首页 > 最新文献

Cellular Oncology最新文献

英文 中文
TRPV1 inhibition suppresses non-small cell lung cancer progression by inhibiting tumour growth and enhancing the immune response. TRPV1抑制通过抑制肿瘤生长和增强免疫反应来抑制非小细胞肺癌癌症的进展。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-06-01 Epub Date: 2023-10-30 DOI: 10.1007/s13402-023-00894-7
Yang Wang, Yu Zhang, Jing Ouyang, Hanying Yi, Shiyu Wang, Dongbo Liu, Yingying Dai, Kun Song, Wenwu Pei, Ziyang Hong, Ling Chen, Wei Zhang, Zhaoqian Liu, Howard L Mcleod, Yijing He

Purpose: TRPV1 is a nonselective Ca2+ channel protein that is widely expressed and plays an important role during the occurrence and development of many cancers. Activation of TRPV1 channels can affect tumour progression by regulating proliferation, apoptosis and migration. Some studies have also shown that activating TRPV1 can affect tumour progression by modulating tumour immunity. However, the effects of TRPV1 on the development of non-small cell lung cancer (NSCLC) have not been explored clearly.

Method: The Cancer Genome Atlas (TCGA) database and spatial transcriptomics datasets from 10 × Genomics were used to analyze TRPV1 expression in various tumour tissues. Cell proliferation and apoptosis were examined by cell counting kit 8 (CCK8), colony formation, and flow cytometry. Immunohistochemistry, qPCR, and western blotting were used to determine the mRNA and protein expression levels of TRPV1 and other related molecules. Tumour xenografts in BALB/C and C57BL/6J mice were used to determine the effects of TRPV1 on NSCLC development in vivo. Neurotransmitter content was examined by LC-MS/MS, ELISA and Immunohistochemistry. Immune cell infiltration was assessed by flow cytometry.

Results: In this study, we found that TRPV1 expression was significantly upregulated in NSCLC and that patients with high TRPV1 expression had a poor prognosis. TRPV1 knockdown can significantly inhibit NSCLC proliferation and induce cell apoptosis through Ca2+-IGF1R signaling. In addition, TRPV1 knockdown resulted in increased infiltration of CD4+ T cells, CD8+ T cells, GZMB+CD8+ T cells and DCs and decreased infiltration of immunosuppressive MDSCs in NSCLC. In addition, TRPV1 knockout effectively decreased the expression of M2 macrophage markers CD163 and increased the expression of M1-associated, costimulatory markers CD86. Knockdown or knockout of TRPV1 significantly inhibit tumour growth and promoted an antitumour immune response through supressing γ-aminobutyric acid (GABA) secretion in NSCLC.

Conclusion: Our study suggests that TRPV1 acts as a tumour promoter in NSCLC, mediating pro-proliferative and anti-apoptotic effects on NSCLC through IGF1R signaling and regulating GABA release to affect the tumour immune response.

目的:TRPV1是一种广泛表达的非选择性Ca2+通道蛋白,在许多癌症的发生和发展过程中发挥着重要作用。TRPV1通道的激活可以通过调节增殖、凋亡和迁移来影响肿瘤的进展。一些研究还表明,激活TRPV1可以通过调节肿瘤免疫来影响肿瘤进展。然而,TRPV1对癌症(NSCLC)发展的影响尚不清楚。方法:利用癌症基因组图谱(TCGA)数据库和10×基因组的空间转录组学数据集分析TRPV1在各种肿瘤组织中的表达。通过细胞计数试剂盒8(CCK8)、集落形成和流式细胞术检测细胞增殖和凋亡。免疫组织化学、qPCR和蛋白质印迹用于测定TRPV1和其他相关分子的mRNA和蛋白质表达水平。使用BALB/C和C57BL/6J小鼠中的肿瘤异种移植物来确定TRPV1对体内NSCLC发展的影响。采用LC-MS/MS、ELISA和免疫组织化学方法检测神经递质含量。通过流式细胞术评估免疫细胞浸润。结果:在本研究中,我们发现TRPV1在NSCLC中的表达显著上调,TRPV1高表达的患者预后较差。TRPV1敲低可通过Ca2+-IGF1R信号传导显著抑制NSCLC增殖并诱导细胞凋亡。此外,TRPV1敲低导致NSCLC中CD4+T细胞、CD8+T细胞,GZMB+CD8+T细胞和DC的浸润增加,以及免疫抑制性MDSCs的浸润减少。此外,TRPV1敲除有效降低了M2巨噬细胞标志物CD163的表达,并增加了M1相关共刺激标志物CD86的表达。敲除或敲除TRPV1可显著抑制肿瘤生长,并通过抑制NSCLC中γ-氨基丁酸(GABA)的分泌促进抗肿瘤免疫反应。结论:我们的研究表明,TRPV1在NSCLC中起肿瘤启动子的作用,通过IGF1R信号传导介导NSCLC的促增殖和抗凋亡作用,并调节GABA的释放以影响肿瘤免疫反应。
{"title":"TRPV1 inhibition suppresses non-small cell lung cancer progression by inhibiting tumour growth and enhancing the immune response.","authors":"Yang Wang, Yu Zhang, Jing Ouyang, Hanying Yi, Shiyu Wang, Dongbo Liu, Yingying Dai, Kun Song, Wenwu Pei, Ziyang Hong, Ling Chen, Wei Zhang, Zhaoqian Liu, Howard L Mcleod, Yijing He","doi":"10.1007/s13402-023-00894-7","DOIUrl":"10.1007/s13402-023-00894-7","url":null,"abstract":"<p><strong>Purpose: </strong>TRPV1 is a nonselective Ca<sup>2+</sup> channel protein that is widely expressed and plays an important role during the occurrence and development of many cancers. Activation of TRPV1 channels can affect tumour progression by regulating proliferation, apoptosis and migration. Some studies have also shown that activating TRPV1 can affect tumour progression by modulating tumour immunity. However, the effects of TRPV1 on the development of non-small cell lung cancer (NSCLC) have not been explored clearly.</p><p><strong>Method: </strong>The Cancer Genome Atlas (TCGA) database and spatial transcriptomics datasets from 10 × Genomics were used to analyze TRPV1 expression in various tumour tissues. Cell proliferation and apoptosis were examined by cell counting kit 8 (CCK8), colony formation, and flow cytometry. Immunohistochemistry, qPCR, and western blotting were used to determine the mRNA and protein expression levels of TRPV1 and other related molecules. Tumour xenografts in BALB/C and C57BL/6J mice were used to determine the effects of TRPV1 on NSCLC development in vivo. Neurotransmitter content was examined by LC-MS/MS, ELISA and Immunohistochemistry. Immune cell infiltration was assessed by flow cytometry.</p><p><strong>Results: </strong>In this study, we found that TRPV1 expression was significantly upregulated in NSCLC and that patients with high TRPV1 expression had a poor prognosis. TRPV1 knockdown can significantly inhibit NSCLC proliferation and induce cell apoptosis through Ca<sup>2+</sup>-IGF1R signaling. In addition, TRPV1 knockdown resulted in increased infiltration of CD4<sup>+</sup> T cells, CD8<sup>+</sup> T cells, GZMB<sup>+</sup>CD8<sup>+</sup> T cells and DCs and decreased infiltration of immunosuppressive MDSCs in NSCLC. In addition, TRPV1 knockout effectively decreased the expression of M2 macrophage markers CD163 and increased the expression of M1-associated, costimulatory markers CD86. Knockdown or knockout of TRPV1 significantly inhibit tumour growth and promoted an antitumour immune response through supressing γ-aminobutyric acid (GABA) secretion in NSCLC.</p><p><strong>Conclusion: </strong>Our study suggests that TRPV1 acts as a tumour promoter in NSCLC, mediating pro-proliferative and anti-apoptotic effects on NSCLC through IGF1R signaling and regulating GABA release to affect the tumour immune response.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"71414929","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
Asymmetric post-translational modifications regulate the nuclear translocation of STAT3 homodimers in response to leukemia inhibitory factor. 非对称翻译后修饰调节 STAT3 同源二聚体在白血病抑制因子作用下的核转位
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-06-01 Epub Date: 2023-12-27 DOI: 10.1007/s13402-023-00911-9
Mickael Diallo, Constança Pimenta, Fernanda Murtinheira, Daniela Martins-Alves, Francisco R Pinto, André Abrantes da Costa, Ricardo Letra-Vilela, Vanesa Martin, Carmen Rodriguez, Mário S Rodrigues, Federico Herrera

STAT3 is a pleiotropic transcription factor overactivated in 70% of solid tumours. We have recently reported that inactivating mutations on residues susceptible to post-translational modifications (PTMs) in only one of the monomers (i.e. asymmetric) caused changes in the cellular distribution of STAT3 homodimers. Here, we used more controlled experimental conditions, i.e. without the interference of endogenous STAT3 (STAT3-/- HeLa cells) and in the presence of a defined cytokine stimulus (Leukemia Inhibitory Factor, LIF), to provide further evidence that asymmetric PTMs affect the nuclear translocation of STAT3 homodimers. Time-lapse microscopy for 20 min after LIF stimulation showed that S727 dephosphorylation (S727A) and K685 inactivation (K685R) slightly enhanced the nuclear translocation of STAT3 homodimers, while K49 inactivation (K49R) delayed STAT3 nuclear translocation. Our findings suggest that asymmetrically modified STAT3 homodimers could be a new level of STAT3 regulation and, therefore, a potential target for cancer therapy.

STAT3是一种多效应转录因子,在70%的实体瘤中被过度激活。我们最近报告说,仅对其中一个单体(即非对称单体)中易受翻译后修饰(PTMs)影响的残基进行失活突变会导致 STAT3 同源二聚体的细胞分布发生变化。在这里,我们采用了更可控的实验条件,即在没有内源性 STAT3(STAT3-/- HeLa 细胞)干扰和有明确细胞因子刺激(白血病抑制因子,LIF)的情况下,进一步证明了不对称 PTM 会影响 STAT3 同源二聚体的核转位。LIF 刺激后 20 分钟的延时显微镜观察表明,S727 去磷酸化(S727A)和 K685 失活(K685R)会轻微增强 STAT3 同源二聚体的核转位,而 K49 失活(K49R)会延迟 STAT3 的核转位。我们的研究结果表明,不对称修饰的STAT3同源二聚体可能是STAT3调控的一个新水平,因此是癌症治疗的一个潜在靶点。
{"title":"Asymmetric post-translational modifications regulate the nuclear translocation of STAT3 homodimers in response to leukemia inhibitory factor.","authors":"Mickael Diallo, Constança Pimenta, Fernanda Murtinheira, Daniela Martins-Alves, Francisco R Pinto, André Abrantes da Costa, Ricardo Letra-Vilela, Vanesa Martin, Carmen Rodriguez, Mário S Rodrigues, Federico Herrera","doi":"10.1007/s13402-023-00911-9","DOIUrl":"10.1007/s13402-023-00911-9","url":null,"abstract":"<p><p>STAT3 is a pleiotropic transcription factor overactivated in 70% of solid tumours. We have recently reported that inactivating mutations on residues susceptible to post-translational modifications (PTMs) in only one of the monomers (i.e. asymmetric) caused changes in the cellular distribution of STAT3 homodimers. Here, we used more controlled experimental conditions, i.e. without the interference of endogenous STAT3 (STAT3-/- HeLa cells) and in the presence of a defined cytokine stimulus (Leukemia Inhibitory Factor, LIF), to provide further evidence that asymmetric PTMs affect the nuclear translocation of STAT3 homodimers. Time-lapse microscopy for 20 min after LIF stimulation showed that S727 dephosphorylation (S727A) and K685 inactivation (K685R) slightly enhanced the nuclear translocation of STAT3 homodimers, while K49 inactivation (K49R) delayed STAT3 nuclear translocation. Our findings suggest that asymmetrically modified STAT3 homodimers could be a new level of STAT3 regulation and, therefore, a potential target for cancer therapy.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11219437/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139040779","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
ITGB2-ICAM1 axis promotes liver metastasis in BAP1-mutated uveal melanoma with retained hypoxia and ECM signatures. ITGB2-ICAM1轴促进BAP1突变葡萄膜黑色素瘤的肝转移,并保留缺氧和ECM特征。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-06-01 Epub Date: 2023-12-27 DOI: 10.1007/s13402-023-00908-4
Jiaoduan Li, Dongyan Cao, Lixin Jiang, Yiwen Zheng, Siyuan Shao, Ai Zhuang, Dongxi Xiang

Purpose: Uveal melanoma (UM) with BAP1 inactivating mutations has a high risk of metastasis, but the mechanism behind BAP1 deficiency driving UM metastasis is unknown.

Methods: We analyzed the single-cell RNA sequencing (scRNA-Seq) data comprised primary and metastatic UM with or without BAP1 mutations (MUTs) to reveal inter- and intra-tumor heterogeneity among different groups. Then, an immune-competent mouse liver metastatic model was used to explore the role of ITGB2-ICAM1 in BAP1-associated UM metastasis.

Results: Cluster 1 tumor cells expressed high levels of genes linked to tumor metastasis, such as GDF15, ATF3, and CDKN1A, all of which are associated with poor prognosis. The strength of communication between terminally exhausted CD8+ T cells and GDF15hiATF3hiCDKN1Ahi tumor cells was enhanced in BAP1-mutated UM, with CellChat analysis predicting strong ITGB2-ICAM1 signaling between them. High expression of either ITGB2 or ICAM1 was a worse prognostic indicator. Using an immune-competent mouse liver metastatic model, we indicated that inhibiting either ICAM1 or ITGB2 prevented liver metastasis in the BAP1-mutated group in vivo. The inhibitors primarily inhibited hypoxia- and ECM-related pathways indicated by changes in the expression of genes such as ADAM8, CAV2, ENO1, PGK1, LOXL2, ITGA5, and VCAN. etc. CONCLUSION: This study suggested that the ITGB2-ICAM1 axis may play a crucial role for BAP1-associated UM metastasis by preserving hypoxia- and ECM- related signatures, which provide a potential strategy for preventing UM metastasis in patients with BAP1 mutation.

目的:存在BAP1失活突变的葡萄膜黑色素瘤(UM)具有很高的转移风险,但BAP1缺失导致UM转移的机制尚不清楚:我们分析了单细胞RNA测序(scRNA-Seq)数据,包括有或没有BAP1突变(MUTs)的原发性和转移性UM,以揭示不同组间和肿瘤内部的异质性。然后,利用免疫功能正常的小鼠肝转移模型来探讨ITGB2-ICAM1在BAP1相关UM转移中的作用:结果:第1组肿瘤细胞表达了高水平的与肿瘤转移相关的基因,如GDF15、ATF3和CDKN1A,所有这些基因都与不良预后相关。在BAP1突变的UM中,终末衰竭的CD8+ T细胞与GDF15hiATF3hiCDKN1Ahi肿瘤细胞之间的通讯强度增强,CellChat分析预测它们之间有很强的ITGB2-ICAM1信号传导。ITGB2或ICAM1的高表达是一个较差的预后指标。我们利用免疫功能健全的小鼠肝转移模型表明,抑制ICAM1或ITGB2可阻止BAP1突变组的体内肝转移。抑制剂主要抑制缺氧和 ECM 相关通路,表现为 ADAM8、CAV2、ENO1、PGK1、LOXL2、ITGA5 和 VCAN 等基因表达的变化。结论:该研究表明,ITGB2-ICAM1 轴可能通过保留缺氧和 ECM 相关特征在 BAP1 相关 UM 转移中发挥关键作用,这为预防 BAP1 突变患者的 UM 转移提供了潜在策略。
{"title":"ITGB2-ICAM1 axis promotes liver metastasis in BAP1-mutated uveal melanoma with retained hypoxia and ECM signatures.","authors":"Jiaoduan Li, Dongyan Cao, Lixin Jiang, Yiwen Zheng, Siyuan Shao, Ai Zhuang, Dongxi Xiang","doi":"10.1007/s13402-023-00908-4","DOIUrl":"10.1007/s13402-023-00908-4","url":null,"abstract":"<p><strong>Purpose: </strong>Uveal melanoma (UM) with BAP1 inactivating mutations has a high risk of metastasis, but the mechanism behind BAP1 deficiency driving UM metastasis is unknown.</p><p><strong>Methods: </strong>We analyzed the single-cell RNA sequencing (scRNA-Seq) data comprised primary and metastatic UM with or without BAP1 mutations (MUTs) to reveal inter- and intra-tumor heterogeneity among different groups. Then, an immune-competent mouse liver metastatic model was used to explore the role of ITGB2-ICAM1 in BAP1-associated UM metastasis.</p><p><strong>Results: </strong>Cluster 1 tumor cells expressed high levels of genes linked to tumor metastasis, such as GDF15, ATF3, and CDKN1A, all of which are associated with poor prognosis. The strength of communication between terminally exhausted CD8<sup>+</sup> T cells and GDF15<sup>hi</sup>ATF3<sup>hi</sup>CDKN1A<sup>hi</sup> tumor cells was enhanced in BAP1-mutated UM, with CellChat analysis predicting strong ITGB2-ICAM1 signaling between them. High expression of either ITGB2 or ICAM1 was a worse prognostic indicator. Using an immune-competent mouse liver metastatic model, we indicated that inhibiting either ICAM1 or ITGB2 prevented liver metastasis in the BAP1-mutated group in vivo. The inhibitors primarily inhibited hypoxia- and ECM-related pathways indicated by changes in the expression of genes such as ADAM8, CAV2, ENO1, PGK1, LOXL2, ITGA5, and VCAN. etc. CONCLUSION: This study suggested that the ITGB2-ICAM1 axis may play a crucial role for BAP1-associated UM metastasis by preserving hypoxia- and ECM- related signatures, which provide a potential strategy for preventing UM metastasis in patients with BAP1 mutation.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139040780","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
Stiffened tumor microenvironment enhances perineural invasion in breast cancer via integrin signaling. 强化肿瘤微环境通过整合素信号增强乳腺癌的神经浸润。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-06-01 Epub Date: 2023-11-28 DOI: 10.1007/s13402-023-00901-x
Bing Han, Xin Guan, Mingyue Ma, Baoling Liang, Linglie Ren, Yutong Liu, Ye Du, Shu-Heng Jiang, Dong Song

Background: Accumulating studies have shown that tumors are regulated by nerves, and there is abundant nerve infiltration in the tumor microenvironment. Many solid tumors including breast cancer (BRCA) have different degrees of perineural invasion (PNI), which is closely related to the tumor occurrence and progression. However, the regulatory mechanism of PNI in BRCA remains largely unexplored.

Methods: PNI-related molecular events are analyzed by the RNAseq data of BRCA samples deposited in The Cancer Genome Atlas (TCGA) database. Extracellular matrix (ECM) components within the tumor microenvironment are analyzed by immunohistochemical staining of α-SMA, Sirius red staining, and Masson trichrome staining. Soft and stiff matrix gels, living cell imaging, and dorsal root ganglion (DRG) coculture assay are used to monitor cancer cell invasiveness towards nerves. Western blotting, qRT-PCR, enzyme-linked immunosorbent assay combined with neutralizing antibody and small molecular inhibitors are employed to decode molecular mechanisms.

Results: Comparative analysis that the ECM was significantly associated with PNI status in the TCGA cohort. BRCA samples with higher α-SMA activity, fibrillar collagen, and collagen content had higher frequency of PNI. Compared with soft matrix, BRCA cells cultured in stiff matrix not only displayed higher cell invasiveness to DRG neurons but also had significant neurotrophic effects. Mechanistically, integrin β1 was identified as a functional receptor to the influence of stiff matrix on BRCA cells. Moreover, stiffened matrix-induced activation of integrin β1 transduces FAK-YAP signal cascade, which enhances cancer invasiveness and the neurotrophic effects. In clinical setting, PNI-positive BRCA samples had higher expression of ITGB1, phosphorylated FAK, YAP, and NGF compared with PNI-negative BRCA samples.

Conclusions: Our findings suggest that stiff matrix induces expression of pro-metastatic and neurotrophic genes through integrin β1-FAK-YAP signals, which finally facilitates PNI in BRCA. Thus, our study provides a new mechanism for PNI in BRCA and highlights nerve-based tumor treatment strategies.

背景:越来越多的研究表明,肿瘤受神经调控,肿瘤微环境中存在丰富的神经浸润。包括乳腺癌(BRCA)在内的许多实体肿瘤都存在不同程度的神经周围浸润(PNI),这与肿瘤的发生和发展密切相关。然而,PNI在BRCA中的调控机制在很大程度上仍未被探索。方法:通过保存在the Cancer Genome Atlas (TCGA)数据库中的BRCA样本RNAseq数据,分析pni相关分子事件。采用免疫组化α-SMA染色、Sirius红染色和Masson三色染色分析肿瘤微环境中细胞外基质(Extracellular matrix, ECM)成分。采用软硬基质凝胶、活细胞成像和背根神经节(DRG)共培养法监测癌细胞对神经的侵袭。采用Western blotting、qRT-PCR、酶联免疫吸附法联合中和抗体和小分子抑制剂解码分子机制。结果:比较分析显示,TCGA队列中ECM与PNI状态显著相关。α-SMA活性高、纤维性胶原蛋白含量高、胶原蛋白含量高的BRCA样品PNI发生率较高。与软基质相比,硬基质中培养的BRCA细胞不仅对DRG神经元具有更高的细胞侵袭性,而且具有显著的神经营养作用。从机制上说,整合素β1被认为是一种功能受体,可以应对硬基质对BRCA细胞的影响。此外,强化基质诱导的整合素β1激活可转导FAK-YAP信号级联,从而增强肿瘤侵袭性和神经营养效应。在临床环境中,与pni阴性的BRCA样本相比,pni阳性的BRCA样本中ITGB1、磷酸化FAK、YAP和NGF的表达更高。结论:我们的研究结果表明,硬基质通过整合素β1-FAK-YAP信号诱导促转移基因和神经营养基因的表达,最终促进BRCA的PNI。因此,我们的研究提供了PNI在BRCA中的新机制,并强调了基于神经的肿瘤治疗策略。
{"title":"Stiffened tumor microenvironment enhances perineural invasion in breast cancer via integrin signaling.","authors":"Bing Han, Xin Guan, Mingyue Ma, Baoling Liang, Linglie Ren, Yutong Liu, Ye Du, Shu-Heng Jiang, Dong Song","doi":"10.1007/s13402-023-00901-x","DOIUrl":"10.1007/s13402-023-00901-x","url":null,"abstract":"<p><strong>Background: </strong>Accumulating studies have shown that tumors are regulated by nerves, and there is abundant nerve infiltration in the tumor microenvironment. Many solid tumors including breast cancer (BRCA) have different degrees of perineural invasion (PNI), which is closely related to the tumor occurrence and progression. However, the regulatory mechanism of PNI in BRCA remains largely unexplored.</p><p><strong>Methods: </strong>PNI-related molecular events are analyzed by the RNAseq data of BRCA samples deposited in The Cancer Genome Atlas (TCGA) database. Extracellular matrix (ECM) components within the tumor microenvironment are analyzed by immunohistochemical staining of α-SMA, Sirius red staining, and Masson trichrome staining. Soft and stiff matrix gels, living cell imaging, and dorsal root ganglion (DRG) coculture assay are used to monitor cancer cell invasiveness towards nerves. Western blotting, qRT-PCR, enzyme-linked immunosorbent assay combined with neutralizing antibody and small molecular inhibitors are employed to decode molecular mechanisms.</p><p><strong>Results: </strong>Comparative analysis that the ECM was significantly associated with PNI status in the TCGA cohort. BRCA samples with higher α-SMA activity, fibrillar collagen, and collagen content had higher frequency of PNI. Compared with soft matrix, BRCA cells cultured in stiff matrix not only displayed higher cell invasiveness to DRG neurons but also had significant neurotrophic effects. Mechanistically, integrin β1 was identified as a functional receptor to the influence of stiff matrix on BRCA cells. Moreover, stiffened matrix-induced activation of integrin β1 transduces FAK-YAP signal cascade, which enhances cancer invasiveness and the neurotrophic effects. In clinical setting, PNI-positive BRCA samples had higher expression of ITGB1, phosphorylated FAK, YAP, and NGF compared with PNI-negative BRCA samples.</p><p><strong>Conclusions: </strong>Our findings suggest that stiff matrix induces expression of pro-metastatic and neurotrophic genes through integrin β1-FAK-YAP signals, which finally facilitates PNI in BRCA. Thus, our study provides a new mechanism for PNI in BRCA and highlights nerve-based tumor treatment strategies.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138446721","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
OTUB1 accelerates hepatocellular carcinoma by stabilizing RACK1 via its non-canonical ubiquitination. OTUB1 通过非规范泛素化稳定 RACK1,从而加速肝细胞癌的发生。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-06-01 Epub Date: 2024-02-05 DOI: 10.1007/s13402-023-00913-7
Liqun Peng, Tiangen Wu, Yingyi Liu, Dongli Zhao, Wenzhi He, Yufeng Yuan

Background: Dysregulated ubiquitination modification occupies a pivotal role in hepatocellular carcinoma (HCC) tumorigenesis and progression. The ubiquitin aldehyde binding 1 (OTUB1) was aberrantly upregulated and exhibited the pro-tumorigenic function in HCC. However, the underlying mechanisms and responsible targets of OTUB1 remain unclear.

Methods: First, bioinformatics analysis, western blot and immunohistochemistry staining were applied to analyze OTUB1 expression in HCC specimens. Then, immunoprecipitation assay-tandem mass spectrometry (MS) combined with the gene set enrichment analysis (GSEA) was used to explore the downstream target of OTUB1. Co-immunoprecipitation and ubiquitination assays were used to identify the mechanisms involved. Finally, we explored the regulatory effect of MAZ on OTUB1 through ChIP-qPCR and dual-luciferase reporter assay.

Results: OTUB1 was broadly elevated in HCC tissues and promoted the proliferation and metastasis of HCC in vitro and in vivo. The receptor for activated C kinase 1 (RACK1) performed as a functional partner of OTUB1 and its hyperactivation was associated with aggressive development and other malignant features in HCC by activating oncogenes transcription. Mechanistically, OTUB1 directly bound to RACK1 at its C-terminal domain and decreased the K48-linked ubiquitination of RACK1 through its non-canonical suppression of ubiquitination activity, which stabilized RACK1 protein levels in HCC cells. Therefore, OTUB1 significantly increased multiple oncogenes expression and activated PI3K/AKT and FAK/ERK signaling in a RACK1-dependent manner in HCC. Moreover, the transcription factor MAZ upregulated OTUB1 expression through identifying a putative response element of OTUB1 promoter area.

Conclusions: Our findings might provide a new therapeutic strategy for HCC by modifying the MAZ-OTUB1-RACK1 axis.

背景:泛素化修饰失调在肝细胞癌(HCC)肿瘤发生和发展过程中起着关键作用。泛素醛结合1(OTUB1)在HCC中异常上调并表现出促癌功能。然而,OTUB1的潜在机制和作用靶点仍不清楚:方法:首先,应用生物信息学分析、Western 印迹和免疫组化染色分析 OTUB1 在 HCC 标本中的表达。然后,利用免疫沉淀分析-串联质谱(MS)结合基因组富集分析(GSEA)探讨OTUB1的下游靶标。通过共免疫沉淀和泛素化实验来确定其中的机制。最后,我们通过 ChIP-qPCR 和双荧光素酶报告实验探讨了 MAZ 对 OTUB1 的调控作用:结果:OTUB1在HCC组织中广泛升高,并在体外和体内促进了HCC的增殖和转移。活化C激酶1受体(RACK1)是OTUB1的功能伙伴,它的过度激活与HCC的侵袭性发展和其他恶性特征有关,因为它激活了癌基因的转录。从机理上讲,OTUB1直接与RACK1的C-末端结构域结合,并通过其非经典的泛素化活性抑制作用减少了RACK1与K48连接的泛素化,从而稳定了HCC细胞中RACK1蛋白的水平。因此,OTUB1 能显著增加 HCC 中多种癌基因的表达,并以 RACK1 依赖性方式激活 PI3K/AKT 和 FAK/ERK 信号转导。此外,转录因子MAZ通过识别OTUB1启动子区域的推定反应元件上调OTUB1的表达:我们的发现可能会通过改变 MAZ-OTUB1-RACK1 轴为 HCC 提供一种新的治疗策略。
{"title":"OTUB1 accelerates hepatocellular carcinoma by stabilizing RACK1 via its non-canonical ubiquitination.","authors":"Liqun Peng, Tiangen Wu, Yingyi Liu, Dongli Zhao, Wenzhi He, Yufeng Yuan","doi":"10.1007/s13402-023-00913-7","DOIUrl":"10.1007/s13402-023-00913-7","url":null,"abstract":"<p><strong>Background: </strong>Dysregulated ubiquitination modification occupies a pivotal role in hepatocellular carcinoma (HCC) tumorigenesis and progression. The ubiquitin aldehyde binding 1 (OTUB1) was aberrantly upregulated and exhibited the pro-tumorigenic function in HCC. However, the underlying mechanisms and responsible targets of OTUB1 remain unclear.</p><p><strong>Methods: </strong>First, bioinformatics analysis, western blot and immunohistochemistry staining were applied to analyze OTUB1 expression in HCC specimens. Then, immunoprecipitation assay-tandem mass spectrometry (MS) combined with the gene set enrichment analysis (GSEA) was used to explore the downstream target of OTUB1. Co-immunoprecipitation and ubiquitination assays were used to identify the mechanisms involved. Finally, we explored the regulatory effect of MAZ on OTUB1 through ChIP-qPCR and dual-luciferase reporter assay.</p><p><strong>Results: </strong>OTUB1 was broadly elevated in HCC tissues and promoted the proliferation and metastasis of HCC in vitro and in vivo. The receptor for activated C kinase 1 (RACK1) performed as a functional partner of OTUB1 and its hyperactivation was associated with aggressive development and other malignant features in HCC by activating oncogenes transcription. Mechanistically, OTUB1 directly bound to RACK1 at its C-terminal domain and decreased the K48-linked ubiquitination of RACK1 through its non-canonical suppression of ubiquitination activity, which stabilized RACK1 protein levels in HCC cells. Therefore, OTUB1 significantly increased multiple oncogenes expression and activated PI3K/AKT and FAK/ERK signaling in a RACK1-dependent manner in HCC. Moreover, the transcription factor MAZ upregulated OTUB1 expression through identifying a putative response element of OTUB1 promoter area.</p><p><strong>Conclusions: </strong>Our findings might provide a new therapeutic strategy for HCC by modifying the MAZ-OTUB1-RACK1 axis.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11219430/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139693350","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
NK cells as powerful therapeutic tool in cancer immunotherapy. NK 细胞是癌症免疫疗法的强大治疗工具。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-06-01 Epub Date: 2024-01-03 DOI: 10.1007/s13402-023-00909-3
Mao Huang, Yixuan Liu, Qijia Yan, Miao Peng, Junshang Ge, Yongzhen Mo, Yumin Wang, Fuyan Wang, Zhaoyang Zeng, Yong Li, Chunmei Fan, Wei Xiong

Background: Natural killer (NK) cells have gained considerable attention and hold great potential for their application in tumor immunotherapy. This is mainly due to their MHC-unrestricted and pan-specific recognition capabilities, as well as their ability to rapidly respond to and eliminate target cells. To artificially generate therapeutic NK cells, various materials can be utilized, such as peripheral blood mononuclear cells (PBMCs), umbilical cord blood (UCB), induced pluripotent stem cells (iPSCs), and NK cell lines. Exploiting the therapeutic potential of NK cells to treat tumors through in vivo and in vitro therapeutic modalities has yielded positive therapeutic results.

Conclusion: This review provides a comprehensive description of NK cell therapeutic approaches for tumors and discusses the current problems associated with these therapeutic approaches and the prospects of NK cell therapy for tumors.

背景:自然杀伤(NK)细胞在肿瘤免疫疗法中的应用受到了广泛关注,并具有巨大潜力。这主要归功于它们不受 MHC 限制的泛特异性识别能力,以及对靶细胞做出快速反应并将其消灭的能力。要人工生成治疗性 NK 细胞,可以利用多种材料,如外周血单核细胞(PBMC)、脐带血(UCB)、诱导多能干细胞(iPSC)和 NK 细胞系。通过体内和体外治疗模式挖掘 NK 细胞治疗肿瘤的潜力已取得了积极的治疗效果:本综述全面介绍了 NK 细胞治疗肿瘤的方法,并讨论了目前与这些治疗方法相关的问题以及 NK 细胞治疗肿瘤的前景。
{"title":"NK cells as powerful therapeutic tool in cancer immunotherapy.","authors":"Mao Huang, Yixuan Liu, Qijia Yan, Miao Peng, Junshang Ge, Yongzhen Mo, Yumin Wang, Fuyan Wang, Zhaoyang Zeng, Yong Li, Chunmei Fan, Wei Xiong","doi":"10.1007/s13402-023-00909-3","DOIUrl":"10.1007/s13402-023-00909-3","url":null,"abstract":"<p><strong>Background: </strong>Natural killer (NK) cells have gained considerable attention and hold great potential for their application in tumor immunotherapy. This is mainly due to their MHC-unrestricted and pan-specific recognition capabilities, as well as their ability to rapidly respond to and eliminate target cells. To artificially generate therapeutic NK cells, various materials can be utilized, such as peripheral blood mononuclear cells (PBMCs), umbilical cord blood (UCB), induced pluripotent stem cells (iPSCs), and NK cell lines. Exploiting the therapeutic potential of NK cells to treat tumors through in vivo and in vitro therapeutic modalities has yielded positive therapeutic results.</p><p><strong>Conclusion: </strong>This review provides a comprehensive description of NK cell therapeutic approaches for tumors and discusses the current problems associated with these therapeutic approaches and the prospects of NK cell therapy for tumors.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139089076","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 MEK/COX-2 axis improve immunotherapy efficacy in dMMR colorectal cancer with PIK3CA overexpression. 靶向MEK/COX-2轴提高PIK3CA过表达的dMMR结直肠癌的免疫疗法疗效
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-06-01 Epub Date: 2024-02-05 DOI: 10.1007/s13402-024-00916-y
Kunwei Peng, Yongxiang Liu, Shousheng Liu, Zining Wang, Huanling Zhang, Wenzhuo He, Yanan Jin, Lei Wang, Xiaojun Xia, Liangping Xia

Purpose: PIK3CA mutation or overexpression is associated with immunotherapy resistance in multiple cancer types, but is also paradoxically associated with benefit of COX-2 inhibition on patient survival of colorectal cancer (CRC) with mismatch repair deficiency (dMMR). This study examined whether and how PIK3CA status affected COX-2-mediated tumor inflammation and immunotherapy response of dMMR CRC.

Methods: Murine colon cancer cells MC38, CT26, and CT26-Mlh1-KO were used to construct PIK3CA knockdown and overexpression models to mimic dMMR CRC with PIK3CA dysregulation, and xenograft models were used to evaluate how PIK3CA regulate COX-2 expression, CD8+ T cells infiltration, tumor growth, and therapy response to anti-PD-L1 treatment using immunocompetent mice. Western blot was carried out to delineate the signaling pathways in human and mouse cancer cells, and immunohistochemical analysis together with bioinformatics analysis using human patient samples.

Results: PIK3CA upregulates COX-2 expression through MEK/ERK signaling pathway independent of AKT signaling to promote tumor inflammation and immunosuppression. PIK3CA knockdown profoundly reduced CT26 tumor growth in a CD8+ T cell-dependent manner, while PIK3CA overexpression significantly inhibited CD8+ T cells infiltration and promoted tumor growth. Furthermore, MEK or COX-2 inhibition augmented the anti-tumor activity of anti-PD-L1 immunotherapy on dMMR CRC mouse models, accompanied with increased CD8+ T cells infiltration and activated tumor microenvironment.

Conclusion: Our results identified that the PIK3CA hyperactivation in dMMR CRC upregulated COX-2 through MEK signaling, which inhibited CD8+ T cells infiltration and promoted tumor growth, together led to immunotherapy resistance. COX-2 or MEK inhibition may relieve therapy resistance and promote therapy efficacy of anti-PD-1/PD-L1 immunotherapy for treating dMMR CRC with PIK3CA overexpression or activating mutation.

目的:PIK3CA突变或过表达与多种癌症类型的免疫治疗耐药性有关,但也与错配修复缺陷(dMMR)结直肠癌(CRC)患者的COX-2抑制对患者生存的益处有关。本研究探讨了 PIK3CA 状态是否以及如何影响 COX-2 介导的肿瘤炎症和 dMMR CRC 的免疫治疗反应:方法:用小鼠结肠癌细胞MC38、CT26和CT26-Mlh1-KO构建PIK3CA敲除和过表达模型,以模拟PIK3CA失调的dMMR CRC;用免疫功能健全的小鼠建立异种移植模型,以评估PIK3CA如何调控COX-2表达、CD8+ T细胞浸润、肿瘤生长以及对抗PD-L1治疗的反应。利用免疫印迹技术对人类和小鼠癌细胞中的信号通路进行了描述,并利用人类患者样本进行了免疫组化分析和生物信息学分析:结果:PIK3CA通过独立于AKT信号的MEK/ERK信号通路上调COX-2的表达,从而促进肿瘤炎症和免疫抑制。PIK3CA敲除可显著降低CT26肿瘤的生长,其生长依赖于CD8+ T细胞,而PIK3CA过表达则可显著抑制CD8+ T细胞浸润并促进肿瘤生长。此外,MEK或COX-2抑制增强了抗PD-L1免疫疗法对dMMR CRC小鼠模型的抗肿瘤活性,同时增加了CD8+ T细胞浸润和激活肿瘤微环境:我们的研究结果表明,PIK3CA在dMMR CRC中的过度激活通过MEK信号转导上调COX-2,从而抑制CD8+ T细胞浸润并促进肿瘤生长,共同导致免疫治疗耐药。抑制COX-2或MEK可缓解抗PD-1/PD-L1免疫疗法治疗PIK3CA过表达或激活突变的dMMR CRC的耐药性并促进疗效。
{"title":"Targeting MEK/COX-2 axis improve immunotherapy efficacy in dMMR colorectal cancer with PIK3CA overexpression.","authors":"Kunwei Peng, Yongxiang Liu, Shousheng Liu, Zining Wang, Huanling Zhang, Wenzhuo He, Yanan Jin, Lei Wang, Xiaojun Xia, Liangping Xia","doi":"10.1007/s13402-024-00916-y","DOIUrl":"10.1007/s13402-024-00916-y","url":null,"abstract":"<p><strong>Purpose: </strong>PIK3CA mutation or overexpression is associated with immunotherapy resistance in multiple cancer types, but is also paradoxically associated with benefit of COX-2 inhibition on patient survival of colorectal cancer (CRC) with mismatch repair deficiency (dMMR). This study examined whether and how PIK3CA status affected COX-2-mediated tumor inflammation and immunotherapy response of dMMR CRC.</p><p><strong>Methods: </strong>Murine colon cancer cells MC38, CT26, and CT26-Mlh1-KO were used to construct PIK3CA knockdown and overexpression models to mimic dMMR CRC with PIK3CA dysregulation, and xenograft models were used to evaluate how PIK3CA regulate COX-2 expression, CD8<sup>+</sup> T cells infiltration, tumor growth, and therapy response to anti-PD-L1 treatment using immunocompetent mice. Western blot was carried out to delineate the signaling pathways in human and mouse cancer cells, and immunohistochemical analysis together with bioinformatics analysis using human patient samples.</p><p><strong>Results: </strong>PIK3CA upregulates COX-2 expression through MEK/ERK signaling pathway independent of AKT signaling to promote tumor inflammation and immunosuppression. PIK3CA knockdown profoundly reduced CT26 tumor growth in a CD8<sup>+</sup> T cell-dependent manner, while PIK3CA overexpression significantly inhibited CD8<sup>+</sup> T cells infiltration and promoted tumor growth. Furthermore, MEK or COX-2 inhibition augmented the anti-tumor activity of anti-PD-L1 immunotherapy on dMMR CRC mouse models, accompanied with increased CD8<sup>+</sup> T cells infiltration and activated tumor microenvironment.</p><p><strong>Conclusion: </strong>Our results identified that the PIK3CA hyperactivation in dMMR CRC upregulated COX-2 through MEK signaling, which inhibited CD8<sup>+</sup> T cells infiltration and promoted tumor growth, together led to immunotherapy resistance. COX-2 or MEK inhibition may relieve therapy resistance and promote therapy efficacy of anti-PD-1/PD-L1 immunotherapy for treating dMMR CRC with PIK3CA overexpression or activating mutation.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139693352","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
Silencing AHNAK promotes nasopharyngeal carcinoma progression by upregulating the ANXA2 protein. 沉默AHNAK可通过上调ANXA2蛋白促进鼻咽癌的进展。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-06-01 Epub Date: 2023-11-14 DOI: 10.1007/s13402-023-00898-3
Xingxing Lu, Yan Mei, Chunmei Fan, Pan Chen, Xiayu Li, Zhaoyang Zeng, Guiyuan Li, Wei Xiong, Bo Xiang, Mei Yi

Purpose: Nasopharyngeal carcinoma (NPC) is an aggressive head and neck disease with a high incidence of distant metastases. Enlargeosomes are cytoplasmic organelles marked by, desmoyokin/AHNAK. This study aimed to evaluate the expression of AHNAK in NPC and its effect on enlargeosomes and to investigate the correlation between AHNAK expression levels and clinical NPC patient characteristics.

Methods: Primary nasopharyngeal carcinoma (NPC) and NPC specimens were evaluated by analyzing public data, and immunohistochemistry. Systematic in vitro and in vivo experiments were performed using different NPC-derived cell lines and mouse models.

Results: In this study, we detected AHNAK and Annexin A2(ANXA2), a protein coating the surface of enlargeosomes, in NPC samples. We found that AHNAK was down-regulated. Down-regulation of AHNAK was associated with poor overall survival in NPC patients. Moreover, transcription factor FOSL1-mediated transcriptional repression was responsible for the low expression of AHNAK by recruiting EZH2. Whereas Annexin A2 was upregulated in human NPC tissues. Upregulation of Annexin A2 was associated with lymph node metastasis and distant metastasis in NPC patients. Functional studies confirmed that silencing of AHNAK enhanced the growth, invasion, and metastatic properties of NPC cells both in vitro and in vivo. In terms of mechanism, loss of AHNAK led to an increase of annexin A2 protein level in NPC cells. Silencing ANXA2 restored NPC cells' migrative and invasive ability upon loss of AHNAK.

Conclusion: Here, we report AHNAK as a tumor suppressor in NPC, which may act through annexin A2 oncogenic signaling in enlargeosome, with potential implications for novel approaches to NPC treatment.

目的:鼻咽癌(NPC)是一种侵袭性头颈部疾病,远处转移发生率高。放大小体是细胞质细胞器,标记为,desmoyokin/AHNAK。本研究旨在评价AHNAK在鼻咽癌中的表达及其对增大体的影响,探讨AHNAK表达水平与鼻咽癌患者临床特征的相关性。方法:采用公开资料分析和免疫组化方法对原发性鼻咽癌(NPC)及其标本进行评价。使用不同的npc衍生细胞系和小鼠模型进行了系统的体外和体内实验。结果:本研究在鼻咽癌样本中检测到AHNAK和膜联蛋白A2(ANXA2),这是一种覆盖在增大体表面的蛋白。我们发现AHNAK被下调。在鼻咽癌患者中,AHNAK的下调与较差的总生存率相关。此外,转录因子fosl1介导的转录抑制是通过募集EZH2导致AHNAK低表达的原因。而在人鼻咽癌组织中,Annexin A2表达上调。膜联蛋白A2的上调与鼻咽癌患者的淋巴结转移和远处转移有关。功能研究证实,在体外和体内,AHNAK的沉默增强了鼻咽癌细胞的生长、侵袭和转移特性。机制上,AHNAK缺失导致鼻咽癌细胞膜联蛋白A2水平升高。沉默ANXA2可以恢复失去AHNAK的鼻咽癌细胞的迁移和侵袭能力。结论:我们报道了AHNAK在鼻咽癌中作为肿瘤抑制因子,可能通过扩大体中的膜联蛋白A2致癌信号传导起作用,这可能对鼻咽癌治疗的新方法有潜在的意义。
{"title":"Silencing AHNAK promotes nasopharyngeal carcinoma progression by upregulating the ANXA2 protein.","authors":"Xingxing Lu, Yan Mei, Chunmei Fan, Pan Chen, Xiayu Li, Zhaoyang Zeng, Guiyuan Li, Wei Xiong, Bo Xiang, Mei Yi","doi":"10.1007/s13402-023-00898-3","DOIUrl":"10.1007/s13402-023-00898-3","url":null,"abstract":"<p><strong>Purpose: </strong>Nasopharyngeal carcinoma (NPC) is an aggressive head and neck disease with a high incidence of distant metastases. Enlargeosomes are cytoplasmic organelles marked by, desmoyokin/AHNAK. This study aimed to evaluate the expression of AHNAK in NPC and its effect on enlargeosomes and to investigate the correlation between AHNAK expression levels and clinical NPC patient characteristics.</p><p><strong>Methods: </strong>Primary nasopharyngeal carcinoma (NPC) and NPC specimens were evaluated by analyzing public data, and immunohistochemistry. Systematic in vitro and in vivo experiments were performed using different NPC-derived cell lines and mouse models.</p><p><strong>Results: </strong>In this study, we detected AHNAK and Annexin A2(ANXA2), a protein coating the surface of enlargeosomes, in NPC samples. We found that AHNAK was down-regulated. Down-regulation of AHNAK was associated with poor overall survival in NPC patients. Moreover, transcription factor FOSL1-mediated transcriptional repression was responsible for the low expression of AHNAK by recruiting EZH2. Whereas Annexin A2 was upregulated in human NPC tissues. Upregulation of Annexin A2 was associated with lymph node metastasis and distant metastasis in NPC patients. Functional studies confirmed that silencing of AHNAK enhanced the growth, invasion, and metastatic properties of NPC cells both in vitro and in vivo. In terms of mechanism, loss of AHNAK led to an increase of annexin A2 protein level in NPC cells. Silencing ANXA2 restored NPC cells' migrative and invasive ability upon loss of AHNAK.</p><p><strong>Conclusion: </strong>Here, we report AHNAK as a tumor suppressor in NPC, which may act through annexin A2 oncogenic signaling in enlargeosome, with potential implications for novel approaches to NPC treatment.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"92157167","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
O-GlcNAcylation of TRIM29 and OGT translation forms a feedback loop to promote adaptive response of PDAC cells to glucose deficiency. TRIM29 的 O-GlcNAcylation 和 OGT 翻译形成反馈回路,促进 PDAC 细胞对葡萄糖缺乏的适应性反应。
IF 4.9 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-06-01 Epub Date: 2024-02-12 DOI: 10.1007/s13402-023-00915-5
Fu-Ying Zhao, Xue Chen, Jia-Mei Wang, Ye Yuan, Chao Li, Jia Sun, Hua-Qin Wang

Purpose: Glucose not only provides energy for tumor cells, but also provides various biomolecules that are essential for their survival, proliferation and invasion. Therefore, it is of great clinical significance to understand the mechanism of how tumor cells adapt to metabolic stress and maintain their survival. The aim of this research was to study the critical role of OGT and TRIM29 O-GlcNAc modification driven adaptability of PDAC cells to low glucose stress, which might have important medical implications for PDAC therapy.

Methods: Western blotting, mass spectrometry and WGA-immunoprecipitation were used to examined the levels of OGT and O-GlcNAc glycosylated proteins in BxPC3 and SW1990 cells in normal culture and under glucose deprivation conditions. Crystal violet assay, flow cytometry, RIP, RT-qPCR, protein stability assay, biotin pull down were used to investigate the mechanism of OGT and TRIM29-mediated adaptive response to glucose deficiency in PDAC cells.

Results: The current study found that under the condition of low glucose culture, the levels of OGT and O-GlcNAc glycosylation in PDAC cells were significantly higher than those in normal culture. Moreover, the high expression of OGT has a protective effect on PDAC cells under low glucose stress. This study confirmed that there was no significant change in mRNA level and protein degradation of OGT under low glucose stress, which was mainly reflected in the increase of protein synthesis. In addition, O-GlcNAc modification at T120 site plays a critical role in the metabolic adaptive responses mediated by TRIM29.

Conclusions: Taken together, our study indicated that O-GlcNAcylation of TRIM29 at T120 site and OGT translation forms a loop feedback to facilitate survival of PDAC under glucose deficiency.

目的:葡萄糖不仅能为肿瘤细胞提供能量,还能提供肿瘤细胞生存、增殖和侵袭所必需的各种生物大分子。因此,了解肿瘤细胞如何适应代谢压力并维持生存的机制具有重要的临床意义。本研究旨在研究OGT和TRIM29 O-GlcNAc修饰驱动PDAC细胞适应低糖应激的关键作用,这可能对PDAC的治疗具有重要的医学意义:方法:采用 Western 印迹、质谱和 WGA 免疫沉淀技术检测正常培养和葡萄糖剥夺条件下 BxPC3 和 SW1990 细胞中 OGT 和 O-GlcNAc 糖基化蛋白的水平。采用水晶紫检测法、流式细胞术、RIP、RT-qPCR、蛋白质稳定性检测、生物素牵引等方法研究了OGT和TRIM29介导的PDAC细胞对葡萄糖缺乏的适应性反应机制:结果:本研究发现,在低糖培养条件下,PDAC 细胞的 OGT 和 O-GlcNAc 糖基化水平明显高于正常培养条件下。此外,OGT的高表达对低糖应激下的PDAC细胞有保护作用。本研究证实,OGT 的 mRNA 水平和蛋白降解在低糖应激下无明显变化,主要体现在蛋白合成的增加。此外,T120位点的O-GlcNAc修饰在TRIM29介导的代谢适应性反应中起着关键作用:综上所述,我们的研究表明,TRIM29在T120位点的O-GlcNAc修饰与OGT翻译形成环路反馈,促进了PDAC在葡萄糖缺乏条件下的存活。
{"title":"O-GlcNAcylation of TRIM29 and OGT translation forms a feedback loop to promote adaptive response of PDAC cells to glucose deficiency.","authors":"Fu-Ying Zhao, Xue Chen, Jia-Mei Wang, Ye Yuan, Chao Li, Jia Sun, Hua-Qin Wang","doi":"10.1007/s13402-023-00915-5","DOIUrl":"10.1007/s13402-023-00915-5","url":null,"abstract":"<p><strong>Purpose: </strong>Glucose not only provides energy for tumor cells, but also provides various biomolecules that are essential for their survival, proliferation and invasion. Therefore, it is of great clinical significance to understand the mechanism of how tumor cells adapt to metabolic stress and maintain their survival. The aim of this research was to study the critical role of OGT and TRIM29 O-GlcNAc modification driven adaptability of PDAC cells to low glucose stress, which might have important medical implications for PDAC therapy.</p><p><strong>Methods: </strong>Western blotting, mass spectrometry and WGA-immunoprecipitation were used to examined the levels of OGT and O-GlcNAc glycosylated proteins in BxPC3 and SW1990 cells in normal culture and under glucose deprivation conditions. Crystal violet assay, flow cytometry, RIP, RT-qPCR, protein stability assay, biotin pull down were used to investigate the mechanism of OGT and TRIM29-mediated adaptive response to glucose deficiency in PDAC cells.</p><p><strong>Results: </strong>The current study found that under the condition of low glucose culture, the levels of OGT and O-GlcNAc glycosylation in PDAC cells were significantly higher than those in normal culture. Moreover, the high expression of OGT has a protective effect on PDAC cells under low glucose stress. This study confirmed that there was no significant change in mRNA level and protein degradation of OGT under low glucose stress, which was mainly reflected in the increase of protein synthesis. In addition, O-GlcNAc modification at T120 site plays a critical role in the metabolic adaptive responses mediated by TRIM29.</p><p><strong>Conclusions: </strong>Taken together, our study indicated that O-GlcNAcylation of TRIM29 at T120 site and OGT translation forms a loop feedback to facilitate survival of PDAC under glucose deficiency.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":4.9,"publicationDate":"2024-06-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139724617","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
ONECUT2 is a druggable driver of luminal to basal breast cancer plasticity. ONECUT2是乳腺癌从管腔型向基底型转变的可药驱动因素。
IF 6.6 2区 医学 Q1 Medicine Pub Date : 2024-05-31 DOI: 10.1007/s13402-024-00957-3
Irene Zamora, Mirian Gutiérrez, Alex Pascual, María J Pajares, Miguel Barajas, Lillian M Perez, Sungyong You, Beatrice S Knudsen, Michael R Freeman, Ignacio J Encío, Mirja Rotinen

Purpose: Tumor heterogeneity complicates patient treatment and can be due to transitioning of cancer cells across phenotypic cell states. This process is associated with the acquisition of independence from an oncogenic driver, such as the estrogen receptor (ER) in breast cancer (BC), resulting in tumor progression, therapeutic failure and metastatic spread. The transcription factor ONECUT2 (OC2) has been shown to be a master regulator protein of metastatic castration-resistant prostate cancer (mCRPC) tumors that promotes lineage plasticity to a drug-resistant neuroendocrine (NEPC) phenotype. Here, we investigate the role of OC2 in the dynamic conversion between different molecular subtypes in BC.

Methods: We analyze OC2 expression and clinical significance in BC using public databases and immunohistochemical staining. In vitro, we perform RNA-Seq, RT-qPCR and western-blot after OC2 enforced expression. We also assess cellular effects of OC2 silencing and inhibition with a drug-like small molecule in vitro and in vivo.

Results: OC2 is highly expressed in a substantial subset of hormone receptor negative human BC tumors and tamoxifen-resistant models, and is associated with poor clinical outcome, lymph node metastasis and heightened clinical stage. OC2 inhibits ER expression and activity, suppresses a gene expression program associated with luminal differentiation and activates a basal-like state at the gene expression level. We also show that OC2 is required for cell growth and survival in metastatic BC models and that it can be targeted with a small molecule inhibitor providing a novel therapeutic strategy for patients with OC2 active tumors.

Conclusions: The transcription factor OC2 is a driver of BC heterogeneity and a potential drug target in distinct cell states within the breast tumors.

目的:肿瘤的异质性使患者的治疗变得复杂,这可能是由于癌细胞在不同表型细胞状态之间的转换。这一过程与从致癌驱动因子(如乳腺癌(BC)中的雌激素受体(ER))中获得独立性有关,从而导致肿瘤进展、治疗失败和转移扩散。转录因子ONECUT2(OC2)已被证明是转移性耐阉割前列腺癌(mCRPC)肿瘤的主调节蛋白,它能促进肿瘤细胞系的可塑性,形成耐药的神经内分泌(NEPC)表型。在此,我们研究了 OC2 在 BC 不同分子亚型之间动态转换中的作用:方法:我们利用公共数据库和免疫组化染色分析了OC2在BC中的表达和临床意义。在体外,我们在 OC2 强化表达后进行 RNA-Seq、RT-qPCR 和 Western-blot 分析。我们还在体外和体内评估了沉默 OC2 和使用类药物小分子抑制 OC2 对细胞的影响:结果:OC2在大量激素受体阴性的人类BC肿瘤亚群和他莫昔芬耐药模型中高表达,并与临床预后差、淋巴结转移和临床分期增高有关。OC2 可抑制 ER 的表达和活性,抑制与管腔分化相关的基因表达程序,并在基因表达水平上激活基底样状态。我们还发现,OC2 是转移性 BC 模型中细胞生长和存活所必需的,它可以用小分子抑制剂作为靶点,为 OC2 活跃的肿瘤患者提供一种新的治疗策略:结论:转录因子OC2是乳腺癌异质性的驱动因素,也是乳腺肿瘤内不同细胞状态的潜在药物靶点。
{"title":"ONECUT2 is a druggable driver of luminal to basal breast cancer plasticity.","authors":"Irene Zamora, Mirian Gutiérrez, Alex Pascual, María J Pajares, Miguel Barajas, Lillian M Perez, Sungyong You, Beatrice S Knudsen, Michael R Freeman, Ignacio J Encío, Mirja Rotinen","doi":"10.1007/s13402-024-00957-3","DOIUrl":"https://doi.org/10.1007/s13402-024-00957-3","url":null,"abstract":"<p><strong>Purpose: </strong>Tumor heterogeneity complicates patient treatment and can be due to transitioning of cancer cells across phenotypic cell states. This process is associated with the acquisition of independence from an oncogenic driver, such as the estrogen receptor (ER) in breast cancer (BC), resulting in tumor progression, therapeutic failure and metastatic spread. The transcription factor ONECUT2 (OC2) has been shown to be a master regulator protein of metastatic castration-resistant prostate cancer (mCRPC) tumors that promotes lineage plasticity to a drug-resistant neuroendocrine (NEPC) phenotype. Here, we investigate the role of OC2 in the dynamic conversion between different molecular subtypes in BC.</p><p><strong>Methods: </strong>We analyze OC2 expression and clinical significance in BC using public databases and immunohistochemical staining. In vitro, we perform RNA-Seq, RT-qPCR and western-blot after OC2 enforced expression. We also assess cellular effects of OC2 silencing and inhibition with a drug-like small molecule in vitro and in vivo.</p><p><strong>Results: </strong>OC2 is highly expressed in a substantial subset of hormone receptor negative human BC tumors and tamoxifen-resistant models, and is associated with poor clinical outcome, lymph node metastasis and heightened clinical stage. OC2 inhibits ER expression and activity, suppresses a gene expression program associated with luminal differentiation and activates a basal-like state at the gene expression level. We also show that OC2 is required for cell growth and survival in metastatic BC models and that it can be targeted with a small molecule inhibitor providing a novel therapeutic strategy for patients with OC2 active tumors.</p><p><strong>Conclusions: </strong>The transcription factor OC2 is a driver of BC heterogeneity and a potential drug target in distinct cell states within the breast tumors.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":null,"pages":null},"PeriodicalIF":6.6,"publicationDate":"2024-05-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141181411","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