首页 > 最新文献

Cellular Oncology最新文献

英文 中文
Non-glycanated ΔDCN isoform in muscle invasive bladder cancer mediates cancer stemness and gemcitabine resistance. 肌浸润性膀胱癌中的非糖化ΔDCN同工酶介导癌症干性和吉西他滨耐药性。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-10-28 DOI: 10.1007/s13402-024-00998-8
Nisha Wu, Jinxiang Wang, Mingming Fan, Yanling Liang, Xiao Wei Qi, Fan Deng, Fangyin Zeng

Background: The small leucine-rich proteoglycan decorin (DCN) is recognized for its diverse roles in tissue homeostasis and malignant progression. Nevertheless, the regulatory effects of DCN on bladder cancer stem cells (BCSCs) and the underlying mechanisms in muscle-invasive bladder cancer (MIBC) remain to be elucidated.

Methods: The study obtained data (including scRNA-seq, clinicopathological characteristics, and survival) were acquired from TCGA and GEO. The BCSCs were cultured by enriching the suspension culture in a serum-free medium, followed by flow cytometry sorting. Overexpression/knockdown was constructed by utilizing lentivirus. The surface biomarkers of cancer stem cells were identified via flow cytometry. Cell proliferation and self-renewal were evaluated by CCK8 and Sphere formation assays, and in vivo tumor growth was evaluated with subcutaneous xenografts.

Results: Total DCN expression was significantly elevated in muscle-invasive bladder cancer (MIBC) and was associated with poor prognosis. The ΔDCN isoform, which lacks glycosylation sites, was identified in bladder cancer stem cells (BCSCs) derived from clinical tissue samples and bladder cancer cell lines. Suppression of ΔDCN expression resulted in a reduction of BCSC stemness. Both in vitro and in vivo experiments indicated that overexpression of full-length DCN inhibited stemness within the extracellular matrix. Conversely, overexpression of ΔDCN and the introduction of exogenous recombinant decorin protein in ΔDCN-knockdown BCSC-SW780 cell lines enhanced stemness within the cytoplasm. The ΔDCN isoform exhibited resistance to gemcitabine chemotherapy in vitro.

Conclusion: Non-glycanated ΔDCN isoforms were identified in bladder cancer stem cells (BCSCs), where they exhibited differential cytoplasmic localization and promoted oncogenic effects by inducing a stemness phenotype and conferring resistance to gemcitabine chemotherapy. These oncogenic effects are in stark contrast to the anti-tumor functions of glycosylated DCN in the extracellular matrix. The ratio of ΔDCN isoforms to glycosylated DCN is pivotal in predicting tumor progression and therapeutic resistance.

背景:富含亮氨酸的小蛋白多糖decorin(DCN)被认为在组织稳态和恶性进展中发挥着多种作用。然而,DCN对膀胱癌干细胞(BSCs)的调控作用及其在肌肉浸润性膀胱癌(MIBC)中的潜在机制仍有待阐明:研究获得的数据(包括 scRNA-seq、临床病理特征和存活率)来自 TCGA 和 GEO。在无血清培养基中富集悬浮培养的 BCSCs,然后进行流式细胞术分选。利用慢病毒构建过表达/基因敲除模型。通过流式细胞术鉴定癌症干细胞的表面生物标志物。细胞增殖和自我更新通过CCK8和球形成试验进行评估,体内肿瘤生长通过皮下异种移植进行评估:结果:在肌浸润性膀胱癌(MIBC)中,DCN总表达量明显升高,且与预后不良有关。在来自临床组织样本和膀胱癌细胞系的膀胱癌干细胞(BCSCs)中发现了缺乏糖基化位点的ΔDCN异构体。抑制ΔDCN的表达可降低膀胱癌干细胞的干性。体外和体内实验都表明,过表达全长DCN会抑制细胞外基质中的干细胞。相反,在ΔDCN敲除的BCSC-SW780细胞系中,过表达ΔDCN和引入外源重组decorin蛋白可增强细胞质内的干性。ΔDCN异构体在体外表现出对吉西他滨化疗的耐药性:结论:在膀胱癌干细胞(BCSCs)中发现了非糖化ΔDCN异构体,它们表现出不同的细胞质定位,并通过诱导干性表型和赋予吉西他滨化疗抗性来促进致癌效应。这些致癌作用与细胞外基质中糖基化 DCN 的抗肿瘤功能形成了鲜明对比。ΔDCN异构体与糖基化DCN的比例在预测肿瘤进展和治疗耐药性方面至关重要。
{"title":"Non-glycanated ΔDCN isoform in muscle invasive bladder cancer mediates cancer stemness and gemcitabine resistance.","authors":"Nisha Wu, Jinxiang Wang, Mingming Fan, Yanling Liang, Xiao Wei Qi, Fan Deng, Fangyin Zeng","doi":"10.1007/s13402-024-00998-8","DOIUrl":"https://doi.org/10.1007/s13402-024-00998-8","url":null,"abstract":"<p><strong>Background: </strong>The small leucine-rich proteoglycan decorin (DCN) is recognized for its diverse roles in tissue homeostasis and malignant progression. Nevertheless, the regulatory effects of DCN on bladder cancer stem cells (BCSCs) and the underlying mechanisms in muscle-invasive bladder cancer (MIBC) remain to be elucidated.</p><p><strong>Methods: </strong>The study obtained data (including scRNA-seq, clinicopathological characteristics, and survival) were acquired from TCGA and GEO. The BCSCs were cultured by enriching the suspension culture in a serum-free medium, followed by flow cytometry sorting. Overexpression/knockdown was constructed by utilizing lentivirus. The surface biomarkers of cancer stem cells were identified via flow cytometry. Cell proliferation and self-renewal were evaluated by CCK8 and Sphere formation assays, and in vivo tumor growth was evaluated with subcutaneous xenografts.</p><p><strong>Results: </strong>Total DCN expression was significantly elevated in muscle-invasive bladder cancer (MIBC) and was associated with poor prognosis. The ΔDCN isoform, which lacks glycosylation sites, was identified in bladder cancer stem cells (BCSCs) derived from clinical tissue samples and bladder cancer cell lines. Suppression of ΔDCN expression resulted in a reduction of BCSC stemness. Both in vitro and in vivo experiments indicated that overexpression of full-length DCN inhibited stemness within the extracellular matrix. Conversely, overexpression of ΔDCN and the introduction of exogenous recombinant decorin protein in ΔDCN-knockdown BCSC-SW780 cell lines enhanced stemness within the cytoplasm. The ΔDCN isoform exhibited resistance to gemcitabine chemotherapy in vitro.</p><p><strong>Conclusion: </strong>Non-glycanated ΔDCN isoforms were identified in bladder cancer stem cells (BCSCs), where they exhibited differential cytoplasmic localization and promoted oncogenic effects by inducing a stemness phenotype and conferring resistance to gemcitabine chemotherapy. These oncogenic effects are in stark contrast to the anti-tumor functions of glycosylated DCN in the extracellular matrix. The ratio of ΔDCN isoforms to glycosylated DCN is pivotal in predicting tumor progression and therapeutic resistance.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-10-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142523456","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting tumor associated macrophages (TAMs) reprograms tumor immune microenvironment to promote solid tumor immunotherapy. 以肿瘤相关巨噬细胞(TAMs)为靶点,重新规划肿瘤免疫微环境,促进实体瘤免疫疗法。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-10-01 Epub Date: 2024-09-05 DOI: 10.1007/s13402-024-00987-x
Chunliu Huang, Xiumei Wang, Lixiang Wang, Yujia Liu, Zijin Xia, Xinyu Wang, Jun Chen
{"title":"Targeting tumor associated macrophages (TAMs) reprograms tumor immune microenvironment to promote solid tumor immunotherapy.","authors":"Chunliu Huang, Xiumei Wang, Lixiang Wang, Yujia Liu, Zijin Xia, Xinyu Wang, Jun Chen","doi":"10.1007/s13402-024-00987-x","DOIUrl":"10.1007/s13402-024-00987-x","url":null,"abstract":"","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142134210","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
SPG21, a potential oncogene targeted by miR-128-3p, amplifies HBx-induced carcinogenesis and chemoresistance via activation of TRPM7-mediated JNK pathway in hepatocellular carcinoma. SPG21是miR-128-3p靶向的潜在癌基因,它通过激活TRPM7介导的肝细胞癌JNK通路,扩大了HBx诱导的癌变和化疗耐药性。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-10-01 Epub Date: 2024-05-16 DOI: 10.1007/s13402-024-00955-5
Ping Zhou, Wei Yao, Lijuan Liu, Qiujin Yan, Xiaobei Chen, Xiaocui Wei, Shuang Ding, Zhao Lv, Fan Zhu

Purpose: Chronic hepatitis B virus (HBV) infection is the primary risk factor for the malignant progression of hepatocellular carcinoma (HCC). It has been reported that HBV X protein (HBx) possesses oncogenic properties, promoting hepatocarcinogenesis and chemoresistance. However, the detailed molecular mechanisms are not fully understood. Here, we aim to investigate the effects of miR-128-3p/SPG21 axis on HBx-induced hepatocarcinogenesis and chemoresistance.

Methods: The expression of SPG21 in HCC was determined using bioinformatics analysis, quantitative real-time PCR (qRT-PCR), western blotting, and immunohistochemistry (IHC). The roles of SPG21 in HCC were elucidated through a series of in vitro and in vivo experiments, including real-time cellular analysis (RTCA), matrigel invasion assay, and xenograft mouse model. Pharmacologic treatment and flow cytometry were performed to demonstrate the potential mechanism of SPG21 in HCC.

Results: SPG21 expression was elevated in HCC tissues compared to adjacent non-tumor tissues (NTs). Moreover, higher SPG21 expression correlated with poor overall survival. Functional assays revealed that SPG21 fostered HCC tumorigenesis and invasion. MiR-128-3p, which targeted SPG21, was downregulated in HCC tissues. Subsequent analyses showed that HBx amplified TRPM7-mediated calcium influx via miR-128-3p/SPG21, thereby activating the c-Jun N-terminal kinase (JNK) pathway. Furthermore, HBx inhibited doxorubicin-induced apoptosis by engaging the JNK pathway through miR-128-3p/SPG21.

Conclusion: The study suggested that SPG21, targeted by miR-128-3p, might be involved in enhancing HBx-induced carcinogenesis and doxorubicin resistance in HCC via the TRPM7/Ca2+/JNK signaling pathway. This insight suggested that SPG21 could be recognized as a potential oncogene, offering a novel perspective on its role as a prognostic factor and a therapeutic target in the context of HCC.

目的:慢性乙型肝炎病毒(HBV)感染是肝细胞癌(HCC)恶性进展的主要风险因素。据报道,HBV X 蛋白(HBx)具有致癌特性,可促进肝癌的发生和化疗耐药性。然而,详细的分子机制尚未完全明了。在此,我们旨在研究 miR-128-3p/SPG21 轴对 HBx 诱导的肝癌发生和化疗耐药性的影响:方法:采用生物信息学分析、定量实时 PCR(qRT-PCR)、Western 印迹和免疫组化(IHC)等方法测定 SPG21 在 HCC 中的表达。通过一系列体内外实验,包括实时细胞分析(RTCA)、matrigel侵袭实验和异种移植小鼠模型,阐明了SPG21在HCC中的作用。通过药物治疗和流式细胞术证明了SPG21在HCC中的潜在作用机制:结果:与邻近的非肿瘤组织(NTs)相比,SPG21在HCC组织中的表达升高。此外,较高的 SPG21 表达与较差的总生存率相关。功能测试显示 SPG21 促进了 HCC 的肿瘤发生和侵袭。针对 SPG21 的 MiR-128-3p 在 HCC 组织中被下调。随后的分析表明,HBx通过miR-128-3p/SPG21扩大了TRPM7介导的钙离子流入,从而激活了c-Jun N-末端激酶(JNK)通路。此外,HBx 通过 miR-128-3p/SPG21 参与 JNK 通路,抑制了多柔比星诱导的细胞凋亡:该研究表明,miR-128-3p靶向的SPG21可能通过TRPM7/Ca2+/JNK信号通路参与增强HBx诱导的癌变和多柔比星在HCC中的抗性。这一发现表明 SPG21 可被视为一种潜在的癌基因,为其在 HCC 中作为预后因素和治疗靶点的作用提供了新的视角。
{"title":"SPG21, a potential oncogene targeted by miR-128-3p, amplifies HBx-induced carcinogenesis and chemoresistance via activation of TRPM7-mediated JNK pathway in hepatocellular carcinoma.","authors":"Ping Zhou, Wei Yao, Lijuan Liu, Qiujin Yan, Xiaobei Chen, Xiaocui Wei, Shuang Ding, Zhao Lv, Fan Zhu","doi":"10.1007/s13402-024-00955-5","DOIUrl":"10.1007/s13402-024-00955-5","url":null,"abstract":"<p><strong>Purpose: </strong>Chronic hepatitis B virus (HBV) infection is the primary risk factor for the malignant progression of hepatocellular carcinoma (HCC). It has been reported that HBV X protein (HBx) possesses oncogenic properties, promoting hepatocarcinogenesis and chemoresistance. However, the detailed molecular mechanisms are not fully understood. Here, we aim to investigate the effects of miR-128-3p/SPG21 axis on HBx-induced hepatocarcinogenesis and chemoresistance.</p><p><strong>Methods: </strong>The expression of SPG21 in HCC was determined using bioinformatics analysis, quantitative real-time PCR (qRT-PCR), western blotting, and immunohistochemistry (IHC). The roles of SPG21 in HCC were elucidated through a series of in vitro and in vivo experiments, including real-time cellular analysis (RTCA), matrigel invasion assay, and xenograft mouse model. Pharmacologic treatment and flow cytometry were performed to demonstrate the potential mechanism of SPG21 in HCC.</p><p><strong>Results: </strong>SPG21 expression was elevated in HCC tissues compared to adjacent non-tumor tissues (NTs). Moreover, higher SPG21 expression correlated with poor overall survival. Functional assays revealed that SPG21 fostered HCC tumorigenesis and invasion. MiR-128-3p, which targeted SPG21, was downregulated in HCC tissues. Subsequent analyses showed that HBx amplified TRPM7-mediated calcium influx via miR-128-3p/SPG21, thereby activating the c-Jun N-terminal kinase (JNK) pathway. Furthermore, HBx inhibited doxorubicin-induced apoptosis by engaging the JNK pathway through miR-128-3p/SPG21.</p><p><strong>Conclusion: </strong>The study suggested that SPG21, targeted by miR-128-3p, might be involved in enhancing HBx-induced carcinogenesis and doxorubicin resistance in HCC via the TRPM7/Ca<sup>2+</sup>/JNK signaling pathway. This insight suggested that SPG21 could be recognized as a potential oncogene, offering a novel perspective on its role as a prognostic factor and a therapeutic target in the context of HCC.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140944885","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Marine derived macrolide bryostatin 4 inhibits the TGF-β signaling pathway against acute erythroleukemia. 海洋衍生大环内酯类药物白屈菜素 4 可抑制 TGF-β 信号通路,对抗急性红细胞白血病。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-10-01 Epub Date: 2024-07-31 DOI: 10.1007/s13402-024-00968-0
Yan-Yu Kou, Jie Liu, Yung-Ting Chang, Li-Yun Liu, Fan Sun, Yi-Lin Li, Jia-Rong Leng, Hou-Wen Lin, Fan Yang

Purpose: Acute erythroleukemia (AEL) is a rare and highly aggressive subtype of acute myeloid leukemia (AML) with an extremely poor prognosis when treated with available drugs. Therefore, new investigational agents capable of inducing remission are urgently required.

Methods: Bioinformatics analysis, western blot and qRT-PCR were used to reveal the potential biological mechanism of bryostatin 4 (B4), an antineoplastic macrolide derived from the marine bryozoan Bugula neritina. Then, in vivo experiments were conducted to evaluate the role of transforming growth factor (TGF)-β signaling in the progression of AEL.

Results: Our results revealed that the proliferation of K562 cells and TF-1 cells was significantly inhibited by B4 at IC50 values of 37 nM and 52 nM, respectively. B4 inhibited TGF-β signaling and its downstream pathway targets, particularly the phosphorylation of Smad2, Smad3, Ras, C-RAF, ERK1/2, and MEK. B4 also played an important role in cell invasion and migration in K562 cells and TF-1 cells by reducing the protein levels of the mesenchymal cell marker vimentin. Moreover, Flow cytometry and western blot analyses demonstrated that B4 induced apoptosis and initiated G0/G1 phase arrest by modulating mitochondrial dysfunction and cyclin-dependent kinase (CDK) expression.

Conclusion: These findings indicated that B4 could inhibit the proliferation, migration, invasion, and TGF-β signaling pathways of AEL cells, thus suggesting that B4 possesses therapeutic potential as a treatment for AEL.

目的:急性红细胞白血病(AEL)是急性髓性白血病(AML)的一种罕见且侵袭性极强的亚型,用现有药物治疗预后极差。因此,迫切需要能够诱导病情缓解的新研究药物:方法:采用生物信息学分析、Western 印迹和 qRT-PCR 技术揭示了从海洋贝类 Bugula neritina 中提取的抗肿瘤大环内酯类药物--白僵菌素 4(B4)的潜在生物机制。然后,进行了体内实验以评估转化生长因子(TGF)-β信号在AEL进展过程中的作用:结果表明,B4能显著抑制K562细胞和TF-1细胞的增殖,IC50值分别为37 nM和52 nM。B4 可抑制 TGF-β 信号转导及其下游通路靶点,尤其是 Smad2、Smad3、Ras、C-RAF、ERK1/2 和 MEK 的磷酸化。B4 还通过降低间质细胞标志物波形蛋白的蛋白水平,在 K562 细胞和 TF-1 细胞的侵袭和迁移中发挥了重要作用。此外,流式细胞术和 Western 印迹分析表明,B4 通过调节线粒体功能障碍和细胞周期蛋白依赖性激酶(CDK)的表达,诱导细胞凋亡并启动 G0/G1 期停滞:这些研究结果表明,B4可抑制AEL细胞的增殖、迁移、侵袭和TGF-β信号通路,从而表明B4具有治疗AEL的潜力。
{"title":"Marine derived macrolide bryostatin 4 inhibits the TGF-β signaling pathway against acute erythroleukemia.","authors":"Yan-Yu Kou, Jie Liu, Yung-Ting Chang, Li-Yun Liu, Fan Sun, Yi-Lin Li, Jia-Rong Leng, Hou-Wen Lin, Fan Yang","doi":"10.1007/s13402-024-00968-0","DOIUrl":"10.1007/s13402-024-00968-0","url":null,"abstract":"<p><strong>Purpose: </strong>Acute erythroleukemia (AEL) is a rare and highly aggressive subtype of acute myeloid leukemia (AML) with an extremely poor prognosis when treated with available drugs. Therefore, new investigational agents capable of inducing remission are urgently required.</p><p><strong>Methods: </strong>Bioinformatics analysis, western blot and qRT-PCR were used to reveal the potential biological mechanism of bryostatin 4 (B4), an antineoplastic macrolide derived from the marine bryozoan Bugula neritina. Then, in vivo experiments were conducted to evaluate the role of transforming growth factor (TGF)-β signaling in the progression of AEL.</p><p><strong>Results: </strong>Our results revealed that the proliferation of K562 cells and TF-1 cells was significantly inhibited by B4 at IC<sub>50</sub> values of 37 nM and 52 nM, respectively. B4 inhibited TGF-β signaling and its downstream pathway targets, particularly the phosphorylation of Smad2, Smad3, Ras, C-RAF, ERK1/2, and MEK. B4 also played an important role in cell invasion and migration in K562 cells and TF-1 cells by reducing the protein levels of the mesenchymal cell marker vimentin. Moreover, Flow cytometry and western blot analyses demonstrated that B4 induced apoptosis and initiated G0/G1 phase arrest by modulating mitochondrial dysfunction and cyclin-dependent kinase (CDK) expression.</p><p><strong>Conclusion: </strong>These findings indicated that B4 could inhibit the proliferation, migration, invasion, and TGF-β signaling pathways of AEL cells, thus suggesting that B4 possesses therapeutic potential as a treatment for AEL.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141856931","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
HVEM in acute lymphocytic leukemia facilitates tumour immune escape by inhibiting CD8+ T cell function. 急性淋巴细胞白血病中的 HVEM 可通过抑制 CD8+ T 细胞功能促进肿瘤免疫逃逸。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-10-01 Epub Date: 2024-05-29 DOI: 10.1007/s13402-024-00959-1
Yujia Liu, Lixiang Wang, Yiyi Li, Cheng Zhong, Xiumei Wang, Xinyu Wang, Zijin Xia, Jing Liao, Chunliu Huang, Chengzhou Mao, Yongyi Feng, Congzhou Luo, Wenhao Mai, Hongrui Song, Hongyu Li, Lin Bao, Danchun Chen, Yue Sheng, Hui Zhang, Xiaolei Wei, Jun Chen, Wei Yi

Purpose: Leukaemia remains a major contributor to global mortality, representing a significant health risk for a substantial number of cancer patients. Despite notable advancements in the field, existing treatments frequently exhibit limited efficacy or recurrence. Here, we explored the potential of abolishing HVEM (herpes virus entry mediator, TNFRSF14) expression in tumours as an effective approach to treat acute lymphoblastic leukaemia (ALL) and prevent its recurrence.

Methods: The clinical correlations between HVEM and leukaemia were revealed by public data analysis. HVEM knockout (KO) murine T cell lymphoblastic leukaemia cell line EL4 were generated using CRISPR-Cas9 technology, and syngeneic subcutaneous tumour models were established to investigate the in vivo function of HVEM. Immunohistochemistry (IHC), RNA-seq and flow cytometry were used to analyse the tumour immune microenvironment (TIME) and tumour draining lymph nodes (dLNs). Immune functions were investigated by depletion of immune subsets in vivo and T cell functional assays in vitro. The HVEM mutant EL4 cell lines were constructed to investigate the functional domain responsible for immune escape.

Results: According to public databases, HVEM is highly expressed in patients with ALL and acute myeloid leukemia (AML) and is negatively correlated with patient prognosis. Genetic deletion of HVEM in EL4 cells markedly inhibited tumour progression and prolonged the survival of tumour-bearing mice. Our experiments proved that HVEM exerted its immunosuppressive effect by inhibiting antitumour function of CD8+ T cell through CRD1 domain both in vivo and in vitro. Additionally, we identified a combination therapy capable of completely eradicating ALL tumours, which induces immune memory toward tumour protection.

Conclusions: Our study reveals the potential mechanisms by which HVEM facilitates ALL progression, and highlights HVEM as a promising target for clinical applications in relapsed ALL therapy.

目的:白血病仍然是造成全球死亡的一个主要因素,对大量癌症患者的健康构成重大威胁。尽管该领域取得了显著进展,但现有治疗方法经常显示出有限的疗效或复发。在此,我们探讨了取消肿瘤中HVEM(疱疹病毒进入介质,TNFRSF14)表达作为治疗急性淋巴细胞白血病(ALL)和防止其复发的有效方法的潜力:方法:通过公开数据分析揭示了HVEM与白血病之间的临床相关性。利用CRISPR-Cas9技术产生HVEM基因敲除(KO)小鼠T细胞淋巴母细胞白血病细胞株EL4,并建立合成皮下肿瘤模型以研究HVEM的体内功能。免疫组织化学(IHC)、RNA-seq和流式细胞术用于分析肿瘤免疫微环境(TIME)和肿瘤引流淋巴结(dLNs)。通过体内免疫亚群耗竭和体外T细胞功能测试研究了免疫功能。构建了HVEM突变体EL4细胞系,以研究导致免疫逃逸的功能域:公共数据库显示,HVEM在ALL和急性髓性白血病(AML)患者中高表达,并与患者预后呈负相关。在EL4细胞中遗传性删除HVEM可明显抑制肿瘤的发展,并延长肿瘤小鼠的生存期。我们的实验证明,HVEM通过CRD1结构域在体内和体外抑制CD8+ T细胞的抗肿瘤功能,从而发挥免疫抑制作用。此外,我们还发现了一种能够彻底根除 ALL 肿瘤的联合疗法,它能诱导肿瘤保护免疫记忆:我们的研究揭示了HVEM促进ALL进展的潜在机制,并强调了HVEM是复发ALL治疗临床应用的一个有希望的靶点。
{"title":"HVEM in acute lymphocytic leukemia facilitates tumour immune escape by inhibiting CD8<sup>+</sup> T cell function.","authors":"Yujia Liu, Lixiang Wang, Yiyi Li, Cheng Zhong, Xiumei Wang, Xinyu Wang, Zijin Xia, Jing Liao, Chunliu Huang, Chengzhou Mao, Yongyi Feng, Congzhou Luo, Wenhao Mai, Hongrui Song, Hongyu Li, Lin Bao, Danchun Chen, Yue Sheng, Hui Zhang, Xiaolei Wei, Jun Chen, Wei Yi","doi":"10.1007/s13402-024-00959-1","DOIUrl":"10.1007/s13402-024-00959-1","url":null,"abstract":"<p><strong>Purpose: </strong>Leukaemia remains a major contributor to global mortality, representing a significant health risk for a substantial number of cancer patients. Despite notable advancements in the field, existing treatments frequently exhibit limited efficacy or recurrence. Here, we explored the potential of abolishing HVEM (herpes virus entry mediator, TNFRSF14) expression in tumours as an effective approach to treat acute lymphoblastic leukaemia (ALL) and prevent its recurrence.</p><p><strong>Methods: </strong>The clinical correlations between HVEM and leukaemia were revealed by public data analysis. HVEM knockout (KO) murine T cell lymphoblastic leukaemia cell line EL4 were generated using CRISPR-Cas9 technology, and syngeneic subcutaneous tumour models were established to investigate the in vivo function of HVEM. Immunohistochemistry (IHC), RNA-seq and flow cytometry were used to analyse the tumour immune microenvironment (TIME) and tumour draining lymph nodes (dLNs). Immune functions were investigated by depletion of immune subsets in vivo and T cell functional assays in vitro. The HVEM mutant EL4 cell lines were constructed to investigate the functional domain responsible for immune escape.</p><p><strong>Results: </strong>According to public databases, HVEM is highly expressed in patients with ALL and acute myeloid leukemia (AML) and is negatively correlated with patient prognosis. Genetic deletion of HVEM in EL4 cells markedly inhibited tumour progression and prolonged the survival of tumour-bearing mice. Our experiments proved that HVEM exerted its immunosuppressive effect by inhibiting antitumour function of CD8<sup>+</sup> T cell through CRD1 domain both in vivo and in vitro. Additionally, we identified a combination therapy capable of completely eradicating ALL tumours, which induces immune memory toward tumour protection.</p><p><strong>Conclusions: </strong>Our study reveals the potential mechanisms by which HVEM facilitates ALL progression, and highlights HVEM as a promising target for clinical applications in relapsed ALL therapy.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141161475","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Tertiary lymphoid structures and their therapeutic implications in cancer. 三级淋巴结构及其对癌症的治疗意义。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-10-01 Epub Date: 2024-08-12 DOI: 10.1007/s13402-024-00975-1
Xun Chen, Pan Wu, Ziqi Liu, Tiansheng Li, Jie Wu, Zhaoyang Zeng, Wenjia Guo, Wei Xiong

Tertiary lymphoid structures (TLSs) are ectopic lymphoid aggregates formed by the structured accumulation of immune cells such as B cells and T cells in non-lymphoid tissues induced by infection, inflammation, and tumors. They play a crucial role in the immune response, particularly in association with tumor development, where they primarily exert anti-tumor immune functions during tumorigenesis. Current research suggests that TLSs inhibit tumor growth by facilitating immune cell infiltration and are correlated with favorable prognosis in various solid tumors, serving as an indicator of immunotherapy effectiveness to some extent. Therefore, TLSs hold great promise as a valuable biomarker. Most importantly, immunotherapies aimed to prompting TLSs formation are anticipated to be potent adjuncts to current cancer treatment. This review focuses on the formation process of TLSs and their potential applications in cancer therapy.

三级淋巴结构(TLS)是由感染、炎症和肿瘤诱导的免疫细胞(如 B 细胞和 T 细胞)在非淋巴组织中结构性聚集形成的异位淋巴聚集体。它们在免疫反应中发挥着至关重要的作用,尤其是在肿瘤发生过程中,它们主要发挥抗肿瘤免疫功能。目前的研究表明,TLS 通过促进免疫细胞浸润来抑制肿瘤生长,并与各种实体瘤的良好预后相关,在一定程度上可作为免疫疗法有效性的指标。因此,TLS 很有希望成为一种有价值的生物标记物。最重要的是,旨在促使 TLSs 形成的免疫疗法有望成为当前癌症治疗的有效辅助手段。本综述将重点讨论 TLSs 的形成过程及其在癌症治疗中的潜在应用。
{"title":"Tertiary lymphoid structures and their therapeutic implications in cancer.","authors":"Xun Chen, Pan Wu, Ziqi Liu, Tiansheng Li, Jie Wu, Zhaoyang Zeng, Wenjia Guo, Wei Xiong","doi":"10.1007/s13402-024-00975-1","DOIUrl":"10.1007/s13402-024-00975-1","url":null,"abstract":"<p><p>Tertiary lymphoid structures (TLSs) are ectopic lymphoid aggregates formed by the structured accumulation of immune cells such as B cells and T cells in non-lymphoid tissues induced by infection, inflammation, and tumors. They play a crucial role in the immune response, particularly in association with tumor development, where they primarily exert anti-tumor immune functions during tumorigenesis. Current research suggests that TLSs inhibit tumor growth by facilitating immune cell infiltration and are correlated with favorable prognosis in various solid tumors, serving as an indicator of immunotherapy effectiveness to some extent. Therefore, TLSs hold great promise as a valuable biomarker. Most importantly, immunotherapies aimed to prompting TLSs formation are anticipated to be potent adjuncts to current cancer treatment. This review focuses on the formation process of TLSs and their potential applications in cancer therapy.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141917841","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Disrupting YAP1-mediated glutamine metabolism induces synthetic lethality alongside ODC1 inhibition in osteosarcoma. 破坏 YAP1 介导的谷氨酰胺代谢可诱导骨肉瘤中的合成致死率,同时抑制 ODC1。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-10-01 Epub Date: 2024-08-08 DOI: 10.1007/s13402-024-00967-1
Hongsheng Wang, Yining Tao, Jing Han, Jiakang Shen, Haoran Mu, Zhuoying Wang, Jinzeng Wang, Xinmeng Jin, Qi Zhang, Yuqin Yang, Jun Lin, Mengxiong Sun, Xiaojun Ma, Ling Ren, Amy K LeBlanc, Jing Xu, Yingqi Hua, Wei Sun
<p><strong>Purpose: </strong>Osteosarcoma, a highly malignant primary bone tumor primarily affecting adolescents, frequently develops resistance to initial chemotherapy, leading to metastasis and limited treatment options. Our study aims to uncover novel therapeutic targets for metastatic and recurrent osteosarcoma.</p><p><strong>Methods: </strong>In this study, we proved the potential of modulating the YAP1-regulated glutamine metabolic pathway to augment the response of OS to DFMO. We initially employed single-cell transcriptomic data to gauge the activation level of polyamine metabolism in MTAP-deleted OS patients. This was further substantiated by transcriptome sequencing data from recurrent and non-recurrent patient tissues, confirming the activation of polyamine metabolism in progressive OS. Through high-throughput drug screening, we pinpointed CIL56, a YAP1 inhibitor, as a promising candidate for a combined therapeutic strategy with DFMO. In vivo, we utilized PDX and CDX models to validate the therapeutic efficacy of this drug combination. In vitro, we conducted western blot analysis, qPCR analysis, immunofluorescence staining, and PuMA experiments to monitor alterations in molecular expression, distribution, and tumor metastasis capability. We employed CCK-8 and colony formation assays to assess the proliferative capacity of cells in the experimental group. We used flow cytometry and reactive oxygen probes to observe changes in ROS and glutamine metabolism within the cells. Finally, we applied RNA-seq in tandem with metabolomics to identify metabolic alterations in OS cells treated with a DFMO and CIL56 combination. This enabled us to intervene and validate the role of the YAP1-mediated glutamine metabolic pathway in DFMO resistance.</p><p><strong>Results: </strong>Through single-cell RNA-seq data analysis, we pinpointed a subset of late-stage OS cells with significantly upregulated polyamine metabolism. This upregulation was further substantiated by transcriptomic profiling of recurrent and non-recurrent OS tissues. High-throughput drug screening revealed a promising combination strategy involving DFMO and CIL56. DFMO treatment curbs the phosphorylation of YAP1 protein in OS cells, promoting nuclear entry and initiating the YAP1-mediated glutamine metabolic pathway. This reduces intracellular ROS levels, countering DFMO's anticancer effect. The therapeutic efficacy of DFMO can be amplified both in vivo and in vitro by combining it with the YAP1 inhibitor CIL56 or the glutaminase inhibitor CB-839. This underscores the significant potential of targeting the YAP1-mediated glutamine metabolic pathway to enhance efficacy of DFMO.</p><p><strong>Conclusion: </strong>Our findings elucidate YAP1-mediated glutamine metabolism as a crucial bypass mechanism against DFMO, following the inhibition of polyamine metabolism. Our study provides valuable insights into the potential role of DFMO in an "One-two Punch" therapy of metastatic and recurrent oste
目的:骨肉瘤是一种高度恶性的原发性骨肿瘤,主要影响青少年,经常对初始化疗产生耐药性,导致转移和治疗选择有限。我们的研究旨在发现治疗转移性和复发性骨肉瘤的新靶点:在这项研究中,我们证明了调节 YAP1 调控的谷氨酰胺代谢途径可增强 OS 对 DFMO 的反应。我们首先利用单细胞转录组数据来衡量MTAP缺失的OS患者体内多胺代谢的激活水平。复发性和非复发性患者组织的转录组测序数据进一步证实了这一点,证实了进行性OS中多胺代谢的激活。通过高通量药物筛选,我们发现 YAP1 抑制剂 CIL56 是与 DFMO 联合治疗策略的理想候选药物。在体内,我们利用 PDX 和 CDX 模型来验证这种药物组合的疗效。在体外,我们进行了 Western 印迹分析、qPCR 分析、免疫荧光染色和 PuMA 实验,以监测分子表达、分布和肿瘤转移能力的变化。我们采用 CCK-8 和集落形成试验来评估实验组细胞的增殖能力。我们使用流式细胞仪和活性氧探针观察细胞内 ROS 和谷氨酰胺代谢的变化。最后,我们将 RNA-seq 与代谢组学结合起来,确定了经 DFMO 和 CIL56 组合处理的 OS 细胞的代谢变化。这使我们能够干预和验证 YAP1 介导的谷氨酰胺代谢途径在 DFMO 抗性中的作用:结果:通过单细胞RNA-seq数据分析,我们确定了多胺代谢显著上调的晚期OS细胞亚群。复发性和非复发性 OS 组织的转录组分析进一步证实了这种上调。高通量药物筛选显示,DFMO和CIL56的组合策略很有前景。DFMO 治疗可抑制 OS 细胞中 YAP1 蛋白的磷酸化,促进核进入并启动 YAP1 介导的谷氨酰胺代谢途径。这降低了细胞内的 ROS 水平,抵消了 DFMO 的抗癌作用。将 DFMO 与 YAP1 抑制剂 CIL56 或谷氨酰胺酶抑制剂 CB-839 结合使用,可在体内和体外扩大 DFMO 的疗效。这凸显了靶向YAP1介导的谷氨酰胺代谢途径以提高DFMO疗效的巨大潜力:结论:我们的研究结果阐明了 YAP1 介导的谷氨酰胺代谢是继抑制多胺代谢之后对 DFMO 起关键作用的旁路机制。我们的研究为 DFMO 在转移性和复发性骨肉瘤的 "一对二冲剂 "疗法中的潜在作用提供了有价值的见解。
{"title":"Disrupting YAP1-mediated glutamine metabolism induces synthetic lethality alongside ODC1 inhibition in osteosarcoma.","authors":"Hongsheng Wang, Yining Tao, Jing Han, Jiakang Shen, Haoran Mu, Zhuoying Wang, Jinzeng Wang, Xinmeng Jin, Qi Zhang, Yuqin Yang, Jun Lin, Mengxiong Sun, Xiaojun Ma, Ling Ren, Amy K LeBlanc, Jing Xu, Yingqi Hua, Wei Sun","doi":"10.1007/s13402-024-00967-1","DOIUrl":"10.1007/s13402-024-00967-1","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Purpose: &lt;/strong&gt;Osteosarcoma, a highly malignant primary bone tumor primarily affecting adolescents, frequently develops resistance to initial chemotherapy, leading to metastasis and limited treatment options. Our study aims to uncover novel therapeutic targets for metastatic and recurrent osteosarcoma.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Methods: &lt;/strong&gt;In this study, we proved the potential of modulating the YAP1-regulated glutamine metabolic pathway to augment the response of OS to DFMO. We initially employed single-cell transcriptomic data to gauge the activation level of polyamine metabolism in MTAP-deleted OS patients. This was further substantiated by transcriptome sequencing data from recurrent and non-recurrent patient tissues, confirming the activation of polyamine metabolism in progressive OS. Through high-throughput drug screening, we pinpointed CIL56, a YAP1 inhibitor, as a promising candidate for a combined therapeutic strategy with DFMO. In vivo, we utilized PDX and CDX models to validate the therapeutic efficacy of this drug combination. In vitro, we conducted western blot analysis, qPCR analysis, immunofluorescence staining, and PuMA experiments to monitor alterations in molecular expression, distribution, and tumor metastasis capability. We employed CCK-8 and colony formation assays to assess the proliferative capacity of cells in the experimental group. We used flow cytometry and reactive oxygen probes to observe changes in ROS and glutamine metabolism within the cells. Finally, we applied RNA-seq in tandem with metabolomics to identify metabolic alterations in OS cells treated with a DFMO and CIL56 combination. This enabled us to intervene and validate the role of the YAP1-mediated glutamine metabolic pathway in DFMO resistance.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;Through single-cell RNA-seq data analysis, we pinpointed a subset of late-stage OS cells with significantly upregulated polyamine metabolism. This upregulation was further substantiated by transcriptomic profiling of recurrent and non-recurrent OS tissues. High-throughput drug screening revealed a promising combination strategy involving DFMO and CIL56. DFMO treatment curbs the phosphorylation of YAP1 protein in OS cells, promoting nuclear entry and initiating the YAP1-mediated glutamine metabolic pathway. This reduces intracellular ROS levels, countering DFMO's anticancer effect. The therapeutic efficacy of DFMO can be amplified both in vivo and in vitro by combining it with the YAP1 inhibitor CIL56 or the glutaminase inhibitor CB-839. This underscores the significant potential of targeting the YAP1-mediated glutamine metabolic pathway to enhance efficacy of DFMO.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Conclusion: &lt;/strong&gt;Our findings elucidate YAP1-mediated glutamine metabolism as a crucial bypass mechanism against DFMO, following the inhibition of polyamine metabolism. Our study provides valuable insights into the potential role of DFMO in an \"One-two Punch\" therapy of metastatic and recurrent oste","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141903355","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
COLEC10 inhibits the stemness of hepatocellular carcinoma by suppressing the activity of β-catenin signaling. COLEC10通过抑制β-catenin信号的活性来抑制肝细胞癌的干性。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-10-01 Epub Date: 2024-07-30 DOI: 10.1007/s13402-024-00972-4
Mei-Na Cai, Dong-Mei Chen, Xin-Ru Chen, Yu-Rong Gu, Chun-Hong Liao, Le-Xin Xiao, Jia-Liang Wang, Bing-Liang Lin, Yue-Hua Huang, Yi-Fan Lian

Background: Liver cancer stem cells (CSCs) contribute to tumor initiation, progression, and recurrence in hepatocellular carcinoma (HCC). The Wnt/β-catenin pathway plays a crucial role in liver cancer stemness, progression, metastasis, and drug resistance, but no clinically approved drugs have targeted this pathway efficiently so far. We aimed to elucidate the role of COLEC10 in HCC stemness.

Methods: The Cancer Genome Atlas (TCGA) and the Clinical Proteomic Tumor Analysis Consortium (CPTAC) databases were employed to search for the association between COLEC10 expression and HCC stemness. Colony formation, sphere formation, side population, and limiting dilution tumor initiation assays were used to identify the regulatory role of COLEC10 overexpression in the stemness of HCC cell lines. Wnt/β-catenin reporter assay and immunoprecipitation were performed to explore the underlying mechanism.

Results: COLEC10 level was negatively correlated with HCC stemness. Elevated COLEC10 led to decreased expressions of EpCAM and AFP (alpha-fetoprotein), two common markers of liver CSCs. Overexpression of COLEC10 inhibited HCC cells from forming colonies and spheres, and reduced the side population numbers in vitro, as well as the tumorigenic capacity in vivo. Mechanically, we demonstrated that overexpression of COLEC10 suppressed the activity of Wnt/β-catenin signaling by upregulating Wnt inhibitory factor WIF1 and reducing the level of cytoplasmic β-catenin. COLEC10 overexpression promoted the interaction of β-catenin with the component of destruction complex CK1α. In addition, KLHL22 (Kelch Like Family Member 22), a reported E3 ligase adaptor predicted to interact with CK1α, could facilitate COLEC10 monoubiquitination and degradation.

Conclusion: COLEC10 inhibits HCC stemness by downregulating the Wnt/β-catenin pathway, which is a promising target for liver CSC therapy.

背景:肝癌干细胞(CSCs)在肝细胞癌(HCC)的肿瘤发生、进展和复发中起着重要作用。Wnt/β-catenin通路在肝癌干细胞的形成、进展、转移和耐药性中起着至关重要的作用,但迄今为止还没有临床批准的药物能有效地靶向这一通路。我们旨在阐明COLEC10在HCC干性中的作用:方法:利用癌症基因组图谱(TCGA)和临床肿瘤蛋白质组学分析联盟(CPTAC)数据库搜索 COLEC10 表达与 HCC 干性之间的关联。研究人员利用集落形成、球形形成、侧群和极限稀释肿瘤启动试验来确定 COLEC10 过表达在 HCC 细胞系干性中的调控作用。研究人员还进行了Wnt/β-catenin报告实验和免疫沉淀以探索其潜在机制:结果:COLEC10水平与HCC干性呈负相关。COLEC10的升高导致EpCAM和AFP(甲胎蛋白)这两种肝脏造血干细胞常见标志物的表达降低。COLEC10的过表达抑制了HCC细胞形成集落和球体,减少了体外的侧群数量和体内的致瘤能力。从机理上讲,我们发现过表达COLEC10可通过上调Wnt抑制因子WIF1和降低胞质β-catenin水平来抑制Wnt/β-catenin信号转导的活性。COLEC10 的过表达促进了β-catenin 与破坏复合体 CK1α 的相互作用。此外,KLHL22(Kelch Like Family Member 22)是一种已报道的E3连接酶适配体,据预测可与CK1α相互作用,可促进COLEC10的单泛素化和降解:结论:COLEC10通过下调Wnt/β-catenin通路抑制HCC干细胞,是肝脏CSC治疗的一个有前景的靶点。
{"title":"COLEC10 inhibits the stemness of hepatocellular carcinoma by suppressing the activity of β-catenin signaling.","authors":"Mei-Na Cai, Dong-Mei Chen, Xin-Ru Chen, Yu-Rong Gu, Chun-Hong Liao, Le-Xin Xiao, Jia-Liang Wang, Bing-Liang Lin, Yue-Hua Huang, Yi-Fan Lian","doi":"10.1007/s13402-024-00972-4","DOIUrl":"10.1007/s13402-024-00972-4","url":null,"abstract":"<p><strong>Background: </strong>Liver cancer stem cells (CSCs) contribute to tumor initiation, progression, and recurrence in hepatocellular carcinoma (HCC). The Wnt/β-catenin pathway plays a crucial role in liver cancer stemness, progression, metastasis, and drug resistance, but no clinically approved drugs have targeted this pathway efficiently so far. We aimed to elucidate the role of COLEC10 in HCC stemness.</p><p><strong>Methods: </strong>The Cancer Genome Atlas (TCGA) and the Clinical Proteomic Tumor Analysis Consortium (CPTAC) databases were employed to search for the association between COLEC10 expression and HCC stemness. Colony formation, sphere formation, side population, and limiting dilution tumor initiation assays were used to identify the regulatory role of COLEC10 overexpression in the stemness of HCC cell lines. Wnt/β-catenin reporter assay and immunoprecipitation were performed to explore the underlying mechanism.</p><p><strong>Results: </strong>COLEC10 level was negatively correlated with HCC stemness. Elevated COLEC10 led to decreased expressions of EpCAM and AFP (alpha-fetoprotein), two common markers of liver CSCs. Overexpression of COLEC10 inhibited HCC cells from forming colonies and spheres, and reduced the side population numbers in vitro, as well as the tumorigenic capacity in vivo. Mechanically, we demonstrated that overexpression of COLEC10 suppressed the activity of Wnt/β-catenin signaling by upregulating Wnt inhibitory factor WIF1 and reducing the level of cytoplasmic β-catenin. COLEC10 overexpression promoted the interaction of β-catenin with the component of destruction complex CK1α. In addition, KLHL22 (Kelch Like Family Member 22), a reported E3 ligase adaptor predicted to interact with CK1α, could facilitate COLEC10 monoubiquitination and degradation.</p><p><strong>Conclusion: </strong>COLEC10 inhibits HCC stemness by downregulating the Wnt/β-catenin pathway, which is a promising target for liver CSC therapy.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141856901","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
TRIM28 promotes tumor growth and metastasis in breast cancer by targeting the BRD7 protein for ubiquitination and degradation. TRIM28 通过靶向 BRD7 蛋白进行泛素化和降解,促进乳腺癌的肿瘤生长和转移。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-10-01 Epub Date: 2024-09-02 DOI: 10.1007/s13402-024-00981-3
Changning Xue, Hanbing Meng, Weihong Niu, Mengna Li, Jianxia Wei, Shipeng Chen, Lemei Zheng, Yumei Duan, Hongyu Deng, Faqing Tang, Songqing Fan, Ming Tan, Wei Xiong, Ming Zhou
<p><strong>Purpose: </strong>Bromodomain-containing protein 7 (BRD7) is downregulated and functions as a tumor suppressor in many types of cancers including breast cancer, and the dysregulation of BRD7 expression is closely related to the development and progression of breast cancer. Whereas little attention has been focused on the regulation of BRD7 protein levels in breast cancer, which needs to be further elucidated.</p><p><strong>Methods: </strong>The protein stability of BRD7 in breast cancer cells and BRD7 protein level in breast cancer tissues was examined by Western Blotting. The potential E3 ubiquitin ligase proteins that interact with the BRD7 was screened by coimmunoprecipitation combined with mass spectrometry analysis in MDA-MB-231 cells. We proved the interaction between BRD7 and tripartite motif containing 28 (TRIM28) through Co-Immunoprecipitation (Co-IP) and immunofluorescence assays. Co-IP and ubiquitination assay were used to explore the specific binding domain between BRD7 and TRIM28 and the ubiquitination site of BRD7. The effects of TRIM28 on the BRD7 protein stability and ubiquitination level was investigated by qPCR, Western Blot and Co-IP assay. CCK-8 and clone formation assays were carried out to assess the effect of TRIM28 on proliferation ability of breast cancer ells. Transwell assay and wound healing assay were used to investigate the effect of TRIM28 on breast cancer cell invasion and migration. Flow cytometry was used to detect the effect of TRIM28 on cell cycle and apoptosis of breast cancer cells. In addition, we confirmed effect of TRIM28 on tumor growth and metastasis by xenograft and metastatic mouse models. We designed some recovery assays to explore the role of recovery BRD7 in TRIM28-mediated promotion of malignant progression of breast cancer in vivo and in vitro. Finally, the clinical significance of TRIM28 and BRD7 was proved by immunohistochemistry.</p><p><strong>Results: </strong>In this study, we demonstrated that BRD7 was an unstable protein and might be regulated by ubiquitination in breast cancer; furthermore, we found that the Coiled-Coil region of TRIM28 could directly bind to N-terminal of BRD7, and TRIM28 mediates BRD7 ubiquitination and degradation dependent on K21 by acting as a potential E3 ubiquitin ligase. Moreover, TRIM28 promoted cell proliferation, migration, invasion, xenograft tumor growth and metastasis, thus playing an oncogenic role in breast cancer. Furthermore, the restoration of BRD7 expression in breast cancer significantly reversed the promotional effects of TRIM28 on malignant progression both in vitro and in vivo. In addition, TRIM28 was highly expressed in the biopsy tissues of breast cancer, and its expression was negatively correlated with BRD7 expression and positively correlated with TNM stage and poor prognosis of BC patients.</p><p><strong>Conclusions: </strong>Our findings provide a novel mechanism by which TRIM28 significantly facilitates BRD7 ubiquitination and deg
目的:含溴结构域蛋白7(Bromodomain-containing protein 7,BRD7)在包括乳腺癌在内的多种癌症中下调并发挥肿瘤抑制因子的功能,BRD7表达失调与乳腺癌的发生和发展密切相关。然而,人们很少关注乳腺癌中BRD7蛋白水平的调控,这需要进一步阐明:方法:采用Western印迹法检测乳腺癌细胞中BRD7蛋白的稳定性和乳腺癌组织中BRD7蛋白的水平。在MDA-MB-231细胞中,通过免疫共沉淀结合质谱分析,筛选出可能与BRD7相互作用的E3泛素连接酶蛋白。我们通过共免疫沉淀(Co-IP)和免疫荧光试验证明了BRD7与含三方基序28(TRIM28)之间的相互作用。Co-IP和泛素化实验用于探索BRD7和TRIM28之间的特异性结合域以及BRD7的泛素化位点。通过qPCR、Western Blot和Co-IP实验研究了TRIM28对BRD7蛋白稳定性和泛素化水平的影响。CCK-8和克隆形成试验评估了TRIM28对乳腺癌细胞增殖能力的影响。Transwell试验和伤口愈合试验用于研究TRIM28对乳腺癌细胞侵袭和迁移的影响。流式细胞术检测了TRIM28对乳腺癌细胞周期和凋亡的影响。此外,我们还通过异种移植和转移小鼠模型证实了 TRIM28 对肿瘤生长和转移的影响。我们设计了一些复原实验来探讨复原 BRD7 在 TRIM28 介导的乳腺癌体内和体外恶性进展中的作用。最后,通过免疫组化证明了TRIM28和BRD7的临床意义:结果:本研究表明,BRD7是一种不稳定蛋白,在乳腺癌中可能受到泛素化的调控;此外,我们还发现TRIM28的Coil-Coil区域可直接与BRD7的N-端结合,TRIM28通过作为潜在的E3泛素连接酶,介导BRD7泛素化和降解,降解过程依赖于K21。此外,TRIM28还能促进细胞增殖、迁移、侵袭、异种移植肿瘤生长和转移,从而在乳腺癌中发挥致癌作用。此外,恢复乳腺癌中BRD7的表达可显著逆转TRIM28在体外和体内对恶性进展的促进作用。此外,TRIM28在乳腺癌活检组织中高表达,其表达与BRD7表达呈负相关,与BC患者的TNM分期和不良预后呈正相关:我们的研究结果提供了一种新的机制,即TRIM28能显著促进BRD7的泛素化和降解,从而促进乳腺癌的恶性进展。靶向TRIM28/BRD7轴可能是临床诊断和治疗乳腺癌的一种新的潜在策略。
{"title":"TRIM28 promotes tumor growth and metastasis in breast cancer by targeting the BRD7 protein for ubiquitination and degradation.","authors":"Changning Xue, Hanbing Meng, Weihong Niu, Mengna Li, Jianxia Wei, Shipeng Chen, Lemei Zheng, Yumei Duan, Hongyu Deng, Faqing Tang, Songqing Fan, Ming Tan, Wei Xiong, Ming Zhou","doi":"10.1007/s13402-024-00981-3","DOIUrl":"10.1007/s13402-024-00981-3","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Purpose: &lt;/strong&gt;Bromodomain-containing protein 7 (BRD7) is downregulated and functions as a tumor suppressor in many types of cancers including breast cancer, and the dysregulation of BRD7 expression is closely related to the development and progression of breast cancer. Whereas little attention has been focused on the regulation of BRD7 protein levels in breast cancer, which needs to be further elucidated.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Methods: &lt;/strong&gt;The protein stability of BRD7 in breast cancer cells and BRD7 protein level in breast cancer tissues was examined by Western Blotting. The potential E3 ubiquitin ligase proteins that interact with the BRD7 was screened by coimmunoprecipitation combined with mass spectrometry analysis in MDA-MB-231 cells. We proved the interaction between BRD7 and tripartite motif containing 28 (TRIM28) through Co-Immunoprecipitation (Co-IP) and immunofluorescence assays. Co-IP and ubiquitination assay were used to explore the specific binding domain between BRD7 and TRIM28 and the ubiquitination site of BRD7. The effects of TRIM28 on the BRD7 protein stability and ubiquitination level was investigated by qPCR, Western Blot and Co-IP assay. CCK-8 and clone formation assays were carried out to assess the effect of TRIM28 on proliferation ability of breast cancer ells. Transwell assay and wound healing assay were used to investigate the effect of TRIM28 on breast cancer cell invasion and migration. Flow cytometry was used to detect the effect of TRIM28 on cell cycle and apoptosis of breast cancer cells. In addition, we confirmed effect of TRIM28 on tumor growth and metastasis by xenograft and metastatic mouse models. We designed some recovery assays to explore the role of recovery BRD7 in TRIM28-mediated promotion of malignant progression of breast cancer in vivo and in vitro. Finally, the clinical significance of TRIM28 and BRD7 was proved by immunohistochemistry.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;In this study, we demonstrated that BRD7 was an unstable protein and might be regulated by ubiquitination in breast cancer; furthermore, we found that the Coiled-Coil region of TRIM28 could directly bind to N-terminal of BRD7, and TRIM28 mediates BRD7 ubiquitination and degradation dependent on K21 by acting as a potential E3 ubiquitin ligase. Moreover, TRIM28 promoted cell proliferation, migration, invasion, xenograft tumor growth and metastasis, thus playing an oncogenic role in breast cancer. Furthermore, the restoration of BRD7 expression in breast cancer significantly reversed the promotional effects of TRIM28 on malignant progression both in vitro and in vivo. In addition, TRIM28 was highly expressed in the biopsy tissues of breast cancer, and its expression was negatively correlated with BRD7 expression and positively correlated with TNM stage and poor prognosis of BC patients.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Conclusions: &lt;/strong&gt;Our findings provide a novel mechanism by which TRIM28 significantly facilitates BRD7 ubiquitination and deg","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142113994","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
PSMD2 overexpression as a biomarker for resistance and prognosis in renal cell carcinoma treated with immune checkpoint and tyrosine kinase inhibitors. PSMD2过表达作为免疫检查点和酪氨酸激酶抑制剂治疗肾细胞癌耐药性和预后的生物标记物。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-10-01 Epub Date: 2024-09-02 DOI: 10.1007/s13402-024-00977-z
Xianglai Xu, Jiahao Wang, Ying Wang, Yanjun Zhu, Jiajun Wang, Jianming Guo

Background: Integrated immune checkpoint inhibitors (ICIs) plus tyrosine kinase inhibitors (TKIs) are now the recommended first-line therapy to manage renal cell carcinoma (mRCC). Proteasome 26S subunit non-ATPase 2 (PSMD2) overexpression in tumors has been correlated with tumor progression. Currently, mRCC lacks an established biomarker for the combination of ICI+TKI.

Methods: This study involved RNA sequencing of RCC patients from two cohorts treated with ICI+TKI (ZS-MRCC and JAVELIN-Renal-101). We utilized immunohistochemistry alongside flow cytometry, aiming at assessing immune cell infiltration and functionality in high-risk localized RCC samples. Response and progression-free survival (PFS) were evaluated relying upon RECIST criteria.

Results: PSMD2 was significantly overexpressed in advanced RCC and among non-responders to ICI+TKI therapy. Overexpressed PSMD2 was correlated with poor PFS in the ZS-MRCC and JAVELIN-101 cohorts. Multivariate Cox analysis validated PSMD2 as an independent PFS predictor. PSMD2 overexpression was related to a reduction in CD8+ T cells, especially GZMB+ CD8+ T cells, besides an increase in PD1+ CD4+ T cells. Additionally, tumors with high PSMD2 levels showed enhanced T cell exhaustion levels and a higher regulatory T cell presence. A Machine Learning (ML) model based on PSMD2 expression and other screened factors was subsequently developed to predict the effectiveness of ICI+TKI.

Conclusions: Elevated PSMD2 expression is linked to resistance and decreased PFS in mRCC patients undergoing ICI+TKI therapy. High PSMD2 levels are also associated with impaired function and increased exhaustion of tumor-infiltrating lymphocytes. An ML model incorporating PSMD2 expression could potentially identify patients who may have a higher likelihood of benefiting from ICI+TKI.

背景:综合免疫检查点抑制剂(ICIs)加酪氨酸激酶抑制剂(TKIs)是目前治疗肾细胞癌(mRCC)的推荐一线疗法。肿瘤中蛋白酶体26S亚基非ATP酶2(PSMD2)的过表达与肿瘤的进展相关。目前,mRCC缺乏一种已确定的生物标志物,可用于ICI+TKI的联合治疗:本研究对两组接受 ICI+TKI 治疗的 RCC 患者(ZS-MRCC 和 JAVELIN-Renal-101)进行了 RNA 测序。我们在使用流式细胞术的同时还使用了免疫组化技术,旨在评估高风险局部RCC样本中的免疫细胞浸润和功能。根据 RECIST 标准评估了反应和无进展生存期(PFS):结果:PSMD2在晚期RCC和对ICI+TKI疗法无应答者中明显过表达。在ZS-MRCC和JAVELIN-101队列中,PSMD2的过表达与不良的PFS相关。多变量Cox分析验证了PSMD2是一个独立的PFS预测因子。PSMD2的过表达与CD8+ T细胞,尤其是GZMB+ CD8+ T细胞的减少有关,此外还与PD1+ CD4+ T细胞的增加有关。此外,PSMD2水平高的肿瘤显示出更高的T细胞衰竭水平和更高的调节性T细胞存在。基于PSMD2表达和其他筛选因素的机器学习(ML)模型随后被开发出来,用于预测ICI+TKI的有效性:结论:PSMD2表达升高与接受ICI+TKI治疗的mRCC患者的耐药性和PFS下降有关。高 PSMD2 水平还与肿瘤浸润淋巴细胞功能受损和耗竭增加有关。包含 PSMD2 表达的 ML 模型有可能识别出更有可能从 ICI+TKI 治疗中获益的患者。
{"title":"PSMD2 overexpression as a biomarker for resistance and prognosis in renal cell carcinoma treated with immune checkpoint and tyrosine kinase inhibitors.","authors":"Xianglai Xu, Jiahao Wang, Ying Wang, Yanjun Zhu, Jiajun Wang, Jianming Guo","doi":"10.1007/s13402-024-00977-z","DOIUrl":"10.1007/s13402-024-00977-z","url":null,"abstract":"<p><strong>Background: </strong>Integrated immune checkpoint inhibitors (ICIs) plus tyrosine kinase inhibitors (TKIs) are now the recommended first-line therapy to manage renal cell carcinoma (mRCC). Proteasome 26S subunit non-ATPase 2 (PSMD2) overexpression in tumors has been correlated with tumor progression. Currently, mRCC lacks an established biomarker for the combination of ICI+TKI.</p><p><strong>Methods: </strong>This study involved RNA sequencing of RCC patients from two cohorts treated with ICI+TKI (ZS-MRCC and JAVELIN-Renal-101). We utilized immunohistochemistry alongside flow cytometry, aiming at assessing immune cell infiltration and functionality in high-risk localized RCC samples. Response and progression-free survival (PFS) were evaluated relying upon RECIST criteria.</p><p><strong>Results: </strong>PSMD2 was significantly overexpressed in advanced RCC and among non-responders to ICI+TKI therapy. Overexpressed PSMD2 was correlated with poor PFS in the ZS-MRCC and JAVELIN-101 cohorts. Multivariate Cox analysis validated PSMD2 as an independent PFS predictor. PSMD2 overexpression was related to a reduction in CD8<sup>+</sup> T cells, especially GZMB<sup>+</sup> CD8<sup>+</sup> T cells, besides an increase in PD1<sup>+</sup> CD4<sup>+</sup> T cells. Additionally, tumors with high PSMD2 levels showed enhanced T cell exhaustion levels and a higher regulatory T cell presence. A Machine Learning (ML) model based on PSMD2 expression and other screened factors was subsequently developed to predict the effectiveness of ICI+TKI.</p><p><strong>Conclusions: </strong>Elevated PSMD2 expression is linked to resistance and decreased PFS in mRCC patients undergoing ICI+TKI therapy. High PSMD2 levels are also associated with impaired function and increased exhaustion of tumor-infiltrating lymphocytes. An ML model incorporating PSMD2 expression could potentially identify patients who may have a higher likelihood of benefiting from ICI+TKI.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142113976","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Cellular 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