首页 > 最新文献

Experimental Hematology & Oncology最新文献

英文 中文
GPR68-ATF4 signaling is a novel prosurvival pathway in glioblastoma activated by acidic extracellular microenvironment. GPR68-ATF4信号传导是胶质母细胞瘤中由酸性细胞外微环境激活的新型促生存途径。
IF 10.9 1区 医学 Q1 Medicine Pub Date : 2024-01-31 DOI: 10.1186/s40164-023-00468-1
Charles H Williams, Leif R Neitzel, Jessica Cornell, Samantha Rea, Ian Mills, Maya S Silver, Jovanni D Ahmad, Konstantin G Birukov, Anna Birukova, Henry Brem, Betty Tyler, Eli E Bar, Charles C Hong

Background: Glioblastoma multiforme (GBM) stands as a formidable challenge in oncology because of its aggressive nature and severely limited treatment options. Despite decades of research, the survival rates for GBM remain effectively stagnant. A defining hallmark of GBM is a highly acidic tumor microenvironment, which is thought to activate pro-tumorigenic pathways. This acidification is the result of altered tumor metabolism favoring aerobic glycolysis, a phenomenon known as the Warburg effect. Low extracellular pH confers radioresistant tumors to glial cells. Notably GPR68, an acid sensing GPCR, is upregulated in radioresistant GBM. Usage of Lorazepam, which has off target agonism of GPR68, is linked to worse clinical outcomes for a variety of cancers. However, the role of tumor microenvironment acidification in GPR68 activation has not been assessed in cancer. Here we interrogate the role of GPR68 specifically in GBM cells using a novel highly specific small molecule inhibitor of GPR68 named Ogremorphin (OGM) to induce the iron mediated cell death pathway: ferroptosis.

Method: OGM was identified in a non-biased zebrafish embryonic development screen and validated with Morpholino and CRISPR based approaches. Next, A GPI-anchored pH reporter, pHluorin2, was stably expressed in U87 glioblastoma cells to probe extracellular acidification. Cell survival assays, via nuclei counting and cell titer glo, were used to demonstrate sensitivity to GPR68 inhibition in twelve immortalized and PDX GBM lines. To determine GPR68 inhibition's mechanism of cell death we use DAVID pathway analysis of RNAseq. Our major indication, ferroptosis, was then confirmed by western blotting and qRT-PCR of reporter genes including TFRC. This finding was further validated by transmission electron microscopy and liperfluo staining to assess lipid peroxidation. Lastly, we use siRNA and CRISPRi to demonstrate the critical role of ATF4 suppression via GPR68 for GBM survival.

Results: We used a pHLourin2 probe to demonstrate how glioblastoma cells acidify their microenvironment to activate the commonly over expressed acid sensing GPCR, GPR68. Using our small molecule inhibitor OGM and genetic means, we show that blocking GPR68 signaling results in robust cell death in all thirteen glioblastoma cell lines tested, irrespective of genetic and phenotypic heterogeneity, or resistance to the mainstay GBM chemotherapeutic temozolomide. We use U87 and U138 glioblastoma cell lines to show how selective induction of ferroptosis occurs in an ATF4-dependent manner. Importantly, OGM was not-acutely toxic to zebrafish and its inhibitory effects were found to spare non-malignant neural cells.

Conclusion: These results indicate GPR68 emerges as a critical sensor for an autocrine pro-tumorigenic signaling cascade triggered by extracellular acidification in glioblastoma cells. In this context, GPR68 suppresses

背景:多形性胶质母细胞瘤(GBM)因其侵袭性强和治疗方案严重受限而成为肿瘤学领域的一项严峻挑战。尽管经过数十年的研究,GBM 的存活率实际上仍然停滞不前。GBM 的一个显著特点是肿瘤微环境呈高度酸性,这被认为会激活肿瘤致病通路。这种酸化是肿瘤新陈代谢改变的结果,有利于有氧糖酵解,这种现象被称为沃伯格效应。细胞外 pH 值低使神经胶质细胞具有抗放射肿瘤性。值得注意的是,GPR68(一种酸感应 GPCR)在耐放射性 GBM 中上调。劳拉西泮对 GPR68 有脱靶激动作用,它的使用与多种癌症的临床预后恶化有关。然而,肿瘤微环境酸化在 GPR68 激活中的作用尚未在癌症中得到评估。在此,我们使用一种名为 Ogremorphin (OGM) 的新型高特异性 GPR68 小分子抑制剂来诱导铁介导的细胞死亡途径:铁凋亡,从而研究 GPR68 在 GBM 细胞中的作用:方法:OGM 是在无偏倚斑马鱼胚胎发育筛选中发现的,并通过基于 Morpholino 和 CRISPR 的方法进行了验证。接下来,在 U87 胶质母细胞瘤细胞中稳定表达了 GPI-anchored pH 报告物 pHluorin2,以探测细胞外酸化。通过细胞核计数和细胞滴度球进行细胞存活测定,以证明十二种永生化和 PDX GBM 株系对 GPR68 抑制的敏感性。为了确定 GPR68 抑制的细胞死亡机制,我们使用了 RNAseq 的 DAVID 通路分析。我们的主要指标--铁突变,随后通过包括 TFRC 在内的报告基因的 Western 印迹和 qRT-PCR 得到了证实。透射电子显微镜和脂质过氧化物染色评估进一步验证了这一发现。最后,我们利用 siRNA 和 CRISPRi 证明了 ATF4 通过 GPR68 对 GBM 生存的抑制作用:我们使用 pHLourin2 探针证明了胶质母细胞瘤细胞如何酸化其微环境以激活普遍过度表达的酸感应 GPCR--GPR68。利用我们的小分子抑制剂 OGM 和基因手段,我们表明阻断 GPR68 信号传导会导致所有 13 个接受测试的胶质母细胞瘤细胞系的细胞大量死亡,而与基因和表型异质性无关,也与对主要的胶质母细胞瘤化疗药物替莫唑胺的耐药性无关。我们使用 U87 和 U138 胶质母细胞瘤细胞系来展示如何以 ATF4 依赖性方式选择性诱导铁变态反应。重要的是,OGM 对斑马鱼无急性毒性,其抑制作用还能使非恶性神经细胞幸免于难:这些结果表明,GPR68 是胶质母细胞瘤细胞由细胞外酸化引发的自分泌促肿瘤信号级联的关键传感器。在这种情况下,GPR68 会抑制 ATF4,抑制 GPR68 会增加 ATF4 的表达,从而导致细胞铁性死亡。这些发现为有选择性地诱导胶质母细胞瘤细胞的铁变态反应,同时保护健康的神经组织提供了一种很有前景的治疗方法。
{"title":"GPR68-ATF4 signaling is a novel prosurvival pathway in glioblastoma activated by acidic extracellular microenvironment.","authors":"Charles H Williams, Leif R Neitzel, Jessica Cornell, Samantha Rea, Ian Mills, Maya S Silver, Jovanni D Ahmad, Konstantin G Birukov, Anna Birukova, Henry Brem, Betty Tyler, Eli E Bar, Charles C Hong","doi":"10.1186/s40164-023-00468-1","DOIUrl":"10.1186/s40164-023-00468-1","url":null,"abstract":"<p><strong>Background: </strong>Glioblastoma multiforme (GBM) stands as a formidable challenge in oncology because of its aggressive nature and severely limited treatment options. Despite decades of research, the survival rates for GBM remain effectively stagnant. A defining hallmark of GBM is a highly acidic tumor microenvironment, which is thought to activate pro-tumorigenic pathways. This acidification is the result of altered tumor metabolism favoring aerobic glycolysis, a phenomenon known as the Warburg effect. Low extracellular pH confers radioresistant tumors to glial cells. Notably GPR68, an acid sensing GPCR, is upregulated in radioresistant GBM. Usage of Lorazepam, which has off target agonism of GPR68, is linked to worse clinical outcomes for a variety of cancers. However, the role of tumor microenvironment acidification in GPR68 activation has not been assessed in cancer. Here we interrogate the role of GPR68 specifically in GBM cells using a novel highly specific small molecule inhibitor of GPR68 named Ogremorphin (OGM) to induce the iron mediated cell death pathway: ferroptosis.</p><p><strong>Method: </strong>OGM was identified in a non-biased zebrafish embryonic development screen and validated with Morpholino and CRISPR based approaches. Next, A GPI-anchored pH reporter, pHluorin2, was stably expressed in U87 glioblastoma cells to probe extracellular acidification. Cell survival assays, via nuclei counting and cell titer glo, were used to demonstrate sensitivity to GPR68 inhibition in twelve immortalized and PDX GBM lines. To determine GPR68 inhibition's mechanism of cell death we use DAVID pathway analysis of RNAseq. Our major indication, ferroptosis, was then confirmed by western blotting and qRT-PCR of reporter genes including TFRC. This finding was further validated by transmission electron microscopy and liperfluo staining to assess lipid peroxidation. Lastly, we use siRNA and CRISPRi to demonstrate the critical role of ATF4 suppression via GPR68 for GBM survival.</p><p><strong>Results: </strong>We used a pHLourin2 probe to demonstrate how glioblastoma cells acidify their microenvironment to activate the commonly over expressed acid sensing GPCR, GPR68. Using our small molecule inhibitor OGM and genetic means, we show that blocking GPR68 signaling results in robust cell death in all thirteen glioblastoma cell lines tested, irrespective of genetic and phenotypic heterogeneity, or resistance to the mainstay GBM chemotherapeutic temozolomide. We use U87 and U138 glioblastoma cell lines to show how selective induction of ferroptosis occurs in an ATF4-dependent manner. Importantly, OGM was not-acutely toxic to zebrafish and its inhibitory effects were found to spare non-malignant neural cells.</p><p><strong>Conclusion: </strong>These results indicate GPR68 emerges as a critical sensor for an autocrine pro-tumorigenic signaling cascade triggered by extracellular acidification in glioblastoma cells. In this context, GPR68 suppresses","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":10.9,"publicationDate":"2024-01-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10829393/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139641950","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
Decoding leukemia at the single-cell level: clonal architecture, classification, microenvironment, and drug resistance 单细胞水平的白血病解码:克隆结构、分类、微环境和耐药性
IF 10.9 1区 医学 Q1 Medicine Pub Date : 2024-01-30 DOI: 10.1186/s40164-024-00479-6
Jianche Liu, Penglei Jiang, Zezhen Lu, Zebin Yu, Pengxu Qian
Leukemias are refractory hematological malignancies, characterized by marked intrinsic heterogeneity which poses significant obstacles to effective treatment. However, traditional bulk sequencing techniques have not been able to effectively unravel the heterogeneity among individual tumor cells. With the emergence of single-cell sequencing technology, it has bestowed upon us an unprecedented resolution to comprehend the mechanisms underlying leukemogenesis and drug resistance across various levels, including the genome, epigenome, transcriptome and proteome. Here, we provide an overview of the currently prevalent single-cell sequencing technologies and a detailed summary of single-cell studies conducted on leukemia, with a specific focus on four key aspects: (1) leukemia’s clonal architecture, (2) frameworks to determine leukemia subtypes, (3) tumor microenvironment (TME) and (4) the drug-resistant mechanisms of leukemia. This review provides a comprehensive summary of current single-cell studies on leukemia and highlights the markers and mechanisms that show promising clinical implications for the diagnosis and treatment of leukemia.
白血病是难治性血液恶性肿瘤,具有明显的内在异质性,给有效治疗带来了巨大障碍。然而,传统的批量测序技术无法有效揭示单个肿瘤细胞之间的异质性。随着单细胞测序技术的出现,它为我们提供了前所未有的分辨率,使我们能够从基因组、表观基因组、转录组和蛋白质组等不同层面理解白血病发生和耐药的机制。在此,我们概述了目前流行的单细胞测序技术,并详细总结了针对白血病开展的单细胞研究,重点关注四个关键方面:(1) 白血病的克隆结构,(2) 确定白血病亚型的框架,(3) 肿瘤微环境 (TME) 和 (4) 白血病的耐药机制。本综述全面总结了目前有关白血病的单细胞研究,并重点介绍了对白血病诊断和治疗具有临床意义的标志物和机制。
{"title":"Decoding leukemia at the single-cell level: clonal architecture, classification, microenvironment, and drug resistance","authors":"Jianche Liu, Penglei Jiang, Zezhen Lu, Zebin Yu, Pengxu Qian","doi":"10.1186/s40164-024-00479-6","DOIUrl":"https://doi.org/10.1186/s40164-024-00479-6","url":null,"abstract":"Leukemias are refractory hematological malignancies, characterized by marked intrinsic heterogeneity which poses significant obstacles to effective treatment. However, traditional bulk sequencing techniques have not been able to effectively unravel the heterogeneity among individual tumor cells. With the emergence of single-cell sequencing technology, it has bestowed upon us an unprecedented resolution to comprehend the mechanisms underlying leukemogenesis and drug resistance across various levels, including the genome, epigenome, transcriptome and proteome. Here, we provide an overview of the currently prevalent single-cell sequencing technologies and a detailed summary of single-cell studies conducted on leukemia, with a specific focus on four key aspects: (1) leukemia’s clonal architecture, (2) frameworks to determine leukemia subtypes, (3) tumor microenvironment (TME) and (4) the drug-resistant mechanisms of leukemia. This review provides a comprehensive summary of current single-cell studies on leukemia and highlights the markers and mechanisms that show promising clinical implications for the diagnosis and treatment of leukemia.","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":10.9,"publicationDate":"2024-01-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139578369","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 focal adhesion kinase boosts immune response in KRAS/LKB1 co-mutated lung adenocarcinoma via remodeling the tumor microenvironment 靶向局灶粘附激酶可通过重塑肿瘤微环境促进 KRAS/LKB1 共突变肺腺癌的免疫反应
IF 10.9 1区 医学 Q1 Medicine Pub Date : 2024-01-30 DOI: 10.1186/s40164-023-00471-6
Meng Qiao, Fei Zhou, Xinyu Liu, Tao Jiang, Haowei Wang, Xuefei Li, Chao Zhao, Lei Cheng, Xiaoxia Chen, Shengxiang Ren, Zaiqi Wang, Caicun Zhou
KRAS mutation is one of the most common oncogenic drivers in NSCLC, however, the response to immunotherapy is heterogeneous owing to the distinct co-occurring genomic alterations. KRAS/LKB1 co-mutated lung adenocarcinoma displays poor response to PD-1 blockade whereas the mechanism remains undetermined. We explored the specific characteristics of tumor microenvironment (TME) in KL tumors using syngeneic KRASG12DLKB1−/− (KL) and KRASG12DTP53−/− (KP) lung cancer mouse models. The impact of focal adhesion kinase (FAK) inhibitor on KL lung tumors was investigated in vitro and in vivo through evaluation of both KL cell lines and KL lung cancer mouse models. We identified KL tumors as “immune-cold” tumors with excessive extracellular matrix (ECM) collagen deposition that formed a physical barrier to block the infiltration of CD8+T cells. Mechanistically, abundant activated cancer-associated fibroblasts (CAFs) resulted from FAK activation contributed to the formation of the unique TME of KL tumors. FAK inhibition with a small molecular inhibitor could remodel the TME by inhibiting CAFs activation, decreasing collagen deposition and further facilitating the infiltration of anti-tumor immune cells, including CD8+ T cells, DC cells and M1-like macrophages into tumors, hence, converting “immune-cold” KL tumors into “immune-hot” tumors. The combined FAK inhibitor and PD-1 blockade therapy synergistically retarded primary and metastatic tumor growth of KL tumors. Our study identified FAK as a promising intervention target for KL tumors and provided basis for the combination of FAK inhibitor with PD-1 blockade in the management of KL lung cancers.
KRAS突变是NSCLC最常见的致癌因素之一,然而,由于共存的基因组改变不同,免疫疗法的反应也不尽相同。KRAS/LKB1共同突变的肺腺癌对PD-1阻断剂的反应较差,但其机制仍未确定。我们利用KRASG12DLKB1-/-(KL)和KRASG12DTP53-/-(KP)肺癌小鼠模型,探索了KL肿瘤微环境(TME)的具体特征。通过对 KL 细胞系和 KL 肺癌小鼠模型进行评估,研究了局灶粘附激酶(FAK)抑制剂在体外和体内对 KL 肺肿瘤的影响。我们发现 KL 肿瘤是一种 "免疫冷 "肿瘤,其细胞外基质(ECM)胶原沉积过多,形成了阻挡 CD8+T 细胞浸润的物理屏障。从机理上讲,FAK 激活导致的大量活化癌相关成纤维细胞(CAFs)促成了 KL 肿瘤独特的 TME 的形成。使用小分子抑制剂抑制FAK可通过抑制CAFs活化重塑TME,减少胶原沉积,进一步促进抗肿瘤免疫细胞(包括CD8+ T细胞、DC细胞和M1样巨噬细胞)浸润肿瘤,从而将 "免疫冷 "的KL肿瘤转化为 "免疫热 "的肿瘤。FAK抑制剂和PD-1阻断剂联合治疗可协同延缓KL肿瘤的原发性和转移性肿瘤生长。我们的研究发现,FAK是一种很有前景的KL肿瘤干预靶点,并为FAK抑制剂与PD-1阻断剂联合治疗KL肺癌提供了依据。
{"title":"Targeting focal adhesion kinase boosts immune response in KRAS/LKB1 co-mutated lung adenocarcinoma via remodeling the tumor microenvironment","authors":"Meng Qiao, Fei Zhou, Xinyu Liu, Tao Jiang, Haowei Wang, Xuefei Li, Chao Zhao, Lei Cheng, Xiaoxia Chen, Shengxiang Ren, Zaiqi Wang, Caicun Zhou","doi":"10.1186/s40164-023-00471-6","DOIUrl":"https://doi.org/10.1186/s40164-023-00471-6","url":null,"abstract":"KRAS mutation is one of the most common oncogenic drivers in NSCLC, however, the response to immunotherapy is heterogeneous owing to the distinct co-occurring genomic alterations. KRAS/LKB1 co-mutated lung adenocarcinoma displays poor response to PD-1 blockade whereas the mechanism remains undetermined. We explored the specific characteristics of tumor microenvironment (TME) in KL tumors using syngeneic KRASG12DLKB1−/− (KL) and KRASG12DTP53−/− (KP) lung cancer mouse models. The impact of focal adhesion kinase (FAK) inhibitor on KL lung tumors was investigated in vitro and in vivo through evaluation of both KL cell lines and KL lung cancer mouse models. We identified KL tumors as “immune-cold” tumors with excessive extracellular matrix (ECM) collagen deposition that formed a physical barrier to block the infiltration of CD8+T cells. Mechanistically, abundant activated cancer-associated fibroblasts (CAFs) resulted from FAK activation contributed to the formation of the unique TME of KL tumors. FAK inhibition with a small molecular inhibitor could remodel the TME by inhibiting CAFs activation, decreasing collagen deposition and further facilitating the infiltration of anti-tumor immune cells, including CD8+ T cells, DC cells and M1-like macrophages into tumors, hence, converting “immune-cold” KL tumors into “immune-hot” tumors. The combined FAK inhibitor and PD-1 blockade therapy synergistically retarded primary and metastatic tumor growth of KL tumors. Our study identified FAK as a promising intervention target for KL tumors and provided basis for the combination of FAK inhibitor with PD-1 blockade in the management of KL lung cancers.","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":10.9,"publicationDate":"2024-01-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139578250","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
Cancer metabolism and carcinogenesis. 癌症新陈代谢和致癌。
IF 10.9 1区 医学 Q1 Medicine Pub Date : 2024-01-29 DOI: 10.1186/s40164-024-00482-x
Jianqiang Yang, Chloe Shay, Nabil F Saba, Yong Teng

Metabolic reprogramming is an emerging hallmark of cancer cells, enabling them to meet increased nutrient and energy demands while withstanding the challenging microenvironment. Cancer cells can switch their metabolic pathways, allowing them to adapt to different microenvironments and therapeutic interventions. This refers to metabolic heterogeneity, in which different cell populations use different metabolic pathways to sustain their survival and proliferation and impact their response to conventional cancer therapies. Thus, targeting cancer metabolic heterogeneity represents an innovative therapeutic avenue with the potential to overcome treatment resistance and improve therapeutic outcomes. This review discusses the metabolic patterns of different cancer cell populations and developmental stages, summarizes the molecular mechanisms involved in the intricate interactions within cancer metabolism, and highlights the clinical potential of targeting metabolic vulnerabilities as a promising therapeutic regimen. We aim to unravel the complex of metabolic characteristics and develop personalized treatment approaches to address distinct metabolic traits, ultimately enhancing patient outcomes.

代谢重编程是癌细胞的一个新特征,它使癌细胞能够满足更多的营养和能量需求,同时抵御微环境的挑战。癌细胞可以改变其代谢途径,从而适应不同的微环境和治疗干预。这就是代谢异质性,不同的细胞群使用不同的代谢途径来维持生存和增殖,并影响它们对传统癌症疗法的反应。因此,以癌症代谢异质性为靶点是一种创新的治疗途径,有可能克服耐药性并改善治疗效果。本综述讨论了不同癌细胞群和发育阶段的代谢模式,总结了癌症代谢中错综复杂的相互作用所涉及的分子机制,并强调了靶向代谢脆弱性作为一种有前途的治疗方案的临床潜力。我们的目标是揭示复杂的代谢特征,并针对不同的代谢特征开发个性化治疗方法,最终提高患者的治疗效果。
{"title":"Cancer metabolism and carcinogenesis.","authors":"Jianqiang Yang, Chloe Shay, Nabil F Saba, Yong Teng","doi":"10.1186/s40164-024-00482-x","DOIUrl":"10.1186/s40164-024-00482-x","url":null,"abstract":"<p><p>Metabolic reprogramming is an emerging hallmark of cancer cells, enabling them to meet increased nutrient and energy demands while withstanding the challenging microenvironment. Cancer cells can switch their metabolic pathways, allowing them to adapt to different microenvironments and therapeutic interventions. This refers to metabolic heterogeneity, in which different cell populations use different metabolic pathways to sustain their survival and proliferation and impact their response to conventional cancer therapies. Thus, targeting cancer metabolic heterogeneity represents an innovative therapeutic avenue with the potential to overcome treatment resistance and improve therapeutic outcomes. This review discusses the metabolic patterns of different cancer cell populations and developmental stages, summarizes the molecular mechanisms involved in the intricate interactions within cancer metabolism, and highlights the clinical potential of targeting metabolic vulnerabilities as a promising therapeutic regimen. We aim to unravel the complex of metabolic characteristics and develop personalized treatment approaches to address distinct metabolic traits, ultimately enhancing patient outcomes.</p>","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":10.9,"publicationDate":"2024-01-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10826200/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139575032","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
A novel AML1-ETO/FTO positive feedback loop promotes leukemogenesis and Ara-C resistance via stabilizing IGFBP2 in t(8;21) acute myeloid leukemia. 新型 AML1-ETO/FTO 正反馈环路通过稳定 t(8;21) 急性髓性白血病中的 IGFBP2 促进白血病生成和 Ara-C 抗性。
IF 10.9 1区 医学 Q1 Medicine Pub Date : 2024-01-24 DOI: 10.1186/s40164-024-00480-z
Wei Zhou, Siying Li, Hong Wang, Jingfeng Zhou, Shuyi Li, Guofeng Chen, Wei Guan, Xianli Fu, Clara Nervi, Li Yu, Yonghui Li

Background: t(8;21)(q22;q22) is one of the most frequent chromosomal abnormalities in acute myeloid leukemia (AML), leading to the generation of the fusion protein AML1-ETO. Despite t(8;21) AML being considered as a subtype with a favorable prognosis, approximately 30-50% of patients experience drug resistance and subsequent relapse. N6-methyladenosine (m6A) is demonstrated to be involved in the development of AML. However, the regulatory mechanisms between AML1-ETO and m6A-related enzymes and the roles of dysregulated m6A modifications in the t(8;21)-leukemogenesis and chemoresistance remain elusive.

Methods: Chromatin immunoprecipitation, dual-luciferase reporter assay, m6A-qPCR, RNA immunoprecipitation, and RNA stability assay were used to investigate a regulatory loop between AML1-ETO and FTO, an m6A demethylase. Gain- and loss-of-function experiments both in vitro and in vivo were further performed. Transcriptome-wide RNA sequencing and m6A sequencing were conducted to identify the potential targets of FTO.

Results: Here we show that FTO is highly expressed in t(8;21) AML, especially in patients with primary refractory disease. The expression of FTO is positively correlated with AML1-ETO, which is attributed to a positive regulatory loop between the AML1-ETO and FTO. Mechanistically, AML1-ETO upregulates FTO expression through inhibiting the transcriptional repression of FTO mediated by PU.1. Meanwhile, FTO promotes the expression of AML1-ETO by inhibiting YTHDF2-mediated AML1-ETO mRNA decay. Inactivation of FTO significantly suppresses cell proliferation, promotes cell differentiation and renders resistant t(8;21) AML cells sensitive to Ara-C. FTO exerts functions by regulating its mRNA targets, especially IGFBP2, in an m6A-dependent manner. Regain of Ara-C tolerance is observed when IGFBP2 is overexpressed in FTO-knockdown t(8;21) AML cells.

Conclusion: Our work reveals a therapeutic potential of targeting AML1-ETO/FTO/IGFBP2 minicircuitry in the treatment for t(8;21) patients with resistance to Ara-C.

背景:t(8;21)(q22;q22)是急性髓性白血病(AML)中最常见的染色体异常之一,可导致融合蛋白AML1-ETO的产生。尽管t(8;21)急性髓细胞白血病被认为是预后良好的亚型,但约有30%-50%的患者会出现耐药性并随后复发。N6-甲基腺苷(m6A)被证实参与了急性髓细胞性白血病的发病。然而,AML1-ETO与m6A相关酶之间的调控机制,以及失调的m6A修饰在t(8;21)-白血病发生和化疗耐药性中的作用仍未确定:方法:采用染色质免疫沉淀、双荧光素酶报告实验、m6A-qPCR、RNA免疫沉淀和RNA稳定性实验研究AML1-ETO和m6A去甲基化酶FTO之间的调控环路。实验还进一步进行了体外和体内功能增益和丧失实验。我们还进行了全转录组 RNA 测序和 m6A 测序,以确定 FTO 的潜在靶标:结果:我们在此发现,FTO在t(8;21)型急性髓细胞白血病中高表达,尤其是在原发性难治性疾病患者中。FTO的表达与AML1-ETO呈正相关,这归因于AML1-ETO与FTO之间的正向调节环。从机理上讲,AML1-ETO 通过抑制 PU.1 介导的 FTO 转录抑制而上调 FTO 的表达。同时,FTO通过抑制YTHDF2介导的AML1-ETO mRNA衰变来促进AML1-ETO的表达。FTO 失活能显著抑制细胞增殖,促进细胞分化,并使耐药的 t(8;21) AML 细胞对 Ara-C 敏感。FTO 通过调节其 mRNA 靶标,尤其是 IGFBP2,以 m6A 依赖性方式发挥功能。当IGFBP2在FTO敲除的t(8;21) AML细胞中过表达时,可观察到Ara-C耐受性的恢复:我们的研究揭示了靶向AML1-ETO/FTO/IGFBP2小环路治疗对Ara-C耐药的t(8;21)患者的潜力。
{"title":"A novel AML1-ETO/FTO positive feedback loop promotes leukemogenesis and Ara-C resistance via stabilizing IGFBP2 in t(8;21) acute myeloid leukemia.","authors":"Wei Zhou, Siying Li, Hong Wang, Jingfeng Zhou, Shuyi Li, Guofeng Chen, Wei Guan, Xianli Fu, Clara Nervi, Li Yu, Yonghui Li","doi":"10.1186/s40164-024-00480-z","DOIUrl":"10.1186/s40164-024-00480-z","url":null,"abstract":"<p><strong>Background: </strong>t(8;21)(q22;q22) is one of the most frequent chromosomal abnormalities in acute myeloid leukemia (AML), leading to the generation of the fusion protein AML1-ETO. Despite t(8;21) AML being considered as a subtype with a favorable prognosis, approximately 30-50% of patients experience drug resistance and subsequent relapse. N<sup>6</sup>-methyladenosine (m<sup>6</sup>A) is demonstrated to be involved in the development of AML. However, the regulatory mechanisms between AML1-ETO and m<sup>6</sup>A-related enzymes and the roles of dysregulated m<sup>6</sup>A modifications in the t(8;21)-leukemogenesis and chemoresistance remain elusive.</p><p><strong>Methods: </strong>Chromatin immunoprecipitation, dual-luciferase reporter assay, m<sup>6</sup>A-qPCR, RNA immunoprecipitation, and RNA stability assay were used to investigate a regulatory loop between AML1-ETO and FTO, an m<sup>6</sup>A demethylase. Gain- and loss-of-function experiments both in vitro and in vivo were further performed. Transcriptome-wide RNA sequencing and m<sup>6</sup>A sequencing were conducted to identify the potential targets of FTO.</p><p><strong>Results: </strong>Here we show that FTO is highly expressed in t(8;21) AML, especially in patients with primary refractory disease. The expression of FTO is positively correlated with AML1-ETO, which is attributed to a positive regulatory loop between the AML1-ETO and FTO. Mechanistically, AML1-ETO upregulates FTO expression through inhibiting the transcriptional repression of FTO mediated by PU.1. Meanwhile, FTO promotes the expression of AML1-ETO by inhibiting YTHDF2-mediated AML1-ETO mRNA decay. Inactivation of FTO significantly suppresses cell proliferation, promotes cell differentiation and renders resistant t(8;21) AML cells sensitive to Ara-C. FTO exerts functions by regulating its mRNA targets, especially IGFBP2, in an m<sup>6</sup>A-dependent manner. Regain of Ara-C tolerance is observed when IGFBP2 is overexpressed in FTO-knockdown t(8;21) AML cells.</p><p><strong>Conclusion: </strong>Our work reveals a therapeutic potential of targeting AML1-ETO/FTO/IGFBP2 minicircuitry in the treatment for t(8;21) patients with resistance to Ara-C.</p>","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":10.9,"publicationDate":"2024-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10807068/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139546037","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
METTL1 mediated tRNA m7G modification promotes leukaemogenesis of AML via tRNA regulated translational control. METTL1 介导的 tRNA m7G 修饰通过 tRNA 调控的翻译控制促进 AML 的白血病生成。
IF 10.9 1区 医学 Q1 Medicine Pub Date : 2024-01-24 DOI: 10.1186/s40164-024-00477-8
Pan Zhao, Lin Xia, Dan Chen, Wei Xu, Huanping Guo, Yinying Xu, Bingbing Yan, Xiao Wu, Yuxia Li, Yunfang Zhang, Xi Zhang

Background: RNA modifications have been proven to play fundamental roles in regulating cellular biology process. Recently, maladjusted N7-methylguanosine (m7G) modification and its modifiers METTL1/WDR4 have been confirmed an oncogene role in multiple cancers. However, the functions and molecular mechanisms of METTL1/WDR4 in acute myeloid leukemia (AML) remain to be determined.

Methods: METTL1/WDR4 expression levels were quantified using qRT-PCR, western blot analysis on AML clinical samples, and bioinformatics analysis on publicly available AML datasets. CCK-8 assays and cell count assays were performed to determine cell proliferation. Flow cytometry assays were conducted to assess cell cycle and apoptosis rates. Multiple techniques were used for mechanism studies in vitro assays, such as northern blotting, liquid chromatography-coupled mass spectrometry (LC-MS/MS), tRNA stability analysis, transcriptome sequencing, small non-coding RNA sequencing, quantitative proteomics, and protein synthesis measurements.

Results: METTL1/WDR4 are significantly elevated in AML patients and associated with poor prognosis. METTL1 knockdown resulted in reduced cell proliferation and increased apoptosis in AML cells. Mechanically, METTL1 knockdown leads to significant decrease of m7G modification abundance on tRNA, which further destabilizes tRNAs and facilitates the biogenesis of tsRNAs in AML cells. In addition, profiling of nascent proteins revealed that METTL1 knockdown and transfection of total tRNAs that were isolated from METTL1 knockdown AML cells decreased global translation efficiency in AML cells.

Conclusions: Taken together, our study demonstrates the important role of METTL1/WDR4 in AML leukaemogenesis, which provides a promising target candidate for AML therapy.

背景:RNA 修饰已被证实在调节细胞生物学过程中发挥着重要作用。最近,N7-甲基鸟苷(m7G)修饰及其修饰因子METTL1/WDR4被证实在多种癌症中起着癌基因的作用。然而,METTL1/WDR4在急性髓性白血病(AML)中的功能和分子机制仍有待确定:方法:使用 qRT-PCR、Western 印迹分析对急性髓性白血病临床样本的 METTL1/WDR4 表达水平进行量化,并对公开的急性髓性白血病数据集进行生物信息学分析。通过 CCK-8 检测和细胞计数检测确定细胞增殖情况。流式细胞仪检测用于评估细胞周期和凋亡率。体外检测中使用了多种技术进行机理研究,如北印迹法、液相色谱耦合质谱法(LC-MS/MS)、tRNA稳定性分析、转录组测序、小非编码RNA测序、定量蛋白质组学和蛋白质合成测量:结果:METTL1/WDR4在急性髓细胞性白血病患者中明显升高,并与预后不良有关。敲除 METTL1 会导致 AML 细胞增殖减少、凋亡增加。从机理上讲,METTL1 基因敲除会导致 tRNA 上的 m7G 修饰丰度显著下降,从而进一步破坏 tRNA 的稳定性,促进 AML 细胞中 tsRNA 的生物生成。此外,新生蛋白质的分析表明,METTL1敲除和转染从METTL1敲除的AML细胞中分离出的总tRNA会降低AML细胞的全局翻译效率:综上所述,我们的研究证明了METTL1/WDR4在AML白血病发生过程中的重要作用,为AML治疗提供了一个前景广阔的候选靶点。
{"title":"METTL1 mediated tRNA m<sup>7</sup>G modification promotes leukaemogenesis of AML via tRNA regulated translational control.","authors":"Pan Zhao, Lin Xia, Dan Chen, Wei Xu, Huanping Guo, Yinying Xu, Bingbing Yan, Xiao Wu, Yuxia Li, Yunfang Zhang, Xi Zhang","doi":"10.1186/s40164-024-00477-8","DOIUrl":"10.1186/s40164-024-00477-8","url":null,"abstract":"<p><strong>Background: </strong>RNA modifications have been proven to play fundamental roles in regulating cellular biology process. Recently, maladjusted N7-methylguanosine (m<sup>7</sup>G) modification and its modifiers METTL1/WDR4 have been confirmed an oncogene role in multiple cancers. However, the functions and molecular mechanisms of METTL1/WDR4 in acute myeloid leukemia (AML) remain to be determined.</p><p><strong>Methods: </strong>METTL1/WDR4 expression levels were quantified using qRT-PCR, western blot analysis on AML clinical samples, and bioinformatics analysis on publicly available AML datasets. CCK-8 assays and cell count assays were performed to determine cell proliferation. Flow cytometry assays were conducted to assess cell cycle and apoptosis rates. Multiple techniques were used for mechanism studies in vitro assays, such as northern blotting, liquid chromatography-coupled mass spectrometry (LC-MS/MS), tRNA stability analysis, transcriptome sequencing, small non-coding RNA sequencing, quantitative proteomics, and protein synthesis measurements.</p><p><strong>Results: </strong>METTL1/WDR4 are significantly elevated in AML patients and associated with poor prognosis. METTL1 knockdown resulted in reduced cell proliferation and increased apoptosis in AML cells. Mechanically, METTL1 knockdown leads to significant decrease of m<sup>7</sup>G modification abundance on tRNA, which further destabilizes tRNAs and facilitates the biogenesis of tsRNAs in AML cells. In addition, profiling of nascent proteins revealed that METTL1 knockdown and transfection of total tRNAs that were isolated from METTL1 knockdown AML cells decreased global translation efficiency in AML cells.</p><p><strong>Conclusions: </strong>Taken together, our study demonstrates the important role of METTL1/WDR4 in AML leukaemogenesis, which provides a promising target candidate for AML therapy.</p>","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":10.9,"publicationDate":"2024-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10807064/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139546002","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
Correction: Breast cancer heterogeneity and its implication in personalized precision therapy. 更正:乳腺癌异质性及其对个性化精准治疗的影响。
IF 10.9 1区 医学 Q1 Medicine Pub Date : 2024-01-23 DOI: 10.1186/s40164-024-00472-z
Liantao Guo, Deguang Kong, Jianhua Liu, Ling Zhan, Lan Luo, Weijie Zheng, Qingyuan Zheng, Chuang Chen, Shengrong Sun
{"title":"Correction: Breast cancer heterogeneity and its implication in personalized precision therapy.","authors":"Liantao Guo, Deguang Kong, Jianhua Liu, Ling Zhan, Lan Luo, Weijie Zheng, Qingyuan Zheng, Chuang Chen, Shengrong Sun","doi":"10.1186/s40164-024-00472-z","DOIUrl":"10.1186/s40164-024-00472-z","url":null,"abstract":"","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":10.9,"publicationDate":"2024-01-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10807152/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139541770","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
ZMIZ2 facilitates hepatocellular carcinoma progression via LEF1 mediated activation of Wnt/β-catenin pathway ZMIZ2 通过 LEF1 介导的 Wnt/β-catenin 通路激活促进肝细胞癌进展
IF 10.9 1区 医学 Q1 Medicine Pub Date : 2024-01-22 DOI: 10.1186/s40164-024-00475-w
Yang Ding, Yumei Ning, Hui Kang, Yuan Yuan, Kun Lin, Chun Wang, Yun Yi, Jianghua He, Lurao Li, Xingxing He, Ying Chang
Hepatocellular carcinoma (HCC) is one of the most common malignancies with a high lethality rate. ZMIZ2 is a transcriptional co-activator implicated in various human diseases. However, the role and molecular mechanism of ZMIZ2 in HCC remains to be elucidated. The expression and prognostic value of ZMIZ2 in HCC was excavated from public databases and explored by bioinformatic analysis. Then the expression of ZMIZ2 and related genes was further validated by quantitative RT-PCR, western blotting, and immunohistochemistry. Loss and gain-of-function experiments were performed in vitro and in vivo to investigate the function of ZMIZ2 in HCC. In addition, transcriptome sequencing and immunoprecipitation was conducted to explore the potential molecular mechanisms of ZMIZ2. ZMIZ2 was highly expressed in HCC and associated with poor prognosis. Silencing ZMIZ2 significantly inhibited HCC cell proliferation, cell cycle process, migration, and invasion in vitro, and also inhibited the progression of HCC in vivo. Additionally, ZMIZ2 expression was correlated with immune cell infiltration in HCC samples. Somatic mutation analysis showed that ZMIZ2 and TP53 mutations jointly affected the progression of HCC. Mechanistically, ZMIZ2 interacted with LEF1 to regulate malignant progression of HCC by activating the Wnt/β-catenin pathway. ZMIZ2 was overexpressed in HCC and associated with poor prognosis. The overexpression of ZMIZ2 was corelated with malignant phenotype, and it facilitated HCC progression via LEF1-mediated activation of the Wnt/β-catenin pathway. Furthermore, ZMIZ2 could be served as a prognostic biomarker and a new therapeutic target for HCC.
肝细胞癌(HCC)是最常见的恶性肿瘤之一,致死率很高。ZMIZ2 是一种转录共激活因子,与多种人类疾病有关。然而,ZMIZ2在HCC中的作用和分子机制仍有待阐明。本研究从公共数据库中挖掘了ZMIZ2在HCC中的表达和预后价值,并通过生物信息学分析进行了探讨。然后通过定量 RT-PCR、Western 印迹和免疫组化进一步验证了 ZMIZ2 及相关基因的表达。为了研究ZMIZ2在HCC中的功能,研究人员进行了体外和体内功能缺失和增益实验。此外,还进行了转录组测序和免疫沉淀,以探索ZMIZ2的潜在分子机制。ZMIZ2在HCC中高表达,并与不良预后相关。沉默ZMIZ2可明显抑制HCC细胞的体外增殖、细胞周期进程、迁移和侵袭,也可抑制HCC在体内的进展。此外,ZMIZ2的表达与HCC样本中的免疫细胞浸润相关。体细胞突变分析表明,ZMIZ2和TP53突变会共同影响HCC的进展。从机制上讲,ZMIZ2与LEF1相互作用,通过激活Wnt/β-catenin通路调控HCC的恶性进展。ZMIZ2在HCC中的过表达与预后不良有关。ZMIZ2的过表达与恶性表型相关,它通过LEF1介导的Wnt/β-catenin通路激活促进了HCC的进展。此外,ZMIZ2可作为HCC的预后生物标志物和新的治疗靶点。
{"title":"ZMIZ2 facilitates hepatocellular carcinoma progression via LEF1 mediated activation of Wnt/β-catenin pathway","authors":"Yang Ding, Yumei Ning, Hui Kang, Yuan Yuan, Kun Lin, Chun Wang, Yun Yi, Jianghua He, Lurao Li, Xingxing He, Ying Chang","doi":"10.1186/s40164-024-00475-w","DOIUrl":"https://doi.org/10.1186/s40164-024-00475-w","url":null,"abstract":"Hepatocellular carcinoma (HCC) is one of the most common malignancies with a high lethality rate. ZMIZ2 is a transcriptional co-activator implicated in various human diseases. However, the role and molecular mechanism of ZMIZ2 in HCC remains to be elucidated. The expression and prognostic value of ZMIZ2 in HCC was excavated from public databases and explored by bioinformatic analysis. Then the expression of ZMIZ2 and related genes was further validated by quantitative RT-PCR, western blotting, and immunohistochemistry. Loss and gain-of-function experiments were performed in vitro and in vivo to investigate the function of ZMIZ2 in HCC. In addition, transcriptome sequencing and immunoprecipitation was conducted to explore the potential molecular mechanisms of ZMIZ2. ZMIZ2 was highly expressed in HCC and associated with poor prognosis. Silencing ZMIZ2 significantly inhibited HCC cell proliferation, cell cycle process, migration, and invasion in vitro, and also inhibited the progression of HCC in vivo. Additionally, ZMIZ2 expression was correlated with immune cell infiltration in HCC samples. Somatic mutation analysis showed that ZMIZ2 and TP53 mutations jointly affected the progression of HCC. Mechanistically, ZMIZ2 interacted with LEF1 to regulate malignant progression of HCC by activating the Wnt/β-catenin pathway. ZMIZ2 was overexpressed in HCC and associated with poor prognosis. The overexpression of ZMIZ2 was corelated with malignant phenotype, and it facilitated HCC progression via LEF1-mediated activation of the Wnt/β-catenin pathway. Furthermore, ZMIZ2 could be served as a prognostic biomarker and a new therapeutic target for HCC.","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":10.9,"publicationDate":"2024-01-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139517668","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
Crosstalk between colorectal CSCs and immune cells in tumorigenesis, and strategies for targeting colorectal CSCs 结直肠癌干细胞与免疫细胞在肿瘤发生过程中的相互关系,以及针对结直肠癌干细胞的策略
IF 10.9 1区 医学 Q1 Medicine Pub Date : 2024-01-22 DOI: 10.1186/s40164-024-00474-x
Qi Zhao, Hong Zong, Pingping Zhu, Chang Su, Wenxue Tang, Zhenzhen Chen, Shuiling Jin
Cancer immunotherapy has emerged as a promising strategy in the treatment of colorectal cancer, and relapse after tumor immunotherapy has attracted increasing attention. Cancer stem cells (CSCs), a small subset of tumor cells with self-renewal and differentiation capacities, are resistant to traditional therapies such as radiotherapy and chemotherapy. Recently, CSCs have been proven to be the cells driving tumor relapse after immunotherapy. However, the mutual interactions between CSCs and cancer niche immune cells are largely uncharacterized. In this review, we focus on colorectal CSCs, CSC-immune cell interactions and CSC-based immunotherapy. Colorectal CSCs are characterized by robust expression of surface markers such as CD44, CD133 and Lgr5; hyperactivation of stemness-related signaling pathways, such as the Wnt/β-catenin, Hippo/Yap1, Jak/Stat and Notch pathways; and disordered epigenetic modifications, including DNA methylation, histone modification, chromatin remodeling, and noncoding RNA action. Moreover, colorectal CSCs express abnormal levels of immune-related genes such as MHC and immune checkpoint molecules and mutually interact with cancer niche cells in multiple tumorigenesis-related processes, including tumor initiation, maintenance, metastasis and drug resistance. To date, many therapies targeting CSCs have been evaluated, including monoclonal antibodies, antibody‒drug conjugates, bispecific antibodies, tumor vaccines adoptive cell therapy, and small molecule inhibitors. With the development of CSC-/niche-targeting technology, as well as the integration of multidisciplinary studies, novel therapies that eliminate CSCs and reverse their immunosuppressive microenvironment are expected to be developed for the treatment of solid tumors, including colorectal cancer.
癌症免疫疗法已成为治疗结直肠癌的一种前景广阔的策略,而肿瘤免疫疗法后的复发问题已引起越来越多的关注。癌症干细胞(CSCs)是一小部分具有自我更新和分化能力的肿瘤细胞,对放疗和化疗等传统疗法具有抗药性。最近,CSCs 被证实是免疫疗法后肿瘤复发的驱动细胞。然而,CSCs 与癌症龛位免疫细胞之间的相互作用在很大程度上还没有定性。在这篇综述中,我们将重点讨论结直肠干细胞、干细胞与免疫细胞之间的相互作用以及基于干细胞的免疫疗法。结直肠癌 CSCs 的特点是表面标志物(如 CD44、CD133 和 Lgr5)的强表达;干性相关信号通路(如 Wnt/β-catenin、Hippo/Yap1、Jak/Stat 和 Notch 通路)的过度激活;以及表观遗传修饰紊乱,包括 DNA 甲基化、组蛋白修饰、染色质重塑和非编码 RNA 作用。此外,结直肠癌 CSCs 表达异常水平的免疫相关基因,如 MHC 和免疫检查点分子,并在多个肿瘤发生相关过程中与癌症龛细胞相互作用,包括肿瘤的发生、维持、转移和耐药性。迄今为止,许多针对 CSCs 的疗法已得到评估,包括单克隆抗体、抗体药物共轭物、双特异性抗体、肿瘤疫苗、收养细胞疗法和小分子抑制剂。随着 CSC/小分子靶向技术的发展以及多学科研究的整合,有望开发出消除 CSCs 并逆转其免疫抑制微环境的新型疗法,用于治疗包括结直肠癌在内的实体瘤。
{"title":"Crosstalk between colorectal CSCs and immune cells in tumorigenesis, and strategies for targeting colorectal CSCs","authors":"Qi Zhao, Hong Zong, Pingping Zhu, Chang Su, Wenxue Tang, Zhenzhen Chen, Shuiling Jin","doi":"10.1186/s40164-024-00474-x","DOIUrl":"https://doi.org/10.1186/s40164-024-00474-x","url":null,"abstract":"Cancer immunotherapy has emerged as a promising strategy in the treatment of colorectal cancer, and relapse after tumor immunotherapy has attracted increasing attention. Cancer stem cells (CSCs), a small subset of tumor cells with self-renewal and differentiation capacities, are resistant to traditional therapies such as radiotherapy and chemotherapy. Recently, CSCs have been proven to be the cells driving tumor relapse after immunotherapy. However, the mutual interactions between CSCs and cancer niche immune cells are largely uncharacterized. In this review, we focus on colorectal CSCs, CSC-immune cell interactions and CSC-based immunotherapy. Colorectal CSCs are characterized by robust expression of surface markers such as CD44, CD133 and Lgr5; hyperactivation of stemness-related signaling pathways, such as the Wnt/β-catenin, Hippo/Yap1, Jak/Stat and Notch pathways; and disordered epigenetic modifications, including DNA methylation, histone modification, chromatin remodeling, and noncoding RNA action. Moreover, colorectal CSCs express abnormal levels of immune-related genes such as MHC and immune checkpoint molecules and mutually interact with cancer niche cells in multiple tumorigenesis-related processes, including tumor initiation, maintenance, metastasis and drug resistance. To date, many therapies targeting CSCs have been evaluated, including monoclonal antibodies, antibody‒drug conjugates, bispecific antibodies, tumor vaccines adoptive cell therapy, and small molecule inhibitors. With the development of CSC-/niche-targeting technology, as well as the integration of multidisciplinary studies, novel therapies that eliminate CSCs and reverse their immunosuppressive microenvironment are expected to be developed for the treatment of solid tumors, including colorectal cancer.","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":10.9,"publicationDate":"2024-01-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139517675","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
Outcomes of allogeneic hematopoietic stem cell transplantation in R/R DLBCL patients with failure of CAR-T therapy CAR-T 疗法失败的 R/R DLBCL 患者接受异基因造血干细胞移植的结果
IF 10.9 1区 医学 Q1 Medicine Pub Date : 2024-01-16 DOI: 10.1186/s40164-024-00473-y
Mengya Cong, Sicheng Ai, Liqing Kang, Mao Jin, Ying Zhu, Caixia Li, Zhengming Jin, Lei Yu, Depei Wu, Haiwen Huang
From October 2017 to June 2022, we retrospectively report outcomes of R/R DLBCL patients with failure of CAR-T therapy, then receiving allo-HSCT. Among 10 patients, 5 were males and 5 females, with a median age of 43.5 (27–52) years. All patients were diagnosed refractory/relapsed diffuse large B cell lymphoma. The median time from CAR-T treatment to transplantation was 84.5 (31–370) days. The median follow-up was 21 (3–69) months. 5/10 patients attained CR and 1/10 patient attained PR during the follow up. The objective response rate (ORR) was 60%. The 1-year overall survival (OS) and progression-free survival (PFS) were 70% and 40%, respectively. At the time of the analysis, 6 patients were still living. During the follow up, four patients have died and the causes were disease relapses and progressions (2 patients), acute renal failure (1 patient), severe pulmonary infection (1 patient). Non-relapse was 20.0%.
2017年10月至2022年6月,我们回顾性报告了CAR-T疗法失败后接受allo-HSCT的R/R DLBCL患者的治疗结果。10名患者中,男性5人,女性5人,中位年龄为43.5(27-52)岁。所有患者均被诊断为难治性/复发性弥漫大B细胞淋巴瘤。从CAR-T治疗到移植的中位时间为84.5(31-370)天。中位随访时间为 21(3-69)个月。随访期间,5/10 的患者达到 CR,1/10 的患者达到 PR。客观反应率(ORR)为60%。1年总生存期(OS)和无进展生存期(PFS)分别为70%和40%。在进行分析时,仍有 6 名患者存活。在随访期间,有4名患者死亡,死亡原因分别是疾病复发和进展(2名患者)、急性肾功能衰竭(1名患者)、严重肺部感染(1名患者)。未复发率为 20.0%。
{"title":"Outcomes of allogeneic hematopoietic stem cell transplantation in R/R DLBCL patients with failure of CAR-T therapy","authors":"Mengya Cong, Sicheng Ai, Liqing Kang, Mao Jin, Ying Zhu, Caixia Li, Zhengming Jin, Lei Yu, Depei Wu, Haiwen Huang","doi":"10.1186/s40164-024-00473-y","DOIUrl":"https://doi.org/10.1186/s40164-024-00473-y","url":null,"abstract":"From October 2017 to June 2022, we retrospectively report outcomes of R/R DLBCL patients with failure of CAR-T therapy, then receiving allo-HSCT. Among 10 patients, 5 were males and 5 females, with a median age of 43.5 (27–52) years. All patients were diagnosed refractory/relapsed diffuse large B cell lymphoma. The median time from CAR-T treatment to transplantation was 84.5 (31–370) days. The median follow-up was 21 (3–69) months. 5/10 patients attained CR and 1/10 patient attained PR during the follow up. The objective response rate (ORR) was 60%. The 1-year overall survival (OS) and progression-free survival (PFS) were 70% and 40%, respectively. At the time of the analysis, 6 patients were still living. During the follow up, four patients have died and the causes were disease relapses and progressions (2 patients), acute renal failure (1 patient), severe pulmonary infection (1 patient). Non-relapse was 20.0%.","PeriodicalId":12180,"journal":{"name":"Experimental Hematology & Oncology","volume":null,"pages":null},"PeriodicalIF":10.9,"publicationDate":"2024-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139474649","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
Experimental Hematology & Oncology
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