan Hu, Zhongguo Zhou, Li Xu, Yaojun Zhang, Jiajie Hou, Minshan Chen","doi":"10.1186/s12943-024-02097-6","DOIUrl":"10.1186/s12943-024-02097-6","url":null,"abstract":"<p><p>Peritumoral hepatocytes are critical components of the liver cancer microenvironment, However, the role of peritumoral hepatocytes in the local tumor immune interface and the underlying molecular mechanisms have not been elucidated. YTHDF2, an RNA N<sup>6</sup>-methyladenosine (m<sup>6</sup>A) reader, is critical for liver tumor progression. The function and regulatory roles of YTHDF2 in peritumoral hepatocytes are unknown. This study demonstrated that oxaliplatin (OXA) upregulated m<sup>6</sup>A modification and YTHDF2 expression in hepatocytes. Studies using tumor-bearing liver-specific Ythdf2 knockout mice revealed that hepatocyte YTHDF2 suppresses liver tumor growth through CD8<sup>+</sup> T cell recruitment and activation. Additionally, YTHDF2 mediated the response to immunotherapy. Mechanistically, OXA upregulated YTHDF2 expression by activating the cGAS-STING signaling pathway and consequently enhanced the therapeutic outcomes of immunotherapeutic interventions. Ythdf2 stabilized Cx3cl1 transcripts in an m<sup>6</sup>A-dependent manner, regulating the interplay between CD8<sup>+</sup> T cells and the progression of liver malignancies. Thus, this study elucidated the novel role of hepatocyte YTHDF2, which promotes therapy-induced antitumor immune responses in the liver. The findings of this study provide valuable insights into the mechanism underlying the therapeutic benefits of targeting YTHDF2.</p>","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":27.7,"publicationDate":"2024-09-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11378438/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142140593","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
Unraveling the key role of chromatin structure in cancer development through epigenetic landscape characterization of oral cancer 通过对口腔癌的表观遗传景观特征分析,揭示染色质结构在癌症发展中的关键作用
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-06 DOI: 10.1186/s12943-024-02100-0
Yue Xue, Lu Liu, Ye Zhang, Yueying He, Jingyao Wang, Zicheng Ma, Tie-jun Li, Jianyun Zhang, Yanyi Huang, Yi Qin Gao
Epigenetic alterations, such as those in chromatin structure and DNA methylation, have been extensively studied in a number of tumor types. But oral cancer, particularly oral adenocarcinoma, has received far less attention. Here, we combined laser-capture microdissection and muti-omics mini-bulk sequencing to systematically characterize the epigenetic landscape of oral cancer, including chromatin architecture, DNA methylation, H3K27me3 modification, and gene expression. In carcinogenesis, tumor cells exhibit reorganized chromatin spatial structures, including compromised compartment structures and altered gene-gene interaction networks. Notably, some structural alterations are observed in phenotypically non-malignant paracancerous but not in normal cells. We developed transformer models to identify the cancer propensity of individual genome loci, thereby determining the carcinogenic status of each sample. Insights into cancer epigenetic landscapes provide evidence that chromatin reorganization is an important hallmark of oral cancer progression, which is also linked with genomic alterations and DNA methylation reprogramming. In particular, regions of frequent copy number alternations in cancer cells are associated with strong spatial insulation in both cancer and normal samples. Aberrant methylation reprogramming in oral squamous cell carcinomas is closely related to chromatin structure and H3K27me3 signals, which are further influenced by intrinsic sequence properties. Our findings indicate that structural changes are both significant and conserved in two distinct types of oral cancer, closely linked to transcriptomic alterations and cancer development. Notably, the structural changes remain markedly evident in oral adenocarcinoma despite the considerably lower incidence of genomic copy number alterations and lesser extent of methylation alterations compared to squamous cell carcinoma. We expect that the comprehensive analysis of epigenetic reprogramming of different types and subtypes of primary oral tumors can provide additional guidance to the design of novel detection and therapy for oral cancer.
表观遗传学改变,如染色质结构和 DNA 甲基化的改变,已在许多肿瘤类型中得到广泛研究。但口腔癌,尤其是口腔腺癌,受到的关注要少得多。在这里,我们结合激光捕获显微切割技术和突变组学微型批量测序技术,系统地描述了口腔癌的表观遗传学特征,包括染色质结构、DNA甲基化、H3K27me3修饰和基因表达。在癌变过程中,肿瘤细胞表现出染色质空间结构的重组,包括受损的区室结构和改变的基因-基因相互作用网络。值得注意的是,在表型为非恶性的癌旁细胞中观察到一些结构改变,而在正常细胞中则没有。我们开发了转化器模型来识别单个基因组位点的癌症倾向,从而确定每个样本的致癌状态。对癌症表观遗传景观的深入研究证明,染色质重组是口腔癌进展的一个重要标志,它还与基因组改变和DNA甲基化重编程有关。特别是,癌细胞中频繁发生拷贝数交替的区域与癌症和正常样本中强烈的空间隔离有关。口腔鳞状细胞癌中的异常甲基化重编程与染色质结构和 H3K27me3 信号密切相关,而染色质结构和 H3K27me3 信号又受到内在序列特性的影响。我们的研究结果表明,在两种不同类型的口腔癌中,结构变化既显著又保守,与转录组改变和癌症发展密切相关。值得注意的是,尽管与鳞状细胞癌相比,口腔腺癌的基因组拷贝数改变发生率低得多,甲基化改变程度也较低,但结构改变在口腔腺癌中仍然非常明显。我们期望,对不同类型和亚型的原发性口腔肿瘤的表观遗传学重编程进行全面分析,能为设计新型口腔癌检测和治疗方法提供更多指导。
{"title":"Unraveling the key role of chromatin structure in cancer development through epigenetic landscape characterization of oral cancer","authors":"Yue Xue, Lu Liu, Ye Zhang, Yueying He, Jingyao Wang, Zicheng Ma, Tie-jun Li, Jianyun Zhang, Yanyi Huang, Yi Qin Gao","doi":"10.1186/s12943-024-02100-0","DOIUrl":"https://doi.org/10.1186/s12943-024-02100-0","url":null,"abstract":"Epigenetic alterations, such as those in chromatin structure and DNA methylation, have been extensively studied in a number of tumor types. But oral cancer, particularly oral adenocarcinoma, has received far less attention. Here, we combined laser-capture microdissection and muti-omics mini-bulk sequencing to systematically characterize the epigenetic landscape of oral cancer, including chromatin architecture, DNA methylation, H3K27me3 modification, and gene expression. In carcinogenesis, tumor cells exhibit reorganized chromatin spatial structures, including compromised compartment structures and altered gene-gene interaction networks. Notably, some structural alterations are observed in phenotypically non-malignant paracancerous but not in normal cells. We developed transformer models to identify the cancer propensity of individual genome loci, thereby determining the carcinogenic status of each sample. Insights into cancer epigenetic landscapes provide evidence that chromatin reorganization is an important hallmark of oral cancer progression, which is also linked with genomic alterations and DNA methylation reprogramming. In particular, regions of frequent copy number alternations in cancer cells are associated with strong spatial insulation in both cancer and normal samples. Aberrant methylation reprogramming in oral squamous cell carcinomas is closely related to chromatin structure and H3K27me3 signals, which are further influenced by intrinsic sequence properties. Our findings indicate that structural changes are both significant and conserved in two distinct types of oral cancer, closely linked to transcriptomic alterations and cancer development. Notably, the structural changes remain markedly evident in oral adenocarcinoma despite the considerably lower incidence of genomic copy number alterations and lesser extent of methylation alterations compared to squamous cell carcinoma. We expect that the comprehensive analysis of epigenetic reprogramming of different types and subtypes of primary oral tumors can provide additional guidance to the design of novel detection and therapy for oral cancer.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-09-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142142539","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 prognostic effect of infiltrating immune cells is shaped by proximal M2 macrophages in lung adenocarcinoma 肺腺癌近端 M2 巨噬细胞决定浸润免疫细胞的预后效果
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-04 DOI: 10.1186/s12943-024-02080-1
Jian-Rong Li, Vikram Shaw, Yupei Lin, Xiang Wang, Muhammad Aminu, Yong Li, Jia Wu, Jianjun Zhang, Christopher I. Amos, Chao Cheng
The spatial arrangement of immune cells within the tumor microenvironment (TME) and their interactions play critical roles in the initiation and development of cancer. Several advanced technologies such as imaging mass cytometry (IMC) providing the immunological landscape of the TME with single-cell resolution. In this study, we develop a new method to quantify the spatial proximity between different cell types based on single-cell spatial data. Using this method on IMC data from 416 lung adenocarcinoma patients, we show that the proximity between different cell types is more correlated with patient prognosis compared to the traditional features such immune cell density and fractions. Consistent with previous reports, our results validate that proximity of T helper (Th) and B cells to cancer cells is associated with survival benefits. More importantly, we discover that the proximity of M2 macrophages to multiple immune cells is associated with poor prognosis. When Th/B cells are stratified into M2-distal and M2-proximal, the abundance of the former but not the latter category of Th/B cells is correlated with enhanced patient survival. Additionally, the abundance of M2-distal and M2-proximal cytotoxic T cells (Tc) is respectively associated with good and poor prognosis. Our results indicate that the prognostic effect of Th, Tc, and B cells in the tumor microenvironment is modulated by the nearby M2 macrophages. The proposed new method proposed can be readily applied to all single-cell spatial data for revealing functional impact of immune cell interactions.
肿瘤微环境(TME)中免疫细胞的空间排列及其相互作用在癌症的发生和发展过程中起着至关重要的作用。成像质谱细胞术(IMC)等一些先进技术以单细胞分辨率提供了肿瘤微环境的免疫学景观。在本研究中,我们根据单细胞空间数据开发了一种量化不同细胞类型之间空间接近性的新方法。通过对 416 例肺部腺癌患者的 IMC 数据使用这种方法,我们发现与免疫细胞密度和分数等传统特征相比,不同细胞类型之间的邻近性与患者预后的相关性更高。与之前的报告一致,我们的研究结果验证了 T 辅助细胞(Th)和 B 细胞与癌细胞的接近度与生存益处相关。更重要的是,我们发现 M2 巨噬细胞接近多种免疫细胞与不良预后有关。当 Th/B 细胞被分层为 M2 远端和 M2 近端时,前一类 Th/B 细胞的丰度而非后一类 Th/B 细胞的丰度与患者生存率的提高相关。此外,M2-远端和M2-近端细胞毒性T细胞(Tc)的数量分别与预后良好和预后不良相关。我们的研究结果表明,肿瘤微环境中 Th、Tc 和 B 细胞的预后效应受附近 M2 巨噬细胞的调节。所提出的新方法可随时应用于所有单细胞空间数据,以揭示免疫细胞相互作用的功能影响。
{"title":"The prognostic effect of infiltrating immune cells is shaped by proximal M2 macrophages in lung adenocarcinoma","authors":"Jian-Rong Li, Vikram Shaw, Yupei Lin, Xiang Wang, Muhammad Aminu, Yong Li, Jia Wu, Jianjun Zhang, Christopher I. Amos, Chao Cheng","doi":"10.1186/s12943-024-02080-1","DOIUrl":"https://doi.org/10.1186/s12943-024-02080-1","url":null,"abstract":"The spatial arrangement of immune cells within the tumor microenvironment (TME) and their interactions play critical roles in the initiation and development of cancer. Several advanced technologies such as imaging mass cytometry (IMC) providing the immunological landscape of the TME with single-cell resolution. In this study, we develop a new method to quantify the spatial proximity between different cell types based on single-cell spatial data. Using this method on IMC data from 416 lung adenocarcinoma patients, we show that the proximity between different cell types is more correlated with patient prognosis compared to the traditional features such immune cell density and fractions. Consistent with previous reports, our results validate that proximity of T helper (Th) and B cells to cancer cells is associated with survival benefits. More importantly, we discover that the proximity of M2 macrophages to multiple immune cells is associated with poor prognosis. When Th/B cells are stratified into M2-distal and M2-proximal, the abundance of the former but not the latter category of Th/B cells is correlated with enhanced patient survival. Additionally, the abundance of M2-distal and M2-proximal cytotoxic T cells (Tc) is respectively associated with good and poor prognosis. Our results indicate that the prognostic effect of Th, Tc, and B cells in the tumor microenvironment is modulated by the nearby M2 macrophages. The proposed new method proposed can be readily applied to all single-cell spatial data for revealing functional impact of immune cell interactions.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-09-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142130688","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 and clustering analyses reveal the mechanisms underlying unmet needs for patients with lung adenocarcinoma and identify potential therapeutic targets. 多组学和聚类分析揭示了肺腺癌患者未满足需求的内在机制,并确定了潜在的治疗靶点。
IF 27.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-02 DOI: 10.1186/s12943-024-02093-w
Ken Asada, Syuzo Kaneko, Ken Takasawa, Kouya Shiraishi, Norio Shinkai, Yoko Shimada, Satoshi Takahashi, Hidenori Machino, Kazuma Kobayashi, 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, Takashi Kohno, Ryuji Hamamoto

Background: The cancer genome contains several driver mutations. However, in some cases, no known drivers have been identified; these remaining areas of unmet needs, leading to limited progress in cancer therapy. Whole-genome sequencing (WGS) can identify non-coding alterations associated with the disease. Consequently, exploration of non-coding regions using WGS and other omics data such as ChIP-sequencing (ChIP-seq) to discern novel alterations and mechanisms related to tumorigenesis have been attractive these days.

Methods: Integrated multi-omics analyses, including WGS, ChIP-seq, DNA methylation, and RNA-sequencing (RNA-seq), were conducted on samples from patients with non-clinically actionable genetic alterations (non-CAGAs) in lung adenocarcinoma (LUAD). Second-level cluster analysis was performed to reinforce the correlations associated with patient survival, as identified by RNA-seq. Subsequent differential gene expression analysis was performed to identify potential druggable targets.

Results: Differences in H3K27ac marks in non-CAGAs LUAD were found and confirmed by analyzing RNA-seq data, in which mastermind-like transcriptional coactivator 2 (MAML2) was suppressed. The down-regulated genes whose expression was correlated to MAML2 expression were associated with patient prognosis. WGS analysis revealed somatic mutations associated with the H3K27ac marks in the MAML2 region and high levels of DNA methylation in MAML2 were observed in tumor samples. The second-level cluster analysis enabled patient stratification and subsequent analyses identified potential therapeutic target genes and treatment options.

Conclusions: We overcome the persistent challenges of identifying alterations or driver mutations in coding regions related to tumorigenesis through a novel approach combining multi-omics data with clinical information to reveal the molecular mechanisms underlying non-CAGAs LUAD, stratify patients to improve patient prognosis, and identify potential therapeutic targets. This approach may be applicable to studies of other cancers with unmet needs.

背景:癌症基因组包含多种驱动突变。然而,在某些情况下,尚未发现已知的驱动基因;这些领域的需求仍未得到满足,导致癌症治疗进展有限。全基因组测序(WGS)可以确定与疾病相关的非编码改变。因此,利用 WGS 和其他组学数据(如 ChIP-测序(ChIP-seq))对非编码区进行探索,以发现与肿瘤发生有关的新的改变和机制,如今已具有吸引力:方法:对肺腺癌(LUAD)中存在非临床可操作基因改变(non-CAGAs)的患者样本进行了多组学综合分析,包括WGS、ChIP-seq、DNA甲基化和RNA测序(RNA-seq)。进行了二级聚类分析,以加强 RNA-seq 确定的与患者生存相关的关联性。随后进行了差异基因表达分析,以确定潜在的药物靶点:结果:通过分析RNA-seq数据,发现并证实了非CAGAs LUAD中H3K27ac标记的差异,其中mastermind样转录辅激活子2(MAML2)受到抑制。与MAML2表达相关的下调基因与患者的预后有关。WGS分析揭示了与MAML2区域H3K27ac标记相关的体细胞突变,并在肿瘤样本中观察到MAML2的高水平DNA甲基化。二级聚类分析实现了患者分层,随后的分析确定了潜在的治疗靶基因和治疗方案:我们通过一种将多组学数据与临床信息相结合的新方法,克服了识别与肿瘤发生相关的编码区改变或驱动突变的长期挑战,揭示了非CAGAs LUAD的分子机制,对患者进行分层以改善患者预后,并确定了潜在的治疗靶点。这种方法可能适用于其他未满足需求的癌症研究。
{"title":"Multi-omics and clustering analyses reveal the mechanisms underlying unmet needs for patients with lung adenocarcinoma and identify potential therapeutic targets.","authors":"Ken Asada, Syuzo Kaneko, Ken Takasawa, Kouya Shiraishi, Norio Shinkai, Yoko Shimada, Satoshi Takahashi, Hidenori Machino, Kazuma Kobayashi, 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, Takashi Kohno, Ryuji Hamamoto","doi":"10.1186/s12943-024-02093-w","DOIUrl":"10.1186/s12943-024-02093-w","url":null,"abstract":"<p><strong>Background: </strong>The cancer genome contains several driver mutations. However, in some cases, no known drivers have been identified; these remaining areas of unmet needs, leading to limited progress in cancer therapy. Whole-genome sequencing (WGS) can identify non-coding alterations associated with the disease. Consequently, exploration of non-coding regions using WGS and other omics data such as ChIP-sequencing (ChIP-seq) to discern novel alterations and mechanisms related to tumorigenesis have been attractive these days.</p><p><strong>Methods: </strong>Integrated multi-omics analyses, including WGS, ChIP-seq, DNA methylation, and RNA-sequencing (RNA-seq), were conducted on samples from patients with non-clinically actionable genetic alterations (non-CAGAs) in lung adenocarcinoma (LUAD). Second-level cluster analysis was performed to reinforce the correlations associated with patient survival, as identified by RNA-seq. Subsequent differential gene expression analysis was performed to identify potential druggable targets.</p><p><strong>Results: </strong>Differences in H3K27ac marks in non-CAGAs LUAD were found and confirmed by analyzing RNA-seq data, in which mastermind-like transcriptional coactivator 2 (MAML2) was suppressed. The down-regulated genes whose expression was correlated to MAML2 expression were associated with patient prognosis. WGS analysis revealed somatic mutations associated with the H3K27ac marks in the MAML2 region and high levels of DNA methylation in MAML2 were observed in tumor samples. The second-level cluster analysis enabled patient stratification and subsequent analyses identified potential therapeutic target genes and treatment options.</p><p><strong>Conclusions: </strong>We overcome the persistent challenges of identifying alterations or driver mutations in coding regions related to tumorigenesis through a novel approach combining multi-omics data with clinical information to reveal the molecular mechanisms underlying non-CAGAs LUAD, stratify patients to improve patient prognosis, and identify potential therapeutic targets. This approach may be applicable to studies of other cancers with unmet needs.</p>","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":27.7,"publicationDate":"2024-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11367768/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142109680","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
LAMTOR1 decreased exosomal PD-L1 to enhance immunotherapy efficacy in non-small cell lung cancer LAMTOR1 可减少外泌体 PD-L1,从而提高非小细胞肺癌的免疫疗法疗效
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-02 DOI: 10.1186/s12943-024-02099-4
Bo Wu, Xin Huang, Xiang Shi, Meixi Jiang, Hongxu Liu, Li Zhao
Great progress has been made in utilizing immune checkpoint blockade (ICB) for the treatment of non-small-cell lung cancer (NSCLC). Therapies targeting programmed cell death protein 1 (PD-1) and its ligand PD-L1, expressed on tumor cells, have demonstrated potential in improving patient survival rates. An unresolved issue involves immune suppression induced by exosomal PD-L1 within the tumor microenvironment (TME), particularly regarding CD8+ T cells. Our study unveiled the crucial involvement of LAMTOR1 in suppressing the exosomes of PD-L1 and promoting CD8+ T cell infiltration in NSCLC. Through its interaction with HRS, LAMTOR1 facilitates PD-L1 lysosomal degradation, thereby reducing exosomal PD-L1 release. Notably, the ability of LAMTOR1 to promote PD-L1 lysosomal degradation relies on a specific ubiquitination site and an HRS binding sequence. The findings suggest that employing LAMTOR1 to construct peptides could serve as a promising strategy for bolstering the efficacy of immunotherapy in NSCLC. The discovery and comprehension of how LAMTOR1 inhibits the release of exosomal PD-L1 offer insights into potential therapeutic strategies for improving immunotherapy. It is imperative to conduct further research and clinical trials to investigate the feasibility and efficacy of targeting LAMTOR1 in NSCLC treatment.
利用免疫检查点阻断疗法(ICB)治疗非小细胞肺癌(NSCLC)已取得了巨大进展。针对肿瘤细胞上表达的程序性细胞死亡蛋白 1(PD-1)及其配体 PD-L1 的疗法在提高患者生存率方面已显示出潜力。一个悬而未决的问题涉及肿瘤微环境(TME)中外泌体 PD-L1 诱导的免疫抑制,尤其是对 CD8+ T 细胞的抑制。我们的研究揭示了LAMTOR1在抑制PD-L1外泌体和促进CD8+ T细胞浸润NSCLC中的关键作用。通过与HRS相互作用,LAMTOR1促进了PD-L1溶酶体降解,从而减少了PD-L1外泌体的释放。值得注意的是,LAMTOR1促进PD-L1溶酶体降解的能力依赖于一个特定的泛素化位点和一个HRS结合序列。研究结果表明,利用 LAMTOR1 构建多肽可能是提高 NSCLC 免疫疗法疗效的有效策略。发现和理解LAMTOR1如何抑制外泌体PD-L1的释放为改进免疫疗法的潜在治疗策略提供了启示。当务之急是开展进一步的研究和临床试验,以调查靶向 LAMTOR1 在 NSCLC 治疗中的可行性和疗效。
{"title":"LAMTOR1 decreased exosomal PD-L1 to enhance immunotherapy efficacy in non-small cell lung cancer","authors":"Bo Wu, Xin Huang, Xiang Shi, Meixi Jiang, Hongxu Liu, Li Zhao","doi":"10.1186/s12943-024-02099-4","DOIUrl":"https://doi.org/10.1186/s12943-024-02099-4","url":null,"abstract":"Great progress has been made in utilizing immune checkpoint blockade (ICB) for the treatment of non-small-cell lung cancer (NSCLC). Therapies targeting programmed cell death protein 1 (PD-1) and its ligand PD-L1, expressed on tumor cells, have demonstrated potential in improving patient survival rates. An unresolved issue involves immune suppression induced by exosomal PD-L1 within the tumor microenvironment (TME), particularly regarding CD8+ T cells. Our study unveiled the crucial involvement of LAMTOR1 in suppressing the exosomes of PD-L1 and promoting CD8+ T cell infiltration in NSCLC. Through its interaction with HRS, LAMTOR1 facilitates PD-L1 lysosomal degradation, thereby reducing exosomal PD-L1 release. Notably, the ability of LAMTOR1 to promote PD-L1 lysosomal degradation relies on a specific ubiquitination site and an HRS binding sequence. The findings suggest that employing LAMTOR1 to construct peptides could serve as a promising strategy for bolstering the efficacy of immunotherapy in NSCLC. The discovery and comprehension of how LAMTOR1 inhibits the release of exosomal PD-L1 offer insights into potential therapeutic strategies for improving immunotherapy. It is imperative to conduct further research and clinical trials to investigate the feasibility and efficacy of targeting LAMTOR1 in NSCLC treatment.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142118219","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
Programmed death receptor (PD-)1/PD-ligand (L)1 in urological cancers : the “all-around warrior” in immunotherapy 泌尿系统癌症中的程序性死亡受体(PD-)1/PD-配体(L)1:免疫疗法中的 "全能战士
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-09-02 DOI: 10.1186/s12943-024-02095-8
Qiang Liu, Yujing Guan, Shenglong Li
Programmed death receptor-1 (PD-1) and its ligand, programmed death ligand-1 (PD-L1) are essential molecules that are key in modulating immune responses. PD-L1 is constitutively expressed on various immune cells, epithelial cells, and cancer cells, where it functions as a co-stimulatory molecule capable of impairing T-cell mediated immune responses. Upon binding to PD-1 on activated T-cells, the PD-1/PD-L1 interaction triggers signaling pathways that can induce T-cell apoptosis or anergy, thereby facilitating the immune escape of tumors. In urological cancers, including bladder cancer (BCa), renal cell carcinoma (RCC), and prostate cancer (PCa), the upregulation of PD-L1 has been demonstrated. It is linked to poor prognosis and enhanced tumor immune evasion. Recent studies have highlighted the significant role of the PD-1/PD-L1 axis in the immune escape mechanisms of urological cancers. The interaction between PD-L1 and PD-1 on T-cells further contributes to immunosuppression by inhibiting T-cell activation and proliferation. Clinical applications of PD-1/PD-L1 checkpoint inhibitors have shown promising efficacy in treating advanced urological cancers, significantly improving patient outcomes. However, resistance to these therapies, either intrinsic or acquired, remains a significant challenge. This review aims to provide a comprehensive overview of the role of the PD-1/PD-L1 signaling pathway in urological cancers. We summarize the regulatory mechanism underlying PD-1 and PD-L1 expression and activity, including genetic, epigenetic, post-transcriptional, and post-translational modifications. Additionally, we discuss current clinical research on PD-1/PD-L1 inhibitors, their therapeutic potential, and the challenges associated with resistance. Understanding these mechanisms is crucial for developing new strategies to overcome therapeutic limitations and enhance the efficacy of cancer immunotherapy.
程序性死亡受体-1(PD-1)及其配体程序性死亡配体-1(PD-L1)是调节免疫反应的重要分子。PD-L1 在各种免疫细胞、上皮细胞和癌细胞上都有表达,它是一种协同刺激分子,能够损害 T 细胞介导的免疫反应。与活化 T 细胞上的 PD-1 结合后,PD-1/PD-L1 相互作用会触发信号通路,诱导 T 细胞凋亡或失活,从而促进肿瘤的免疫逃逸。在膀胱癌(BCa)、肾细胞癌(RCC)和前列腺癌(PCa)等泌尿系统癌症中,PD-L1 的上调已得到证实。这与预后不良和肿瘤免疫逃避能力增强有关。最近的研究强调了 PD-1/PD-L1 轴在泌尿系统癌症免疫逃避机制中的重要作用。T 细胞上的 PD-L1 和 PD-1 之间的相互作用通过抑制 T 细胞的活化和增殖进一步促进了免疫抑制。PD-1/PD-L1 检查点抑制剂的临床应用已显示出治疗晚期泌尿系统癌症的良好疗效,显著改善了患者的预后。然而,这些疗法的耐药性,无论是内在的还是后天获得的,仍然是一个重大挑战。本综述旨在全面概述 PD-1/PD-L1 信号通路在泌尿系统癌症中的作用。我们总结了 PD-1 和 PD-L1 表达和活性的调控机制,包括遗传、表观遗传、转录后和翻译后修饰。此外,我们还讨论了目前有关 PD-1/PD-L1 抑制剂的临床研究、其治疗潜力以及与耐药性相关的挑战。了解这些机制对于开发新的策略以克服治疗限制和提高癌症免疫疗法的疗效至关重要。
{"title":"Programmed death receptor (PD-)1/PD-ligand (L)1 in urological cancers : the “all-around warrior” in immunotherapy","authors":"Qiang Liu, Yujing Guan, Shenglong Li","doi":"10.1186/s12943-024-02095-8","DOIUrl":"https://doi.org/10.1186/s12943-024-02095-8","url":null,"abstract":"Programmed death receptor-1 (PD-1) and its ligand, programmed death ligand-1 (PD-L1) are essential molecules that are key in modulating immune responses. PD-L1 is constitutively expressed on various immune cells, epithelial cells, and cancer cells, where it functions as a co-stimulatory molecule capable of impairing T-cell mediated immune responses. Upon binding to PD-1 on activated T-cells, the PD-1/PD-L1 interaction triggers signaling pathways that can induce T-cell apoptosis or anergy, thereby facilitating the immune escape of tumors. In urological cancers, including bladder cancer (BCa), renal cell carcinoma (RCC), and prostate cancer (PCa), the upregulation of PD-L1 has been demonstrated. It is linked to poor prognosis and enhanced tumor immune evasion. Recent studies have highlighted the significant role of the PD-1/PD-L1 axis in the immune escape mechanisms of urological cancers. The interaction between PD-L1 and PD-1 on T-cells further contributes to immunosuppression by inhibiting T-cell activation and proliferation. Clinical applications of PD-1/PD-L1 checkpoint inhibitors have shown promising efficacy in treating advanced urological cancers, significantly improving patient outcomes. However, resistance to these therapies, either intrinsic or acquired, remains a significant challenge. This review aims to provide a comprehensive overview of the role of the PD-1/PD-L1 signaling pathway in urological cancers. We summarize the regulatory mechanism underlying PD-1 and PD-L1 expression and activity, including genetic, epigenetic, post-transcriptional, and post-translational modifications. Additionally, we discuss current clinical research on PD-1/PD-L1 inhibitors, their therapeutic potential, and the challenges associated with resistance. Understanding these mechanisms is crucial for developing new strategies to overcome therapeutic limitations and enhance the efficacy of cancer immunotherapy. ","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142118220","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
Cellular senescence and SASP in tumor progression and therapeutic opportunities 肿瘤进展中的细胞衰老和 SASP 以及治疗机会
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-31 DOI: 10.1186/s12943-024-02096-7
Zening Dong, Yahan Luo, Zhangchen Yuan, Yu Tian, Tianqiang Jin, Feng Xu
Cellular senescence (CS), a permanent and irreversible arrest of the cell cycle and proliferation leading to the degeneration of cellular structure and function, has been implicated in various key physiological and pathological processes, particularly in cancer. Initially, CS was recognized as a barrier to tumorigenesis, serving as an intrinsic defense mechanism to protect cells from malignant transformation. However, increasing evidence suggests that senescent cells can promote tumor progression to overt malignancy, primarily through a set of factors known as senescence-associated secretory phenotypes (SASPs), including chemokines, growth factors, cytokines, and stromal metalloproteinases. These factors significantly reshape the tumor microenvironment (TME), enabling tumors to evade immune destruction. Interestingly, some studies have also suggested that SASPs may impede tumor development by enhancing immunosurveillance. These opposing roles highlight the complexity and heterogeneity of CS and SASPs in diverse cancers. Consequently, there has been growing interest in pharmacological interventions targeting CS or SASPs in cancer therapy, such as senolytics and senomorphics, to either promote the clearance of senescent cells or mitigate the harmful effects of SASPs. In this review, we will interpret the concept of CS, delve into the role of SASPs in reshaping the TME, and summarize recent advances in anti-tumor strategies targeting CS or SASPs.
细胞衰老(CS)是细胞周期和增殖的一种永久性、不可逆转的停滞,会导致细胞结构和功能的退化。最初,CS 被认为是肿瘤发生的屏障,是保护细胞免受恶性转化的内在防御机制。然而,越来越多的证据表明,衰老细胞可促进肿瘤进展为明显的恶性肿瘤,主要是通过一系列称为衰老相关分泌表型(SASPs)的因子,包括趋化因子、生长因子、细胞因子和基质金属蛋白酶。这些因子极大地重塑了肿瘤微环境(TME),使肿瘤得以逃避免疫破坏。有趣的是,一些研究还表明 SASPs 可通过增强免疫监视来阻碍肿瘤的发展。这些相反的作用凸显了 CS 和 SASPs 在不同癌症中的复杂性和异质性。因此,人们越来越关注在癌症治疗中针对 CS 或 SASPs 的药物干预,如衰老剂和衰老形态剂,以促进衰老细胞的清除或减轻 SASPs 的有害影响。在这篇综述中,我们将解读CS的概念,深入探讨SASPs在重塑TME中的作用,并总结针对CS或SASPs的抗肿瘤策略的最新进展。
{"title":"Cellular senescence and SASP in tumor progression and therapeutic opportunities","authors":"Zening Dong, Yahan Luo, Zhangchen Yuan, Yu Tian, Tianqiang Jin, Feng Xu","doi":"10.1186/s12943-024-02096-7","DOIUrl":"https://doi.org/10.1186/s12943-024-02096-7","url":null,"abstract":"Cellular senescence (CS), a permanent and irreversible arrest of the cell cycle and proliferation leading to the degeneration of cellular structure and function, has been implicated in various key physiological and pathological processes, particularly in cancer. Initially, CS was recognized as a barrier to tumorigenesis, serving as an intrinsic defense mechanism to protect cells from malignant transformation. However, increasing evidence suggests that senescent cells can promote tumor progression to overt malignancy, primarily through a set of factors known as senescence-associated secretory phenotypes (SASPs), including chemokines, growth factors, cytokines, and stromal metalloproteinases. These factors significantly reshape the tumor microenvironment (TME), enabling tumors to evade immune destruction. Interestingly, some studies have also suggested that SASPs may impede tumor development by enhancing immunosurveillance. These opposing roles highlight the complexity and heterogeneity of CS and SASPs in diverse cancers. Consequently, there has been growing interest in pharmacological interventions targeting CS or SASPs in cancer therapy, such as senolytics and senomorphics, to either promote the clearance of senescent cells or mitigate the harmful effects of SASPs. In this review, we will interpret the concept of CS, delve into the role of SASPs in reshaping the TME, and summarize recent advances in anti-tumor strategies targeting CS or SASPs.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-08-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142101946","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
Joint single-cell genetic and transcriptomic analysis reveal pre-malignant SCP-like subclones in human neuroblastoma 单细胞遗传学和转录组学联合分析揭示了人类神经母细胞瘤的恶性前 SCP 样亚克隆
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-31 DOI: 10.1186/s12943-024-02091-y
Thale K. Olsen, Jörg Otte, Shenglin Mei, Bethel Tesfai Embaie, Polina Kameneva, Huaitao Cheng, Teng Gao, Vasilios Zachariadis, Ioanna Tsea, Åsa Björklund, Emil Kryukov, Ziyi Hou, Anna Johansson, Erik Sundström, Tommy Martinsson, Susanne Fransson, Jakob Stenman, Shahrzad Shirazi Fard, John Inge Johnsen, Per Kogner, Igor Adameyko, Martin Enge, Peter V. Kharchenko, Ninib Baryawno
Neuroblastoma (NB) is a heterogeneous embryonal malignancy and the deadliest tumor of infancy. It is a complex disease that can result in diverse clinical outcomes. In some children, tumors regress spontaneously. Others respond well to existing treatments. But for the high-risk group, which constitutes approximately 40% of all patients, the prognosis remains dire despite collaborative efforts in basic and clinical research. While its exact cellular origin is still under debate, NB is assumed to arise from the neural crest cell lineage including multipotent Schwann cell precursors (SCPs), which differentiate into sympatho-adrenal cell states eventually producing chromaffin cells and sympathoblasts. To investigate clonal development of neuroblastoma cell states, we performed haplotype-specific analysis of human tumor samples using single-cell multi-omics, including joint DNA/RNA sequencing of sorted single cells (DNTR-seq). Samples were also assessed using immunofluorescence stainings and fluorescence in-situ hybridization (FISH). Beyond adrenergic tumor cells, we identify subpopulations of aneuploid SCP-like cells, characterized by clonal expansion, whole-chromosome 17 gains, as well as expression programs of proliferation, apoptosis, and a non-immunomodulatory phenotype. Aneuploid pre-malignant SCP-like cells represent a novel feature of NB. Genetic evidence and tumor phylogeny suggest that these clones and malignant adrenergic populations originate from aneuploidy-prone cells of migrating neural crest or SCP origin, before lineage commitment to sympatho-adrenal cell states. Our findings expand the phenotypic spectrum of NB cell states. Considering the multipotency of SCPs in development, we suggest that the transformation of fetal SCPs may represent one possible mechanism of tumor initiation in NB with chromosome 17 aberrations as a characteristic element.
神经母细胞瘤(NB)是一种异质性胚胎性恶性肿瘤,也是婴儿期最致命的肿瘤。它是一种复杂的疾病,可导致不同的临床结果。有些患儿的肿瘤会自然消退。另一些则对现有的治疗方法反应良好。但是,对于约占所有患者 40% 的高危人群来说,尽管基础和临床研究人员通力合作,但预后仍然不容乐观。尽管其确切的细胞起源仍存在争议,但人们认为 NB 起源于神经嵴细胞系,包括多能的许旺细胞前体(SCPs),它们分化成交感肾上腺细胞状态,最终产生嗜铬细胞和交感母细胞。为了研究神经母细胞瘤细胞状态的克隆发展,我们使用单细胞多组学技术对人类肿瘤样本进行了单倍型特异性分析,包括对分选的单细胞进行DNA/RNA联合测序(DNTR-seq)。我们还利用免疫荧光染色和荧光原位杂交(FISH)对样本进行了评估。除了肾上腺素能肿瘤细胞外,我们还发现了非整倍体 SCP 样细胞亚群,其特点是克隆扩增、全染色体 17 增殖以及增殖、凋亡和非免疫调节表型的表达程序。非整倍体恶性前SCP样细胞代表了NB的一种新特征。遗传学证据和肿瘤系统发育表明,这些克隆和恶性肾上腺素能细胞群起源于易发生非整倍体的移行神经嵴细胞或SCP细胞,然后才进入交感肾上腺细胞状态。我们的发现扩大了 NB 细胞状态的表型谱。考虑到 SCP 在发育过程中的多潜能性,我们认为胎儿 SCP 的转化可能代表了以 17 号染色体畸变为特征要素的 NB 肿瘤发生的一种可能机制。
{"title":"Joint single-cell genetic and transcriptomic analysis reveal pre-malignant SCP-like subclones in human neuroblastoma","authors":"Thale K. Olsen, Jörg Otte, Shenglin Mei, Bethel Tesfai Embaie, Polina Kameneva, Huaitao Cheng, Teng Gao, Vasilios Zachariadis, Ioanna Tsea, Åsa Björklund, Emil Kryukov, Ziyi Hou, Anna Johansson, Erik Sundström, Tommy Martinsson, Susanne Fransson, Jakob Stenman, Shahrzad Shirazi Fard, John Inge Johnsen, Per Kogner, Igor Adameyko, Martin Enge, Peter V. Kharchenko, Ninib Baryawno","doi":"10.1186/s12943-024-02091-y","DOIUrl":"https://doi.org/10.1186/s12943-024-02091-y","url":null,"abstract":"Neuroblastoma (NB) is a heterogeneous embryonal malignancy and the deadliest tumor of infancy. It is a complex disease that can result in diverse clinical outcomes. In some children, tumors regress spontaneously. Others respond well to existing treatments. But for the high-risk group, which constitutes approximately 40% of all patients, the prognosis remains dire despite collaborative efforts in basic and clinical research. While its exact cellular origin is still under debate, NB is assumed to arise from the neural crest cell lineage including multipotent Schwann cell precursors (SCPs), which differentiate into sympatho-adrenal cell states eventually producing chromaffin cells and sympathoblasts. To investigate clonal development of neuroblastoma cell states, we performed haplotype-specific analysis of human tumor samples using single-cell multi-omics, including joint DNA/RNA sequencing of sorted single cells (DNTR-seq). Samples were also assessed using immunofluorescence stainings and fluorescence in-situ hybridization (FISH). Beyond adrenergic tumor cells, we identify subpopulations of aneuploid SCP-like cells, characterized by clonal expansion, whole-chromosome 17 gains, as well as expression programs of proliferation, apoptosis, and a non-immunomodulatory phenotype. Aneuploid pre-malignant SCP-like cells represent a novel feature of NB. Genetic evidence and tumor phylogeny suggest that these clones and malignant adrenergic populations originate from aneuploidy-prone cells of migrating neural crest or SCP origin, before lineage commitment to sympatho-adrenal cell states. Our findings expand the phenotypic spectrum of NB cell states. Considering the multipotency of SCPs in development, we suggest that the transformation of fetal SCPs may represent one possible mechanism of tumor initiation in NB with chromosome 17 aberrations as a characteristic element.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-08-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142100696","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
Targeting m7G-enriched circKDM1A prevents colorectal cancer progression 靶向富含 m7G 的 circKDM1A 可预防结直肠癌进展
IF 37.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2024-08-30 DOI: 10.1186/s12943-024-02090-z
Zhenqiang Sun, Yanxin Xu, Chaohua Si, Xiaoke Wu, Yaxin Guo, Chen Chen, Chengzeng Wang
Plenty of circRNAs have been reported to play an important role in colorectal cancer (CRC), while the reason of abnormal circRNA expression in cancer still keep elusive. Here, we found that m7G RNA modifications were enriched in some circRNAs, these m7G modifications in circRNAs were catalyzed by METTL1, and the GG motif was the main site preference for m7G modifications in circRNAs. We further confirmed that METTL1 played a cancer-promoting role in CRC. We then screened a highly expressed circRNA, called circKDM1A, and found that METTL1 prevented the degradation of circKDM1A by m7G modification. CircKDM1A was further verified to promote proliferation, invasion and migration of CRC in vivo and in vitro. Its cancer-promoting ability was weakened after the m7G site mutation. CircKDM1A was verified to activate AKT pathway by upregulating PDK1, consequently promoting CRC progression. These results suggest that m7G-modified circRNA promotes CRC progression via activating AKT pathway. Our study uncovers an essential physiological function and mechanism of METTL1-mediated m7G modification in the regulation of circRNA stability and cancer progression.
有报道称,大量的circRNA在结直肠癌(CRC)中发挥着重要作用,而癌症中circRNA异常表达的原因却仍然扑朔迷离。在这里,我们发现m7G RNA修饰在一些circRNA中富集,这些circRNA中的m7G修饰由METTL1催化,而GG基序是circRNA中m7G修饰的主要偏好位点。我们进一步证实了 METTL1 在 CRC 中的促癌作用。随后,我们筛选了一种高表达的 circRNA(名为 circKDM1A),发现 METTL1 通过 m7G 修饰阻止了 circKDM1A 的降解。经进一步验证,circKDM1A能促进体内和体外CRC的增殖、侵袭和迁移。m7G位点突变后,其促癌能力减弱。经证实,CircKDM1A 可通过上调 PDK1 激活 AKT 通路,从而促进 CRC 的进展。这些结果表明,m7G修饰的circRNA通过激活AKT通路促进CRC的进展。我们的研究揭示了 METTL1 介导的 m7G 修饰在调控 circRNA 稳定性和癌症进展中的重要生理功能和机制。
{"title":"Targeting m7G-enriched circKDM1A prevents colorectal cancer progression","authors":"Zhenqiang Sun, Yanxin Xu, Chaohua Si, Xiaoke Wu, Yaxin Guo, Chen Chen, Chengzeng Wang","doi":"10.1186/s12943-024-02090-z","DOIUrl":"https://doi.org/10.1186/s12943-024-02090-z","url":null,"abstract":"Plenty of circRNAs have been reported to play an important role in colorectal cancer (CRC), while the reason of abnormal circRNA expression in cancer still keep elusive. Here, we found that m7G RNA modifications were enriched in some circRNAs, these m7G modifications in circRNAs were catalyzed by METTL1, and the GG motif was the main site preference for m7G modifications in circRNAs. We further confirmed that METTL1 played a cancer-promoting role in CRC. We then screened a highly expressed circRNA, called circKDM1A, and found that METTL1 prevented the degradation of circKDM1A by m7G modification. CircKDM1A was further verified to promote proliferation, invasion and migration of CRC in vivo and in vitro. Its cancer-promoting ability was weakened after the m7G site mutation. CircKDM1A was verified to activate AKT pathway by upregulating PDK1, consequently promoting CRC progression. These results suggest that m7G-modified circRNA promotes CRC progression via activating AKT pathway. Our study uncovers an essential physiological function and mechanism of METTL1-mediated m7G modification in the regulation of circRNA stability and cancer progression.","PeriodicalId":19000,"journal":{"name":"Molecular Cancer","volume":null,"pages":null},"PeriodicalIF":37.3,"publicationDate":"2024-08-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142100697","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
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