首页 > 最新文献

Journal of Experimental & Clinical Cancer Research最新文献

英文 中文
Identification of circulating tumor cells captured by the FDA-cleared Parsortix® PC1 system from the peripheral blood of metastatic breast cancer patients using immunofluorescence and cytopathological evaluations. 利用免疫荧光和细胞病理学评估,从转移性乳腺癌患者的外周血中鉴定经 FDA 批准的 Parsortix® PC1 系统捕获的循环肿瘤细胞。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-21 DOI: 10.1186/s13046-024-03149-x
Mariacristina Ciccioli, Kyukwang Kim, Negar Khazan, Joseph D Khoury, Martin J Cooke, M Craig Miller, Daniel J O'Shannessy, Anne-Sophie Pailhes-Jimenez, Richard G Moore

Circulating Tumor Cells (CTCs) may serve as a non-invasive source of tumor material to investigate an individual's disease in real-time. The Parsortix® PC1 System, the first FDA-cleared medical device for the capture and harvest of CTCs from peripheral blood of metastatic breast cancer (MBC) patients for use in subsequent user-validated downstream analyses, enables the epitope-independent capture of CTCs with diverse phenotypes based on cell size and deformability. The aim of this study was to determine the proportion of MBC patients and self-declared female healthy volunteers (HVs) that had CTCs identified using immunofluorescence (IF) or Wright-Giemsa (WG) staining. Peripheral blood from 76 HVs and 76 MBC patients was processed on Parsortix® PC1 Systems. Harvested cells were cytospun onto a charged slide and immunofluorescently stained for identification of CTCs expressing epithelial markers. The IF slides were subsequently WG-stained and analyzed for CTC identification based on morphological features of malignant cells. All testing was performed by operators blinded to the clinical status of each subject. CTCs were identified on the IF slides in 45.3% (≥ 1) / 24.0% (≥ 5) of the MBC patients (range = 0 - 125, mean = 7) and in 6.9% (≥ 1) / 2.8% (≥ 5) of the HVs (range = 0 - 28, mean = 1). Among the MBC patients with ≥ 1 CTC, 70.6% had only CK + /EpCAM- CTCs, with none having EpCAM + /CK- CTCs. CTC clusters were identified in 56.0% of the CTC-positive patients. On the WG-stained slides, CTCs were identified in 42.9% (≥ 1) / 21.4% (≥ 5) of the MBC patients (range = 0 - 41, mean = 4) and 4.3% (≥ 1) / 2.9% (≥ 5) of the HVs (range = 0 - 14, mean = 0). This study demonstrated the ability of the Parsortix® PC1 System to capture and harvest CTCs from a significantly larger proportion of MBC patients compared to HVs when coupled with both IF and WG cytomorphological assessment. The presence of epithelial cells in subjects without diagnosed disease has been previously described, with their significance being unclear. Interestingly, a high proportion of the identified CTCs did not express EpCAM, highlighting the limitations of using EpCAM-based approaches.

循环肿瘤细胞 (CTC) 可作为一种非侵入性的肿瘤材料来源,用于实时调查个人疾病。Parsortix® PC1 系统是首款通过 FDA 认证的医疗设备,可从转移性乳腺癌(MBC)患者的外周血中捕获和采集 CTCs,用于后续的用户验证下游分析。本研究旨在确定使用免疫荧光(IF)或赖特-吉氏(WG)染色法鉴定出 CTC 的 MBC 患者和自我申报的女性健康志愿者(HV)的比例。76 名健康志愿者和 76 名乳腺癌患者的外周血在 Parsortix® PC1 系统上进行处理。采集的细胞被置于带电载玻片上并进行免疫荧光染色,以识别表达上皮标记的 CTC。随后对免疫荧光载玻片进行 WG 染色,并根据恶性细胞的形态特征进行 CTC 鉴定分析。所有检测均由对每位受检者的临床状况保密的操作人员进行。45.3% (≥ 1) / 24.0% (≥ 5) 的 MBC 患者(范围 = 0 - 125,平均 = 7)和 6.9% (≥ 1) / 2.8% (≥ 5) 的 HV 患者(范围 = 0 - 28,平均 = 1)在 IF 切片上鉴定出了 CTC。在≥1个CTC的MBC患者中,70.6%只有CK + /EpCAM- CTC,没有EpCAM + /CK- CTC。在 56.0% 的 CTC 阳性患者中发现了 CTC 簇。在 WG 染色的切片上,42.9%(≥ 1)/21.4%(≥ 5)的 MBC 患者(范围 = 0 - 41,平均 = 4)和 4.3%(≥ 1)/2.9%(≥ 5)的 HV 患者(范围 = 0 - 14,平均 = 0)中发现了 CTC。这项研究表明,与 HVs 相比,Parsortix® PC1 系统在结合 IF 和 WG 细胞形态学评估时,能从更大比例的 MBC 患者身上捕获和收获 CTCs。以前曾描述过未确诊疾病的受试者体内存在上皮细胞,但其意义尚不明确。有趣的是,已鉴定的 CTC 中有很大一部分不表达 EpCAM,这凸显了基于 EpCAM 方法的局限性。
{"title":"Identification of circulating tumor cells captured by the FDA-cleared Parsortix<sup>®</sup> PC1 system from the peripheral blood of metastatic breast cancer patients using immunofluorescence and cytopathological evaluations.","authors":"Mariacristina Ciccioli, Kyukwang Kim, Negar Khazan, Joseph D Khoury, Martin J Cooke, M Craig Miller, Daniel J O'Shannessy, Anne-Sophie Pailhes-Jimenez, Richard G Moore","doi":"10.1186/s13046-024-03149-x","DOIUrl":"10.1186/s13046-024-03149-x","url":null,"abstract":"<p><p>Circulating Tumor Cells (CTCs) may serve as a non-invasive source of tumor material to investigate an individual's disease in real-time. The Parsortix<sup>®</sup> PC1 System, the first FDA-cleared medical device for the capture and harvest of CTCs from peripheral blood of metastatic breast cancer (MBC) patients for use in subsequent user-validated downstream analyses, enables the epitope-independent capture of CTCs with diverse phenotypes based on cell size and deformability. The aim of this study was to determine the proportion of MBC patients and self-declared female healthy volunteers (HVs) that had CTCs identified using immunofluorescence (IF) or Wright-Giemsa (WG) staining. Peripheral blood from 76 HVs and 76 MBC patients was processed on Parsortix<sup>®</sup> PC1 Systems. Harvested cells were cytospun onto a charged slide and immunofluorescently stained for identification of CTCs expressing epithelial markers. The IF slides were subsequently WG-stained and analyzed for CTC identification based on morphological features of malignant cells. All testing was performed by operators blinded to the clinical status of each subject. CTCs were identified on the IF slides in 45.3% (≥ 1) / 24.0% (≥ 5) of the MBC patients (range = 0 - 125, mean = 7) and in 6.9% (≥ 1) / 2.8% (≥ 5) of the HVs (range = 0 - 28, mean = 1). Among the MBC patients with ≥ 1 CTC, 70.6% had only CK + /EpCAM- CTCs, with none having EpCAM + /CK- CTCs. CTC clusters were identified in 56.0% of the CTC-positive patients. On the WG-stained slides, CTCs were identified in 42.9% (≥ 1) / 21.4% (≥ 5) of the MBC patients (range = 0 - 41, mean = 4) and 4.3% (≥ 1) / 2.9% (≥ 5) of the HVs (range = 0 - 14, mean = 0). This study demonstrated the ability of the Parsortix<sup>®</sup> PC1 System to capture and harvest CTCs from a significantly larger proportion of MBC patients compared to HVs when coupled with both IF and WG cytomorphological assessment. The presence of epithelial cells in subjects without diagnosed disease has been previously described, with their significance being unclear. Interestingly, a high proportion of the identified CTCs did not express EpCAM, highlighting the limitations of using EpCAM-based approaches.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"240"},"PeriodicalIF":11.4,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11337573/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142019445","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Two-polarized roles of transcription factor FOSB in lung cancer progression and prognosis: dependent on p53 status. 转录因子 FOSB 在肺癌进展和预后中的两极作用:取决于 p53 状态。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-21 DOI: 10.1186/s13046-024-03161-1
Hongchao Zhang, Guopei Zhang, Mingyang Xiao, Su Cui, Cuihong Jin, Jinghua Yang, Shengwen Wu, Xiaobo Lu

Background: Activator protein-1 (AP-1) represents a transcription factor family that has garnered growing attention for its extensive involvement in tumor biology. However, the roles of the AP-1 family in the evolution of lung cancer remain poorly characterized. FBJ Murine Osteosarcoma Viral Oncogene Homolog B (FOSB), a classic AP-1 family member, was previously reported to play bewilderingly two-polarized roles in non-small cell lung cancer (NSCLC) as an enigmatic double-edged sword, for which the reasons and significance warrant further elucidation.

Methods and results: Based on the bioinformatics analysis of a large NSCLC cohort from the TCGA database, our current work found the well-known tumor suppressor gene TP53 served as a key code to decipher the two sides of FOSB - its expression indicated a positive prognosis in NSCLC patients harboring wild-type TP53 while a negative one in those harboring mutant TP53. By constructing a panel of syngeneically derived NSCLC cells expressing p53 in different statuses, the radically opposite prognostic effects of FOSB expression in NSCLC population were validated, with the TP53-R248Q mutation site emerging as particularly meaningful. Transcriptome sequencing showed that FOSB overexpression elicited diversifying transcriptomic landscapes across NSCLC cells with varying genetic backgrounds of TP53 and, combined with the validation by RT-qPCR, PREX1 (TP53-Null), IGFBP5 (TP53-WT), AKR1C3, and ALDH3A1 (TP53-R248Q) were respectively identified as p53-dependent transcriptional targets of FOSB. Subsequently, the heterogenous impacts of FOSB on the tumor biology in NSCLC cells via the above selective transcriptional targets were confirmed in vitro and in vivo. Mechanistic investigations revealed that wild-type or mutant p53 might guide FOSB to recognize and bind to distinct promoter sequences via protein-protein interactions to transcriptionally activate specific target genes, thereby creating disparate influences on the progression and prognosis in NSCLC.

Conclusions: FOSB expression holds promise as a novel prognostic biomarker for NSCLC in combination with a given genetic background of TP53, and the unique interactions between FOSB and p53 may serve as underlying intervention targets for NSCLC.

背景:激活蛋白-1(AP-1)是一个转录因子家族,因其广泛参与肿瘤生物学而日益受到关注。然而,AP-1 家族在肺癌演化过程中的作用仍然鲜为人知。FBJ Murine Osteosarcoma Viral Oncogene Homolog B(FOSB)是典型的AP-1家族成员,之前有报道称它在非小细胞肺癌(NSCLC)中扮演着令人困惑的两极角色,是一把神秘的双刃剑,其原因和意义值得进一步阐明:基于对 TCGA 数据库中一个大型 NSCLC 队列的生物信息学分析,我们目前的工作发现,著名的肿瘤抑制基因 TP53 是破译 FOSB 两面性的关键代码--其表达在携带野生型 TP53 的 NSCLC 患者中显示出积极的预后,而在携带突变型 TP53 的患者中则显示出消极的预后。通过构建一个在不同状态下表达 p53 的合成基因衍生 NSCLC 细胞面板,验证了 FOSB 表达在 NSCLC 群体中截然相反的预后效果,其中 TP53-R248Q 突变位点的表达尤为重要。转录组测序显示,FOSB的过表达在具有不同TP53遗传背景的NSCLC细胞中引起了多样化的转录组景观,结合RT-qPCR的验证,PREX1(TP53-Null)、IGFBP5(TP53-WT)、AKR1C3和ALDH3A1(TP53-R248Q)分别被确定为FOSB的p53依赖性转录靶标。随后,在体外和体内证实了FOSB通过上述选择性转录靶点对NSCLC细胞肿瘤生物学的不同影响。机理研究发现,野生型或突变型p53可能通过蛋白-蛋白相互作用引导FOSB识别并结合到不同的启动子序列,转录激活特定的靶基因,从而对NSCLC的进展和预后产生不同的影响:结论:结合特定的TP53遗传背景,FOSB的表达有望成为NSCLC的新型预后生物标志物,FOSB与p53之间的独特相互作用可能成为NSCLC的潜在干预靶点。
{"title":"Two-polarized roles of transcription factor FOSB in lung cancer progression and prognosis: dependent on p53 status.","authors":"Hongchao Zhang, Guopei Zhang, Mingyang Xiao, Su Cui, Cuihong Jin, Jinghua Yang, Shengwen Wu, Xiaobo Lu","doi":"10.1186/s13046-024-03161-1","DOIUrl":"10.1186/s13046-024-03161-1","url":null,"abstract":"<p><strong>Background: </strong>Activator protein-1 (AP-1) represents a transcription factor family that has garnered growing attention for its extensive involvement in tumor biology. However, the roles of the AP-1 family in the evolution of lung cancer remain poorly characterized. FBJ Murine Osteosarcoma Viral Oncogene Homolog B (FOSB), a classic AP-1 family member, was previously reported to play bewilderingly two-polarized roles in non-small cell lung cancer (NSCLC) as an enigmatic double-edged sword, for which the reasons and significance warrant further elucidation.</p><p><strong>Methods and results: </strong>Based on the bioinformatics analysis of a large NSCLC cohort from the TCGA database, our current work found the well-known tumor suppressor gene TP53 served as a key code to decipher the two sides of FOSB - its expression indicated a positive prognosis in NSCLC patients harboring wild-type TP53 while a negative one in those harboring mutant TP53. By constructing a panel of syngeneically derived NSCLC cells expressing p53 in different statuses, the radically opposite prognostic effects of FOSB expression in NSCLC population were validated, with the TP53-R248Q mutation site emerging as particularly meaningful. Transcriptome sequencing showed that FOSB overexpression elicited diversifying transcriptomic landscapes across NSCLC cells with varying genetic backgrounds of TP53 and, combined with the validation by RT-qPCR, PREX1 (TP53-Null), IGFBP5 (TP53-WT), AKR1C3, and ALDH3A1 (TP53-R248Q) were respectively identified as p53-dependent transcriptional targets of FOSB. Subsequently, the heterogenous impacts of FOSB on the tumor biology in NSCLC cells via the above selective transcriptional targets were confirmed in vitro and in vivo. Mechanistic investigations revealed that wild-type or mutant p53 might guide FOSB to recognize and bind to distinct promoter sequences via protein-protein interactions to transcriptionally activate specific target genes, thereby creating disparate influences on the progression and prognosis in NSCLC.</p><p><strong>Conclusions: </strong>FOSB expression holds promise as a novel prognostic biomarker for NSCLC in combination with a given genetic background of TP53, and the unique interactions between FOSB and p53 may serve as underlying intervention targets for NSCLC.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"237"},"PeriodicalIF":11.4,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11337850/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142009859","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Areca nut-induced metabolic reprogramming and M2 differentiation promote OPMD malignant transformation. 亚麻仁诱导的代谢重编程和M2分化促进了OPMD的恶性转化。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-20 DOI: 10.1186/s13046-024-03163-z
Shyng-Shiou F Yuan, Leong-Perng Chan, Hieu D H Nguyen, Chang-Wei Su, Yuk-Kwan Chen, Jeff Yi-Fu Chen, Shigetaka Shimodaira, Stephen Chu-Sung Hu, Steven Lo, Yen-Yun Wang

Background: Betel quid and its major ingredient, areca nut, are recognized by IARC as major risk factors in oral cancer development. Areca nut extract (ANE) exposure has been linked to OPMD progression and malignant transformation to OSCC. However, the detailed mechanism through which ANE acts on other cell types in the oral microenvironment to promote oral carcinogenesis remains elusive.

Methods: Immunoprofiling of macrophages associated with OPMD and OSCC was carried out by immunohistochemical and immunofluorescence staining. Phosphokinase and cytokine arrays and western blotting were performed to determine the underlying mechanisms. Transwell assays were used to evaluate the migration-promoting effect of ANE. Hamster model was finally applied to confirm the in vivo effect of ANE.

Results: We reported that M2 macrophages positively correlated with oral cancer progression. ANE induced M2 macrophage differentiation, CREB phosphorylation and VCAM-1 secretion and increased mitochondrial metabolism. Conditioned medium and VCAM-1 from ANE-treated macrophages promoted migration and mesenchymal phenotypes in oral precancer cells. In vivo studies showed that ANE enhanced M2 polarization and related signaling pathways in the oral buccal tissues of hamsters.

Conclusion: Our study provides novel mechanisms for areca nut-induced oral carcinogenesis, demonstrating that areca nut promotes M2 macrophage differentiation and secretion of oncogenic cytokines that critically activate malignant transformation of oral premalignant cells.

背景:国际癌症研究机构(IARC)认为,槟榔及其主要成分槟榔是导致口腔癌的主要危险因素。接触阿月浑子提取物(ANE)与口腔微小病变(OPMD)的发展和恶性转化为口腔癌(OSCC)有关。然而,ANE作用于口腔微环境中其他细胞类型以促进口腔癌发生的详细机制仍未确定:方法:通过免疫组织化学和免疫荧光染色对与 OPMD 和 OSCC 相关的巨噬细胞进行免疫分析。方法:通过免疫组织化学和免疫荧光染色对与OPMD和OSCC相关的巨噬细胞进行免疫分析,并进行磷酸激酶和细胞因子阵列分析和免疫印迹,以确定其潜在机制。采用 Transwell 试验评估 ANE 的迁移促进作用。最后应用仓鼠模型证实了 ANE 的体内效应:结果:我们发现M2巨噬细胞与口腔癌的进展呈正相关。ANE诱导M2巨噬细胞分化、CREB磷酸化和VCAM-1分泌,并增加线粒体代谢。经ANE处理的巨噬细胞的培养基和VCAM-1可促进口腔癌前病变细胞的迁移和间质表型。体内研究表明,ANE 增强了仓鼠口腔颊组织中的 M2 极化和相关信号通路:我们的研究为山苍子诱导口腔癌的发生提供了新的机制,证明山苍子能促进 M2 巨噬细胞分化并分泌致癌细胞因子,从而关键性地激活口腔癌前病变细胞的恶性转化。
{"title":"Areca nut-induced metabolic reprogramming and M2 differentiation promote OPMD malignant transformation.","authors":"Shyng-Shiou F Yuan, Leong-Perng Chan, Hieu D H Nguyen, Chang-Wei Su, Yuk-Kwan Chen, Jeff Yi-Fu Chen, Shigetaka Shimodaira, Stephen Chu-Sung Hu, Steven Lo, Yen-Yun Wang","doi":"10.1186/s13046-024-03163-z","DOIUrl":"10.1186/s13046-024-03163-z","url":null,"abstract":"<p><strong>Background: </strong>Betel quid and its major ingredient, areca nut, are recognized by IARC as major risk factors in oral cancer development. Areca nut extract (ANE) exposure has been linked to OPMD progression and malignant transformation to OSCC. However, the detailed mechanism through which ANE acts on other cell types in the oral microenvironment to promote oral carcinogenesis remains elusive.</p><p><strong>Methods: </strong>Immunoprofiling of macrophages associated with OPMD and OSCC was carried out by immunohistochemical and immunofluorescence staining. Phosphokinase and cytokine arrays and western blotting were performed to determine the underlying mechanisms. Transwell assays were used to evaluate the migration-promoting effect of ANE. Hamster model was finally applied to confirm the in vivo effect of ANE.</p><p><strong>Results: </strong>We reported that M2 macrophages positively correlated with oral cancer progression. ANE induced M2 macrophage differentiation, CREB phosphorylation and VCAM-1 secretion and increased mitochondrial metabolism. Conditioned medium and VCAM-1 from ANE-treated macrophages promoted migration and mesenchymal phenotypes in oral precancer cells. In vivo studies showed that ANE enhanced M2 polarization and related signaling pathways in the oral buccal tissues of hamsters.</p><p><strong>Conclusion: </strong>Our study provides novel mechanisms for areca nut-induced oral carcinogenesis, demonstrating that areca nut promotes M2 macrophage differentiation and secretion of oncogenic cytokines that critically activate malignant transformation of oral premalignant cells.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"233"},"PeriodicalIF":11.4,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11334407/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142005750","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The deubiquitinase USP15 drives malignant progression of gastric cancer through glucose metabolism remodeling. 去泛素化酶USP15通过葡萄糖代谢重塑驱动胃癌恶性进展
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-20 DOI: 10.1186/s13046-024-03152-2
Longtao Huangfu, Huanbo Zhu, Gangjian Wang, Junbing Chen, Yongqi Wang, Biao Fan, Xiaoyang Wang, Qian Yao, Ting Guo, Jing Han, Ying Hu, Hong Du, Xiaomei Li, Jiafu Ji, Xiaofang Xing

Background: Ubiquitin-specific protease 15 (USP15) exhibits amplifications in various tumors, including gastric cancer (GC), yet its biological function and mechanisms in GC progression remain elusive.

Methods: Here, we established stable USP15 knockdown or overexpression GC cell lines and explored the potential mechanism of USP15 in GC. Besides, we also identified interacting targets of USP15.

Results: USP15 knockdown significantly impeded cell proliferation, invasion, epithelial-mesenchymal transition, and distal colonization in xenograft models, while enhancing oxaliplatin's antitumor effect. USP15 was involved in ubiquitination modification of glycolytic regulators. Silencing of USP15 suppressed glycolytic activity and impaired mitochondrial functions. Interference with USP15 expression reversed tumor progression and distal colonization in vivo. HKDC1 and IGF2BP3 were found as core interacting targets of USP15, and HKDC1 was identified as a substrate for ubiquitination modification by USP15, whereby USP15 regulated glucose metabolism activity by inhibiting the ubiquitination degradation of HKDC1.

Conclusions: Our study unveiled aberrantly high expression of USP15 in GC tissues, correlating with malignant progression and nonresponse to neoadjuvant therapy. USP15 inhibitors, if developed, could be effective in promoting chemotherapy through glucose metabolism remodeling.

背景:方法:我们建立了稳定的 USP15 敲除或过表达 GC 细胞系,并探索了 USP15 在 GC 中的潜在机制。此外,我们还发现了USP15的相互作用靶点:结果:在异种移植模型中,敲除 USP15 能明显抑制细胞增殖、侵袭、上皮-间质转化和远端定植,同时增强奥沙利铂的抗肿瘤效果。USP15 参与了糖酵解调节因子的泛素化修饰。沉默 USP15 会抑制糖酵解活性并损害线粒体功能。干扰 USP15 的表达可逆转体内肿瘤的进展和远端定植。研究发现,HKDC1和IGF2BP3是USP15的核心相互作用靶标,HKDC1是USP15泛素化修饰的底物,USP15通过抑制HKDC1的泛素化降解来调节糖代谢活性:我们的研究揭示了 USP15 在 GC 组织中的异常高表达,这与恶性进展和对新辅助治疗无反应有关。如果开发出 USP15 抑制剂,可通过重塑葡萄糖代谢有效促进化疗。
{"title":"The deubiquitinase USP15 drives malignant progression of gastric cancer through glucose metabolism remodeling.","authors":"Longtao Huangfu, Huanbo Zhu, Gangjian Wang, Junbing Chen, Yongqi Wang, Biao Fan, Xiaoyang Wang, Qian Yao, Ting Guo, Jing Han, Ying Hu, Hong Du, Xiaomei Li, Jiafu Ji, Xiaofang Xing","doi":"10.1186/s13046-024-03152-2","DOIUrl":"10.1186/s13046-024-03152-2","url":null,"abstract":"<p><strong>Background: </strong>Ubiquitin-specific protease 15 (USP15) exhibits amplifications in various tumors, including gastric cancer (GC), yet its biological function and mechanisms in GC progression remain elusive.</p><p><strong>Methods: </strong>Here, we established stable USP15 knockdown or overexpression GC cell lines and explored the potential mechanism of USP15 in GC. Besides, we also identified interacting targets of USP15.</p><p><strong>Results: </strong>USP15 knockdown significantly impeded cell proliferation, invasion, epithelial-mesenchymal transition, and distal colonization in xenograft models, while enhancing oxaliplatin's antitumor effect. USP15 was involved in ubiquitination modification of glycolytic regulators. Silencing of USP15 suppressed glycolytic activity and impaired mitochondrial functions. Interference with USP15 expression reversed tumor progression and distal colonization in vivo. HKDC1 and IGF2BP3 were found as core interacting targets of USP15, and HKDC1 was identified as a substrate for ubiquitination modification by USP15, whereby USP15 regulated glucose metabolism activity by inhibiting the ubiquitination degradation of HKDC1.</p><p><strong>Conclusions: </strong>Our study unveiled aberrantly high expression of USP15 in GC tissues, correlating with malignant progression and nonresponse to neoadjuvant therapy. USP15 inhibitors, if developed, could be effective in promoting chemotherapy through glucose metabolism remodeling.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"235"},"PeriodicalIF":11.4,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11334570/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142009857","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Repurposed AT9283 triggers anti-tumoral effects by targeting MKK3 oncogenic functions in Colorectal Cancer. 靶向 MKK3 致癌功能的 AT9283 在结直肠癌中发挥抗肿瘤作用。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-20 DOI: 10.1186/s13046-024-03150-4
Valentina Piastra, Federica Ganci, Andrea Sacconi, Angelina Pranteda, Matteo Allegretti, Roberta Bernardini, Martina Serra, Barbara Lupo, Emanuela Dell'Aquila, Gianluigi Ferretti, Edoardo Pescarmona, Armando Bartolazzi, Giovanni Blandino, Livio Trusolino, Gianluca Bossi

Background: Colorectal cancer (CRC) is the third most common type of cancer and the second leading cause of cancer-related deaths worldwide, with a survival rate near to 10% when diagnosed at an advanced stage. Hence, the identification of new molecular targets to design more selective and efficient therapies is urgently required. The Mitogen activated protein kinase kinase 3 (MKK3) is a dual-specificity threonine/tyrosine protein kinase that, activated in response to cellular stress and inflammatory stimuli, regulates a plethora of biological processes. Previous studies revealed novel MKK3 roles in supporting tumor malignancy, as its depletion induces autophagy and cell death in cancer lines of different tumor types, including CRC. Therefore, MKK3 may represent an interesting new therapeutic target in advanced CRC, however selective MKK3 inhibitors are currently not available.

Methods: The study involved transcriptomic based drug repurposing approach and confirmatory assays with CRC lines, primary colonocytes and a subset of CRC patient-derived organoids (PDO). Investigations in vitro and in vivo were addressed.

Results: The repurposing approach identified the multitargeted kinase inhibitor AT9283 as a putative compound with MKK3 depletion-mimicking activities. Indeed, AT9283 drops phospho- and total-MKK3 protein levels in tested CRC models. Likely the MKK3 silencing, AT9283 treatment: i) inhibited cell proliferation promoting autophagy and cell death in tested CRC lines and PDOs; ii) resulted well-tolerated by CCD-18Co colonocytes; iii) reduced cancer cell motility inhibiting CRC cell migration and invasion; iv) inhibited COLO205 xenograft tumor growth. Mechanistically, AT9283 abrogated MKK3 protein levels mainly through the inhibition of aurora kinase A (AURKA), impacting on MKK3/AURKA protein-protein interaction and protein stability therefore uncovering the relevance of MKK3/AURKA crosstalk in sustaining CRC malignancy in vitro and in vivo.

Conclusion: Overall, we demonstrated that the anti-tumoral effects triggered by AT9283 treatment recapitulated the MKK3 depletion effects in all tested CRC models in vitro and in vivo, suggesting that AT9283 is a repurposed drug. According to its good tolerance when tested with primary colonocytes (CCD-18CO), AT9283 is a promising drug for the development of novel therapeutic strategies to target MKK3 oncogenic functions in late-stage and metastatic CRC patients.

背景:结直肠癌(CRC)是第三大常见癌症类型,也是全球癌症相关死亡的第二大原因,晚期确诊患者的存活率接近 10%。因此,迫切需要确定新的分子靶点,以设计更具选择性和更有效的疗法。丝裂原活化蛋白激酶激酶 3(MKK3)是一种具有双重特异性的苏氨酸/酪氨酸蛋白激酶,在细胞应激和炎症刺激下被激活,调节大量的生物过程。先前的研究揭示了 MKK3 在支持肿瘤恶性程度方面的新作用,因为在包括 CRC 在内的不同肿瘤类型的癌系中,消耗 MKK3 会诱导自噬和细胞死亡。因此,MKK3 可能是晚期 CRC 的一个有趣的新治疗靶点,但目前还没有选择性的 MKK3 抑制剂:研究涉及基于转录组学的药物再利用方法,以及对 CRC 株系、原代结肠细胞和 CRC 患者衍生的器官组织(PDO)子集的确证试验。研究涉及体外和体内研究:结果:再利用方法发现多靶点激酶抑制剂 AT9283 是一种具有 MKK3 缺失模拟活性的假定化合物。事实上,在测试的 CRC 模型中,AT9283 降低了磷酸化和总 MKK3 蛋白水平。可能是由于 MKK3 的沉默作用,AT9283 治疗:i)抑制细胞增殖,促进自噬和细胞死亡;ii)CCD-18Co 结肠细胞耐受性良好;iii)降低癌细胞的运动性,抑制 CRC 细胞的迁移和侵袭;iv)抑制 COLO205 异种移植肿瘤的生长。从机理上讲,AT9283主要通过抑制极光激酶A(AURKA)来降低MKK3蛋白水平,影响MKK3/AURKA蛋白-蛋白相互作用和蛋白稳定性,从而揭示了MKK3/AURKA串扰在体外和体内维持CRC恶性肿瘤的相关性:总之,我们证明了在所有测试的体外和体内 CRC 模型中,AT9283 治疗引发的抗肿瘤效应再现了 MKK3 的耗竭效应,这表明 AT9283 是一种再利用药物。AT9283在原代结肠细胞(CCD-18CO)测试中表现出良好的耐受性,因此是一种很有前景的药物,可用于开发针对晚期和转移性 CRC 患者 MKK3 致癌功能的新型治疗策略。
{"title":"Repurposed AT9283 triggers anti-tumoral effects by targeting MKK3 oncogenic functions in Colorectal Cancer.","authors":"Valentina Piastra, Federica Ganci, Andrea Sacconi, Angelina Pranteda, Matteo Allegretti, Roberta Bernardini, Martina Serra, Barbara Lupo, Emanuela Dell'Aquila, Gianluigi Ferretti, Edoardo Pescarmona, Armando Bartolazzi, Giovanni Blandino, Livio Trusolino, Gianluca Bossi","doi":"10.1186/s13046-024-03150-4","DOIUrl":"10.1186/s13046-024-03150-4","url":null,"abstract":"<p><strong>Background: </strong>Colorectal cancer (CRC) is the third most common type of cancer and the second leading cause of cancer-related deaths worldwide, with a survival rate near to 10% when diagnosed at an advanced stage. Hence, the identification of new molecular targets to design more selective and efficient therapies is urgently required. The Mitogen activated protein kinase kinase 3 (MKK3) is a dual-specificity threonine/tyrosine protein kinase that, activated in response to cellular stress and inflammatory stimuli, regulates a plethora of biological processes. Previous studies revealed novel MKK3 roles in supporting tumor malignancy, as its depletion induces autophagy and cell death in cancer lines of different tumor types, including CRC. Therefore, MKK3 may represent an interesting new therapeutic target in advanced CRC, however selective MKK3 inhibitors are currently not available.</p><p><strong>Methods: </strong>The study involved transcriptomic based drug repurposing approach and confirmatory assays with CRC lines, primary colonocytes and a subset of CRC patient-derived organoids (PDO). Investigations in vitro and in vivo were addressed.</p><p><strong>Results: </strong>The repurposing approach identified the multitargeted kinase inhibitor AT9283 as a putative compound with MKK3 depletion-mimicking activities. Indeed, AT9283 drops phospho- and total-MKK3 protein levels in tested CRC models. Likely the MKK3 silencing, AT9283 treatment: i) inhibited cell proliferation promoting autophagy and cell death in tested CRC lines and PDOs; ii) resulted well-tolerated by CCD-18Co colonocytes; iii) reduced cancer cell motility inhibiting CRC cell migration and invasion; iv) inhibited COLO205 xenograft tumor growth. Mechanistically, AT9283 abrogated MKK3 protein levels mainly through the inhibition of aurora kinase A (AURKA), impacting on MKK3/AURKA protein-protein interaction and protein stability therefore uncovering the relevance of MKK3/AURKA crosstalk in sustaining CRC malignancy in vitro and in vivo.</p><p><strong>Conclusion: </strong>Overall, we demonstrated that the anti-tumoral effects triggered by AT9283 treatment recapitulated the MKK3 depletion effects in all tested CRC models in vitro and in vivo, suggesting that AT9283 is a repurposed drug. According to its good tolerance when tested with primary colonocytes (CCD-18CO), AT9283 is a promising drug for the development of novel therapeutic strategies to target MKK3 oncogenic functions in late-stage and metastatic CRC patients.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"234"},"PeriodicalIF":11.4,"publicationDate":"2024-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11334304/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142009856","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
SelK promotes glioblastoma cell proliferation by inhibiting β-TrCP1 mediated ubiquitin-dependent degradation of CDK4. SelK通过抑制β-TrCP1介导的CDK4泛素依赖性降解,促进胶质母细胞瘤细胞增殖。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-19 DOI: 10.1186/s13046-024-03157-x
Jizhen Li, Lingling Zhao, Zerui Wu, Shirui Huang, Junyu Wang, Yuanyuan Chang, Li Liu, Honglei Jin, Jianglong Lu, Chuanshu Huang, Qipeng Xie, Haishan Huang, Zhipeng Su

Background: Glioblastoma (GB) is recognized as one of the most aggressive brain tumors, with a median survival of 14.6 months. However, there are still some patients whose survival time was greater than 3 years, and the biological reasons behind this clinical phenomenon arouse our research interests. By conducting proteomic analysis on tumor tissues obtained from GB patients who survived over 3 years compared to those who survived less than 1 year, we identified a significant upregulation of SelK in patients with shorter survival times. Therefore, we hypothesized that SelK may be an important indicator related to the occurrence and progression of GBM.

Methods: Proteomics and immunohistochemistry from GB patients were analyzed to investigate the correlation between SelK and clinical prognosis. Cellular phenotypes were evaluated by cell cycle analysis, cell viability assays, and xenograft models. Immunoblots and co-immunoprecipitation were conducted to verify SelK-mediated ubiquitin-dependent degradation of CDK4.

Results: SelK was found to be significantly upregulated in GB samples from short-term survivors (≤ 1 year) compared to those from long-term survivors (≥ 3 years), and its expression levels were negatively correlated with clinical prognosis. Knocking down of SelK expression reduced GB cell viability, induced G0/G1 phase arrest, and impaired the growth of transplanted glioma cells in nude mice. Down-regulation of SelK-induced ER stress leads to a reduction in the expression of SKP2 and an up-regulation of β-TrCP1 expression. Up-regulation of β-TrCP1, thereby accelerating the ubiquitin-dependent degradation of CDK4 and ultimately inhibiting the malignant proliferation of the GB cells.

Conclusion: This study discovered a significant increase in SelK expression in GB patients with poor prognosis, revealing a negative correlation between SelK expression and patient outcomes. Further mechanistic investigations revealed that SelK enhances the proliferation of GB cells by targeting the endoplasmic reticulum stress/SKP2/β-TrCP1/CDK4 axis.

背景:胶质母细胞瘤(GB)是公认的侵袭性最强的脑肿瘤之一,中位生存期为 14.6 个月。然而,仍有一些患者的生存期超过 3 年,这一临床现象背后的生物学原因引起了我们的研究兴趣。通过对存活时间超过 3 年的 GB 患者与存活时间不足 1 年的患者的肿瘤组织进行蛋白质组学分析,我们发现在存活时间较短的患者中,SelK 有明显的上调。因此,我们推测SelK可能是与GBM的发生和发展相关的一个重要指标:方法:对GB患者的蛋白质组学和免疫组化进行分析,研究SelK与临床预后的相关性。细胞表型通过细胞周期分析、细胞活力测定和异种移植模型进行评估。通过免疫印迹和共免疫沉淀来验证SelK介导的泛素依赖性CDK4降解:结果:与长期存活者(≥3年)相比,SelK在短期存活者(≤1年)的GB样本中明显上调,其表达水平与临床预后呈负相关。敲除SelK的表达可降低GB细胞的存活率,诱导G0/G1期停滞,并损害裸鼠移植胶质瘤细胞的生长。SelK诱导的ER应激的下调导致SKP2表达的减少和β-TrCP1表达的上调。β-TrCP1的上调,从而加速CDK4的泛素依赖性降解,最终抑制GB细胞的恶性增殖:本研究发现,在预后不良的GB患者中,SelK的表达明显增加,揭示了SelK表达与患者预后之间的负相关。进一步的机理研究发现,SelK通过靶向内质网应激/SKP2/β-TrCP1/CDK4轴来增强GB细胞的增殖。
{"title":"SelK promotes glioblastoma cell proliferation by inhibiting β-TrCP1 mediated ubiquitin-dependent degradation of CDK4.","authors":"Jizhen Li, Lingling Zhao, Zerui Wu, Shirui Huang, Junyu Wang, Yuanyuan Chang, Li Liu, Honglei Jin, Jianglong Lu, Chuanshu Huang, Qipeng Xie, Haishan Huang, Zhipeng Su","doi":"10.1186/s13046-024-03157-x","DOIUrl":"10.1186/s13046-024-03157-x","url":null,"abstract":"<p><strong>Background: </strong>Glioblastoma (GB) is recognized as one of the most aggressive brain tumors, with a median survival of 14.6 months. However, there are still some patients whose survival time was greater than 3 years, and the biological reasons behind this clinical phenomenon arouse our research interests. By conducting proteomic analysis on tumor tissues obtained from GB patients who survived over 3 years compared to those who survived less than 1 year, we identified a significant upregulation of SelK in patients with shorter survival times. Therefore, we hypothesized that SelK may be an important indicator related to the occurrence and progression of GBM.</p><p><strong>Methods: </strong>Proteomics and immunohistochemistry from GB patients were analyzed to investigate the correlation between SelK and clinical prognosis. Cellular phenotypes were evaluated by cell cycle analysis, cell viability assays, and xenograft models. Immunoblots and co-immunoprecipitation were conducted to verify SelK-mediated ubiquitin-dependent degradation of CDK4.</p><p><strong>Results: </strong>SelK was found to be significantly upregulated in GB samples from short-term survivors (≤ 1 year) compared to those from long-term survivors (≥ 3 years), and its expression levels were negatively correlated with clinical prognosis. Knocking down of SelK expression reduced GB cell viability, induced G0/G1 phase arrest, and impaired the growth of transplanted glioma cells in nude mice. Down-regulation of SelK-induced ER stress leads to a reduction in the expression of SKP2 and an up-regulation of β-TrCP1 expression. Up-regulation of β-TrCP1, thereby accelerating the ubiquitin-dependent degradation of CDK4 and ultimately inhibiting the malignant proliferation of the GB cells.</p><p><strong>Conclusion: </strong>This study discovered a significant increase in SelK expression in GB patients with poor prognosis, revealing a negative correlation between SelK expression and patient outcomes. Further mechanistic investigations revealed that SelK enhances the proliferation of GB cells by targeting the endoplasmic reticulum stress/SKP2/β-TrCP1/CDK4 axis.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"231"},"PeriodicalIF":11.4,"publicationDate":"2024-08-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11331741/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142001179","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting treatment resistance: unveiling the potential of RNA methylation regulators and TG-101,209 in pan-cancer neoadjuvant therapy. 靶向治疗耐药性:揭示 RNA 甲基化调节剂和 TG-101,209 在泛癌症新辅助治疗中的潜力。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-19 DOI: 10.1186/s13046-024-03111-x
Yaoyao Zhou, Ziyun Liu, Cheng Gong, Jie Zhang, Jing Zhao, Xia Zhang, Xiangyu Liu, Bin Li, Rui Li, Zhenyu Shi, Yongjie Xie, Li Bao

Background: Tumor recurrence and mortality rates remain challenging in cancer patients despite comprehensive treatment. Neoadjuvant chemotherapy and immunotherapy aim to eliminate residual tumor cells, reducing the risk of recurrence. However, drug resistance during neoadjuvant therapy is a significant hurdle. Recent studies suggest a correlation between RNA methylation regulators (RMRs) and response to neoadjuvant therapy.

Methods: Using a multi-center approach, we integrated advanced techniques such as single-cell transcriptomics, whole-genome sequencing, RNA sequencing, proteomics, machine learning, and in vivo/in vitro experiments. Analyzing pan-cancer cohorts, the association between neoadjuvant chemotherapy/immunotherapy effectiveness and RNA methylation using single-cell sequencing was investigated. Multi-omics analysis and machine learning algorithms identified genomic variations, transcriptional dysregulation, and prognostic relevance of RMRs, revealing distinct molecular subtypes guiding pan-cancer neoadjuvant therapy stratification.

Results: Our analysis unveiled a strong link between neoadjuvant therapy efficacy and RNA methylation dynamics, supported by pan-cancer single-cell sequencing data. Integration of omics data and machine learning algorithms identified RMR genomic variations, transcriptional dysregulation, and prognostic implications in pan-cancer. High-RMR-expressing tumors displayed increased genomic alterations, an immunosuppressive microenvironment, poorer prognosis, and resistance to neoadjuvant therapy. Molecular investigations and in vivo/in vitro experiments have substantiated that the JAK inhibitor TG-101,209 exerts notable effects on the immune microenvironment of tumors, rendering high-RMR-expressing pan-cancer tumors, particularly in pancreatic cancer, more susceptible to chemotherapy and immunotherapy.

Conclusions: This study emphasizes the pivotal role of RMRs in pan-cancer neoadjuvant therapy, serving as predictive biomarkers for monitoring the tumor microenvironment, patient prognosis, and therapeutic response. Distinct molecular subtypes of RMRs aid individualized stratification in neoadjuvant therapy. Combining TG-101,209 adjuvant therapy presents a promising strategy to enhance the sensitivity of high-RMR-expressing tumors to chemotherapy and immunotherapy. However, further validation studies are necessary to fully understand the clinical utility of RNA methylation regulators and their impact on patient outcomes.

背景:尽管进行了综合治疗,但癌症患者的肿瘤复发率和死亡率仍然很高。新辅助化疗和免疫疗法旨在消除残余肿瘤细胞,降低复发风险。然而,新辅助治疗期间的耐药性是一个重大障碍。最近的研究表明,RNA甲基化调节因子(RMRs)与新辅助治疗反应之间存在相关性:我们采用多中心方法,整合了单细胞转录组学、全基因组测序、RNA测序、蛋白质组学、机器学习和体内/体外实验等先进技术。通过分析泛癌症队列,利用单细胞测序研究了新辅助化疗/免疫治疗效果与RNA甲基化之间的关联。多组学分析和机器学习算法确定了RMRs的基因组变异、转录失调和预后相关性,揭示了指导泛癌新辅助治疗分层的不同分子亚型:在泛癌症单细胞测序数据的支持下,我们的分析揭示了新辅助疗法疗效与RNA甲基化动态之间的密切联系。通过整合omics数据和机器学习算法,我们发现了RMR基因组变异、转录失调以及对泛癌预后的影响。高RMR表达的肿瘤显示出更多的基因组改变、免疫抑制微环境、较差的预后以及对新辅助治疗的耐药性。分子研究和体内/体外实验证实,JAK抑制剂TG-101,209对肿瘤的免疫微环境有显著影响,使高RMR表达的泛癌肿瘤,尤其是胰腺癌,更容易接受化疗和免疫治疗:这项研究强调了RMRs在泛癌症新辅助治疗中的关键作用,它是监测肿瘤微环境、患者预后和治疗反应的预测性生物标志物。RMRs的不同分子亚型有助于新辅助治疗中的个体化分层。结合 TG-101,209 辅助治疗是提高高 RMR 表达肿瘤对化疗和免疫疗法敏感性的一种有前途的策略。然而,要充分了解 RNA 甲基化调节剂的临床用途及其对患者预后的影响,还需要进一步的验证研究。
{"title":"Targeting treatment resistance: unveiling the potential of RNA methylation regulators and TG-101,209 in pan-cancer neoadjuvant therapy.","authors":"Yaoyao Zhou, Ziyun Liu, Cheng Gong, Jie Zhang, Jing Zhao, Xia Zhang, Xiangyu Liu, Bin Li, Rui Li, Zhenyu Shi, Yongjie Xie, Li Bao","doi":"10.1186/s13046-024-03111-x","DOIUrl":"10.1186/s13046-024-03111-x","url":null,"abstract":"<p><strong>Background: </strong>Tumor recurrence and mortality rates remain challenging in cancer patients despite comprehensive treatment. Neoadjuvant chemotherapy and immunotherapy aim to eliminate residual tumor cells, reducing the risk of recurrence. However, drug resistance during neoadjuvant therapy is a significant hurdle. Recent studies suggest a correlation between RNA methylation regulators (RMRs) and response to neoadjuvant therapy.</p><p><strong>Methods: </strong>Using a multi-center approach, we integrated advanced techniques such as single-cell transcriptomics, whole-genome sequencing, RNA sequencing, proteomics, machine learning, and in vivo/in vitro experiments. Analyzing pan-cancer cohorts, the association between neoadjuvant chemotherapy/immunotherapy effectiveness and RNA methylation using single-cell sequencing was investigated. Multi-omics analysis and machine learning algorithms identified genomic variations, transcriptional dysregulation, and prognostic relevance of RMRs, revealing distinct molecular subtypes guiding pan-cancer neoadjuvant therapy stratification.</p><p><strong>Results: </strong>Our analysis unveiled a strong link between neoadjuvant therapy efficacy and RNA methylation dynamics, supported by pan-cancer single-cell sequencing data. Integration of omics data and machine learning algorithms identified RMR genomic variations, transcriptional dysregulation, and prognostic implications in pan-cancer. High-RMR-expressing tumors displayed increased genomic alterations, an immunosuppressive microenvironment, poorer prognosis, and resistance to neoadjuvant therapy. Molecular investigations and in vivo/in vitro experiments have substantiated that the JAK inhibitor TG-101,209 exerts notable effects on the immune microenvironment of tumors, rendering high-RMR-expressing pan-cancer tumors, particularly in pancreatic cancer, more susceptible to chemotherapy and immunotherapy.</p><p><strong>Conclusions: </strong>This study emphasizes the pivotal role of RMRs in pan-cancer neoadjuvant therapy, serving as predictive biomarkers for monitoring the tumor microenvironment, patient prognosis, and therapeutic response. Distinct molecular subtypes of RMRs aid individualized stratification in neoadjuvant therapy. Combining TG-101,209 adjuvant therapy presents a promising strategy to enhance the sensitivity of high-RMR-expressing tumors to chemotherapy and immunotherapy. However, further validation studies are necessary to fully understand the clinical utility of RNA methylation regulators and their impact on patient outcomes.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"232"},"PeriodicalIF":11.4,"publicationDate":"2024-08-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11331809/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142005751","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Correction: Different p53 genotypes regulating different phosphorylation sites and subcellular location of CDC25C associated with the formation of polyploid giant cancer cells. 更正:调节 CDC25C 不同磷酸化位点和亚细胞位置的不同 p53 基因型与多倍体巨型癌细胞的形成有关。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-17 DOI: 10.1186/s13046-024-03155-z
Kai Liu, Minying Zheng, Qi Zhao, Kexin Zhang, Zugui Li, Fangmei Fu, Hao Zhang, Jiaxing Du, Yuwei Li, Shiwu Zhang
{"title":"Correction: Different p53 genotypes regulating different phosphorylation sites and subcellular location of CDC25C associated with the formation of polyploid giant cancer cells.","authors":"Kai Liu, Minying Zheng, Qi Zhao, Kexin Zhang, Zugui Li, Fangmei Fu, Hao Zhang, Jiaxing Du, Yuwei Li, Shiwu Zhang","doi":"10.1186/s13046-024-03155-z","DOIUrl":"10.1186/s13046-024-03155-z","url":null,"abstract":"","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"229"},"PeriodicalIF":11.4,"publicationDate":"2024-08-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11329984/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141996802","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Correction: Deliberation concerning the role of M1-type macrophage subset in oral carcinogenesis. 更正:关于 M1 型巨噬细胞亚群在口腔癌发生中的作用的讨论。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-17 DOI: 10.1186/s13046-024-03162-0
Chen-Xi Li, Zhong-Cheng Gong, Jing-Wen Yu
{"title":"Correction: Deliberation concerning the role of M1-type macrophage subset in oral carcinogenesis.","authors":"Chen-Xi Li, Zhong-Cheng Gong, Jing-Wen Yu","doi":"10.1186/s13046-024-03162-0","DOIUrl":"10.1186/s13046-024-03162-0","url":null,"abstract":"","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"228"},"PeriodicalIF":11.4,"publicationDate":"2024-08-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11330006/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141996801","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
tsRNA-GlyGCC promotes colorectal cancer progression and 5-FU resistance by regulating SPIB. tsRNA-GlyGCC通过调节SPIB促进结直肠癌的进展和5-FU耐药性。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-17 DOI: 10.1186/s13046-024-03132-6
Rong Xu, Ashuai Du, Xinpei Deng, Wei Du, Kaiying Zhang, Jianbo Li, Yingxue Lu, Xiaoli Wei, Qinglong Yang, Hailin Tang

Background: tRNA-derived small RNAs (tsRNAs) are newly discovered non-coding RNA, which are generated from tRNAs and are reported to participate in several biological processes in diseases, especially cancer; however, the mechanism of tsRNA involvement in colorectal cancer (CRC) and 5-fluorouracil (5-FU) is still unclear.

Methods: RNA sequencing was performed to identify differential expression of tsRNAs in CRC tissues. CCK8, colony formation, transwell assays, and tumor sphere assays were used to investigate the role of tsRNA-GlyGCC in 5-FU resistance in CRC. TargetScan and miRanda were used to identify the target genes of tsRNA-GlyGCC. Biotin pull-down, RNA pull-down, luciferase assay, ChIP, and western blotting were used to explore the underlying molecular mechanisms of action of tsRNA-GlyGCC. The MeRIP assay was used to investigate the N(7)-methylguanosine RNA modification of tsRNA-GlyGCC.

Results: In this study, we uncovered the feature of tsRNAs in human CRC tissues and confirmed a specific 5' half tRNA, 5'tiRNA-Gly-GCC (tsRNA-GlyGCC), which is upregulated in CRC tissues and modulated by METTL1-mediated N(7)-methylguanosine tRNA modification. In vitro and in vivo experiments revealed the oncogenic role of tsRNA-GlyGCC in 5-FU drug resistance in CRC. Remarkably, our results showed that tsRNA-GlyGCC modulated the JAK1/STAT6 signaling pathway by targeting SPIB. Poly (β-amino esters) were synthesized to assist the delivery of 5-FU and tsRNA-GlyGCC inhibitor, which effectively inhibited tumor growth and enhanced CRC sensitive to 5-FU without obvious adverse effects in subcutaneous tumor.

Conclusions: Our study revealed a specific tsRNA-GlyGCC-engaged pathway in CRC progression. Targeting tsRNA-GlyGCC in combination with 5-FU may provide a promising nanotherapeutic strategy for the treatment of 5-FU-resistance CRC.

背景:tRNA衍生的小RNA(tsRNAs)是新发现的非编码RNA,由tRNAs产生,据报道参与多种疾病的生物学过程,尤其是癌症;然而,tsRNA参与结直肠癌(CRC)和5-氟尿嘧啶(5-FU)的机制仍不清楚:方法:进行 RNA 测序以确定 tsRNA 在 CRC 组织中的差异表达。采用 CCK8、菌落形成、transwell 试验和肿瘤球试验研究 tsRNA-GlyGCC 在 CRC 耐 5-FU 中的作用。利用 TargetScan 和 miRanda 鉴定 tsRNA-GlyGCC 的靶基因。生物素牵引、RNA牵引、荧光素酶检测、ChIP和Western印迹被用来探索tsRNA-GlyGCC的潜在分子作用机制。MeRIP试验用于研究tsRNA-GlyGCC的N(7)-甲基鸟苷RNA修饰:本研究揭示了人 CRC 组织中 tsRNAs 的特征,并证实了一种特异的 5' 半 tRNA,即 5'tiRNA-Gly-GCC (tsRNA-GlyGCC),它在 CRC 组织中上调,并受 METTL1 介导的 N(7)-methylguanosine tRNA 修饰的调节。体外和体内实验揭示了 tsRNA-GlyGCC 在 5-FU 耐药性中的致癌作用。值得注意的是,我们的研究结果表明,tsRNA-GlyGCC通过靶向SPIB调节了JAK1/STAT6信号通路。通过合成聚(β-氨基酯)来辅助递送5-FU和tsRNA-GlyGCC抑制剂,可有效抑制肿瘤生长,提高CRC对5-FU的敏感性,且对皮下肿瘤无明显不良反应:我们的研究揭示了tsRNA-GlyGCC在CRC进展中的特异性参与途径。结论:我们的研究揭示了tsRNA-GlyGCC在CRC进展过程中的特异性参与途径,靶向tsRNA-GlyGCC与5-FU联用可为5-FU耐药CRC的治疗提供一种前景广阔的纳米治疗策略。
{"title":"tsRNA-GlyGCC promotes colorectal cancer progression and 5-FU resistance by regulating SPIB.","authors":"Rong Xu, Ashuai Du, Xinpei Deng, Wei Du, Kaiying Zhang, Jianbo Li, Yingxue Lu, Xiaoli Wei, Qinglong Yang, Hailin Tang","doi":"10.1186/s13046-024-03132-6","DOIUrl":"10.1186/s13046-024-03132-6","url":null,"abstract":"<p><strong>Background: </strong>tRNA-derived small RNAs (tsRNAs) are newly discovered non-coding RNA, which are generated from tRNAs and are reported to participate in several biological processes in diseases, especially cancer; however, the mechanism of tsRNA involvement in colorectal cancer (CRC) and 5-fluorouracil (5-FU) is still unclear.</p><p><strong>Methods: </strong>RNA sequencing was performed to identify differential expression of tsRNAs in CRC tissues. CCK8, colony formation, transwell assays, and tumor sphere assays were used to investigate the role of tsRNA-GlyGCC in 5-FU resistance in CRC. TargetScan and miRanda were used to identify the target genes of tsRNA-GlyGCC. Biotin pull-down, RNA pull-down, luciferase assay, ChIP, and western blotting were used to explore the underlying molecular mechanisms of action of tsRNA-GlyGCC. The MeRIP assay was used to investigate the N(7)-methylguanosine RNA modification of tsRNA-GlyGCC.</p><p><strong>Results: </strong>In this study, we uncovered the feature of tsRNAs in human CRC tissues and confirmed a specific 5' half tRNA, 5'tiRNA-Gly-GCC (tsRNA-GlyGCC), which is upregulated in CRC tissues and modulated by METTL1-mediated N(7)-methylguanosine tRNA modification. In vitro and in vivo experiments revealed the oncogenic role of tsRNA-GlyGCC in 5-FU drug resistance in CRC. Remarkably, our results showed that tsRNA-GlyGCC modulated the JAK1/STAT6 signaling pathway by targeting SPIB. Poly (β-amino esters) were synthesized to assist the delivery of 5-FU and tsRNA-GlyGCC inhibitor, which effectively inhibited tumor growth and enhanced CRC sensitive to 5-FU without obvious adverse effects in subcutaneous tumor.</p><p><strong>Conclusions: </strong>Our study revealed a specific tsRNA-GlyGCC-engaged pathway in CRC progression. Targeting tsRNA-GlyGCC in combination with 5-FU may provide a promising nanotherapeutic strategy for the treatment of 5-FU-resistance CRC.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"230"},"PeriodicalIF":11.4,"publicationDate":"2024-08-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11330149/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141996803","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Journal of Experimental & Clinical Cancer Research
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