首页 > 最新文献

Journal of Experimental & Clinical Cancer Research最新文献

英文 中文
Progesterone boosts abiraterone-driven target and NK cell therapies against glioblastoma. 黄体酮可促进阿比特龙驱动的靶向和NK细胞疗法对抗胶质母细胞瘤。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-06 DOI: 10.1186/s13046-024-03144-2
Hsien-Chung Chen, Hong-Yi Lin, Yung-Hsiao Chiang, Wen-Bin Yang, Chung-Han Wang, Pei-Yu Yang, Siou-Lian Hu, Tsung-I Hsu

Introduction: Glioblastoma (GBM) poses a significant challenge in oncology, with median survival times barely extending beyond a year due to resistance to standard therapies like temozolomide (TMZ). This study introduces a novel therapeutic strategy combining progesterone (Prog) and abiraterone (Abi) aimed at enhancing GBM treatment efficacy by modulating the tumor microenvironment and augmenting NK cell-mediated immunity.

Methods: We employed in vitro and in vivo GBM models to assess the effects of Prog and Abi on cell viability, proliferation, apoptosis, and the immune microenvironment. Techniques included cell viability assays, Glo-caspase 3/7 apoptosis assays, RNA-seq and qPCR for gene expression, Seahorse analysis for mitochondrial function, HPLC-MS for metabolomics analysis, and immune analysis by flow cytometry to quantify NK cell infiltration.

Results: Prog significantly reduced the IC50 of Abi in TMZ-resistant GBM cell, suggesting the enhanced cytotoxicity. Treatment induced greater apoptosis than either agent alone, suppressed tumor growth, and prolonged survival in mouse models. Notably, there was an increase in CD3-/CD19-/CD56+/NK1.1+ NK cell infiltration in treated tumors, indicating a shift towards an anti-tumor immune microenvironment. The combination therapy also resulted in a reduction of MGMT expression and a suppression of mitochondrial respiration and glycolysis in GBM cells.

Conclusion: The combination of Prog and Abi represents a promising therapeutic approach for GBM, showing potential in suppressing tumor growth, extending survival, and modulating the immune microenvironment. These findings warrant further exploration into the clinical applicability of this strategy to improve outcomes for GBM patients.

简介胶质母细胞瘤(GBM)是肿瘤学领域的一项重大挑战,由于对替莫唑胺(TMZ)等标准疗法产生耐药性,中位生存时间几乎不超过一年。本研究介绍了一种结合黄体酮(Prog)和阿比特龙(Abi)的新型治疗策略,旨在通过调节肿瘤微环境和增强NK细胞介导的免疫力来提高GBM的治疗效果:我们采用体外和体内 GBM 模型来评估 Prog 和 Abi 对细胞活力、增殖、凋亡和免疫微环境的影响。技术包括细胞活力测定、Glo-caspase 3/7凋亡测定、基因表达的RNA-seq和qPCR、线粒体功能的Seahorse分析、代谢组学分析的HPLC-MS,以及通过流式细胞术量化NK细胞浸润的免疫分析:结果:Prog明显降低了阿比在TMZ耐药GBM细胞中的IC50,表明其细胞毒性增强。与单独使用两种药物相比,Prog 能诱导更多的细胞凋亡,抑制肿瘤生长,延长小鼠模型的存活时间。值得注意的是,经治疗的肿瘤中 CD3-/CD19-/CD56+/NK1.1+ NK 细胞浸润增加,表明向抗肿瘤免疫微环境转变。联合疗法还降低了 MGMT 的表达,抑制了 GBM 细胞的线粒体呼吸和糖酵解:Prog和Abi的联合疗法是一种治疗GBM的有前途的方法,在抑制肿瘤生长、延长生存期和调节免疫微环境方面显示出潜力。这些发现值得进一步探索这一策略的临床适用性,以改善 GBM 患者的预后。
{"title":"Progesterone boosts abiraterone-driven target and NK cell therapies against glioblastoma.","authors":"Hsien-Chung Chen, Hong-Yi Lin, Yung-Hsiao Chiang, Wen-Bin Yang, Chung-Han Wang, Pei-Yu Yang, Siou-Lian Hu, Tsung-I Hsu","doi":"10.1186/s13046-024-03144-2","DOIUrl":"10.1186/s13046-024-03144-2","url":null,"abstract":"<p><strong>Introduction: </strong>Glioblastoma (GBM) poses a significant challenge in oncology, with median survival times barely extending beyond a year due to resistance to standard therapies like temozolomide (TMZ). This study introduces a novel therapeutic strategy combining progesterone (Prog) and abiraterone (Abi) aimed at enhancing GBM treatment efficacy by modulating the tumor microenvironment and augmenting NK cell-mediated immunity.</p><p><strong>Methods: </strong>We employed in vitro and in vivo GBM models to assess the effects of Prog and Abi on cell viability, proliferation, apoptosis, and the immune microenvironment. Techniques included cell viability assays, Glo-caspase 3/7 apoptosis assays, RNA-seq and qPCR for gene expression, Seahorse analysis for mitochondrial function, HPLC-MS for metabolomics analysis, and immune analysis by flow cytometry to quantify NK cell infiltration.</p><p><strong>Results: </strong>Prog significantly reduced the IC50 of Abi in TMZ-resistant GBM cell, suggesting the enhanced cytotoxicity. Treatment induced greater apoptosis than either agent alone, suppressed tumor growth, and prolonged survival in mouse models. Notably, there was an increase in CD3<sup>-</sup>/CD19<sup>-</sup>/CD56<sup>+</sup>/NK1.1<sup>+</sup> NK cell infiltration in treated tumors, indicating a shift towards an anti-tumor immune microenvironment. The combination therapy also resulted in a reduction of MGMT expression and a suppression of mitochondrial respiration and glycolysis in GBM cells.</p><p><strong>Conclusion: </strong>The combination of Prog and Abi represents a promising therapeutic approach for GBM, showing potential in suppressing tumor growth, extending survival, and modulating the immune microenvironment. These findings warrant further exploration into the clinical applicability of this strategy to improve outcomes for GBM patients.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"218"},"PeriodicalIF":11.4,"publicationDate":"2024-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11302026/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141894766","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
Regulatory role of lncH19 in RAC1 alternative splicing: implication for RAC1B expression in colorectal cancer. lncH19 在 RAC1 替代剪接中的调控作用:对结直肠癌中 RAC1B 表达的影响。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-05 DOI: 10.1186/s13046-024-03139-z
Aurora Cordaro, Maria Magdalena Barreca, Chiara Zichittella, Marco Loria, Denise Anello, Goffredo Arena, Nicolina Sciaraffa, Claudia Coronnello, Giuseppe Pizzolanti, Riccardo Alessandro, Alice Conigliaro

Aberrant alternative splicing events play a critical role in cancer biology, contributing to tumor invasion, metastasis, epithelial-mesenchymal transition, and drug resistance. Recent studies have shown that alternative splicing is a key feature for transcriptomic variations in colorectal cancer, which ranks third among malignant tumors worldwide in both incidence and mortality. Long non-coding RNAs can modulate this process by acting as trans-regulatory agents, recruiting splicing factors, or driving them to specific targeted genes. LncH19 is a lncRNA dis-regulated in several tumor types and, in colorectal cancer, it plays a critical role in tumor onset, progression, and metastasis. In this paper, we found, that in colorectal cancer cells, the long non-coding RNA H19 can bind immature RNAs and splicing factors as hnRNPM and RBFOX2. Through bioinformatic analysis, we identified 57 transcripts associated with lncH19 and containing binding sites for both splicing factors, hnRNPM, and RBFOX2. Among these transcripts, we identified the mRNA of the GTPase-RAC1, whose alternatively spliced isoform, RAC1B, has been ascribed several roles in the malignant transformation. We confirmed, in vitro, the binding of the splicing factors to both the transcripts RAC1 and lncH19. Loss and gain of expression experiments in two colorectal cancer cell lines (SW620 and HCT116) demonstrated that lncH19 is required for RAC1B expression and, through RAC1B, it induces c-Myc and Cyclin-D increase. In vivo, investigation from biopsies of colorectal cancer patients showed higher levels of all the explored genes (lncH19, RAC1B, c-Myc and Cyclin-D) concerning the healthy counterpart, thus supporting our in vitro model. In addition, we identified a positive correlation between lncH19 and RAC1B in colorectal cancer patients. Finally, we demonstrated that lncH19, as a shuttle, drives the splicing factors RBFOX2 and hnRNPM to RAC1 allowing exon retention and RAC1B expression. The data shown in this paper represent the first evidence of a new mechanism of action by which lncH19 carries out its functions as an oncogene by prompting colorectal cancer through the modulation of alternative splicing.

畸变的替代剪接事件在癌症生物学中起着至关重要的作用,有助于肿瘤的侵袭、转移、上皮-间质转化和耐药性。最近的研究表明,替代剪接是结直肠癌转录组变异的一个关键特征,而结直肠癌的发病率和死亡率在全球恶性肿瘤中均排名第三。长非编码 RNA 可作为反式调节剂、招募剪接因子或将其驱动到特定的目标基因上,从而调节这一过程。LncH19 是一种在多种肿瘤类型中失调的 lncRNA,在结直肠癌中,它在肿瘤的发生、发展和转移中起着关键作用。在本文中,我们发现在结直肠癌细胞中,长非编码 RNA H19 可以结合未成熟 RNA 和剪接因子 hnRNPM 和 RBFOX2。通过生物信息学分析,我们发现了 57 个与 lncH19 相关的转录本,这些转录本含有剪接因子 hnRNPM 和 RBFOX2 的结合位点。在这些转录本中,我们发现了 GTPase-RAC1 的 mRNA,其另类剪接异构体 RAC1B 在恶性转化中发挥了多种作用。我们在体外证实了剪接因子与转录本 RAC1 和 lncH19 的结合。在两种结直肠癌细胞系(SW620 和 HCT116)中进行的表达缺失和表达增益实验表明,lncH19 是 RAC1B 表达所必需的,它通过 RAC1B 诱导 c-Myc 和 Cyclin-D 的增加。在体内,对结直肠癌患者活组织的调查显示,与健康人相比,所有研究基因(lncH19、RAC1B、c-Myc 和 Cyclin-D)的水平都更高,从而支持了我们的体外模型。此外,我们还发现结直肠癌患者体内的 lncH19 与 RAC1B 呈正相关。最后,我们证明,lncH19 作为一种穿梭器,可将剪接因子 RBFOX2 和 hnRNPM 驱动到 RAC1 上,从而实现外显子保留和 RAC1B 表达。本文显示的数据首次证明了一种新的作用机制,即lncH19通过调节替代剪接促使结直肠癌的发生,从而发挥其作为致癌基因的功能。
{"title":"Regulatory role of lncH19 in RAC1 alternative splicing: implication for RAC1B expression in colorectal cancer.","authors":"Aurora Cordaro, Maria Magdalena Barreca, Chiara Zichittella, Marco Loria, Denise Anello, Goffredo Arena, Nicolina Sciaraffa, Claudia Coronnello, Giuseppe Pizzolanti, Riccardo Alessandro, Alice Conigliaro","doi":"10.1186/s13046-024-03139-z","DOIUrl":"10.1186/s13046-024-03139-z","url":null,"abstract":"<p><p>Aberrant alternative splicing events play a critical role in cancer biology, contributing to tumor invasion, metastasis, epithelial-mesenchymal transition, and drug resistance. Recent studies have shown that alternative splicing is a key feature for transcriptomic variations in colorectal cancer, which ranks third among malignant tumors worldwide in both incidence and mortality. Long non-coding RNAs can modulate this process by acting as trans-regulatory agents, recruiting splicing factors, or driving them to specific targeted genes. LncH19 is a lncRNA dis-regulated in several tumor types and, in colorectal cancer, it plays a critical role in tumor onset, progression, and metastasis. In this paper, we found, that in colorectal cancer cells, the long non-coding RNA H19 can bind immature RNAs and splicing factors as hnRNPM and RBFOX2. Through bioinformatic analysis, we identified 57 transcripts associated with lncH19 and containing binding sites for both splicing factors, hnRNPM, and RBFOX2. Among these transcripts, we identified the mRNA of the GTPase-RAC1, whose alternatively spliced isoform, RAC1B, has been ascribed several roles in the malignant transformation. We confirmed, in vitro, the binding of the splicing factors to both the transcripts RAC1 and lncH19. Loss and gain of expression experiments in two colorectal cancer cell lines (SW620 and HCT116) demonstrated that lncH19 is required for RAC1B expression and, through RAC1B, it induces c-Myc and Cyclin-D increase. In vivo, investigation from biopsies of colorectal cancer patients showed higher levels of all the explored genes (lncH19, RAC1B, c-Myc and Cyclin-D) concerning the healthy counterpart, thus supporting our in vitro model. In addition, we identified a positive correlation between lncH19 and RAC1B in colorectal cancer patients. Finally, we demonstrated that lncH19, as a shuttle, drives the splicing factors RBFOX2 and hnRNPM to RAC1 allowing exon retention and RAC1B expression. The data shown in this paper represent the first evidence of a new mechanism of action by which lncH19 carries out its functions as an oncogene by prompting colorectal cancer through the modulation of alternative splicing.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"217"},"PeriodicalIF":11.4,"publicationDate":"2024-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11299361/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141890689","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
Examining the evidence for mutual modulation between m6A modification and circular RNAs: current knowledge and future prospects. 研究 m6A 修饰与环状 RNA 之间相互调节的证据:现有知识与未来展望。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-03 DOI: 10.1186/s13046-024-03136-2
Xiaozhu Tang, Mengjie Guo, Yuanjiao Zhang, Junxian Lv, Chunyan Gu, Ye Yang

The resistance of cancer cells to treatment significantly impedes the success of therapy, leading to the recurrence of various types of cancers. Understanding the specific mechanisms of therapy resistance may offer novel approaches for alleviating drug resistance in cancer. Recent research has shown a reciprocal relationship between circular RNAs (circRNAs) and N6-methyladenosine (m6A) modification, and their interaction can affect the resistance and sensitivity of cancer therapy. This review aims to summarize the latest developments in the m6A modification of circRNAs and their importance in regulating therapy resistance in cancer. Furthermore, we explore their mutual interaction and exact mechanisms and provide insights into potential future approaches for reversing cancer resistance.

癌细胞对治疗的耐药性极大地阻碍了治疗的成功,导致各类癌症复发。了解抗药性的具体机制可为缓解癌症抗药性提供新方法。最近的研究表明,环状 RNA(circRNA)和 N6-甲基腺苷(m6A)修饰之间存在相互影响的关系,它们之间的相互作用会影响癌症治疗的耐药性和敏感性。本综述旨在总结循环 RNA 的 m6A 修饰及其在调节癌症耐药性方面的重要性的最新进展。此外,我们还探讨了它们之间的相互作用和确切机制,并对未来逆转癌症耐药性的潜在方法提出了见解。
{"title":"Examining the evidence for mutual modulation between m6A modification and circular RNAs: current knowledge and future prospects.","authors":"Xiaozhu Tang, Mengjie Guo, Yuanjiao Zhang, Junxian Lv, Chunyan Gu, Ye Yang","doi":"10.1186/s13046-024-03136-2","DOIUrl":"10.1186/s13046-024-03136-2","url":null,"abstract":"<p><p>The resistance of cancer cells to treatment significantly impedes the success of therapy, leading to the recurrence of various types of cancers. Understanding the specific mechanisms of therapy resistance may offer novel approaches for alleviating drug resistance in cancer. Recent research has shown a reciprocal relationship between circular RNAs (circRNAs) and N6-methyladenosine (m6A) modification, and their interaction can affect the resistance and sensitivity of cancer therapy. This review aims to summarize the latest developments in the m6A modification of circRNAs and their importance in regulating therapy resistance in cancer. Furthermore, we explore their mutual interaction and exact mechanisms and provide insights into potential future approaches for reversing cancer resistance.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"216"},"PeriodicalIF":11.4,"publicationDate":"2024-08-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11297759/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141879677","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 landscape of circulating tumor HPV DNA and TTMV-HPVDNA for surveillance of HPV-oropharyngeal carcinoma: systematic review and meta-analysis. 循环肿瘤 HPV DNA 和 TTMV-HPVDNA 监测 HPV 口咽癌的前景:系统回顾和荟萃分析。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-03 DOI: 10.1186/s13046-024-03137-1
Flaminia Campo, Oreste Iocca, Francesca Paolini, Valentina Manciocco, Silvia Moretto, Armando De Virgilio, Claudio Moretti, Antonello Vidiri, Aldo Venuti, Paolo Bossi, Giovanni Blandino, Raul Pellini

Background: Human papilloma virus (HPV) related cancers of the oropharynx are rapidly increasing in incidence and may soon represent the majority of all head and neck cancers. Improved monitoring and surveillance methods are thus an urgent need in public health.

Main text: The goal is to highlight the current potential and limitations of liquid biopsy through a meta analytic study on ctHPVDNA and TTMV-HPVDNA. It was performed a Literature search on articles published until December 2023 using three different databases: MEDLINE, Embase, and Cochrane Library. Studies that evaluated post-treatment ctHPVDNA and TTMV-HPVDNA in patients with HPV + OPSCC, studies reporting complete data on the diagnostic accuracy in recurrence, or in which the number of true positives, false positives, true negatives, and false negatives was extractable, and methods of detection of viral DNA clearly defined. The meta-analysis was conducted following the Meta-analysis Of Observational Studies in Epidemiology (MOOSE) reporting guidelines. The aim of this meta-analysis was to evaluate the sensitivity, specificity, and accuracy of ctHPVDNA and TTMV by ddPCR to define its efficacy in clinical setting for the follow up of HPV-OPSCC.

Conclusion: The 12 studies included in the meta-analysis provided a total of 1311 patients for the analysis (398 valuated with ctHPVDNA and 913 with TTMV-HPVDNA). Pooled sensitivity and specificity were 86% (95% CI: 78%-91%) and 96% (95% CI: 91%-99%), respectively; negative and positive likelihood ratios were 0.072 (95% CI: 0.057-0.093) and 24.7 (95% CI: 6.5-93.2), respectively; pooled DOR was 371.66 (95% CI: 179.1-918). The area under the curve (AUC) was 0.81 (95% CI, 0.67-0.91). Liquid biopsy for the identification of cell free DNA might identify earlier recurrence in HPV + OPSCC patients. At the present time, liquid biopsy protocol needs to be standardized and liquid biopsy cannot yet be used in clinical setting. In the future, a multidimensional integrated approach which links multiple clinical, radiological, and laboratory data will contribute to obtain the best follow-up strategies for the follow-up of HPV-OPSCC.

背景:与人乳头状瘤病毒(HPV)相关的口咽癌发病率迅速上升,可能很快就会占到所有头颈部癌症的大多数。因此,公共卫生迫切需要改进监测和监控方法:目的是通过对ctHPVDNA和TTMV-HPVDNA的元分析研究,强调液体活检目前的潜力和局限性。该研究利用三个不同的数据库对2023年12月之前发表的文章进行了文献检索:MEDLINE、Embase 和 Cochrane Library。这些研究评估了HPV + OPSCC患者治疗后的ctHPVDNA和TTMV-HPVDNA,报告了复发诊断准确性的完整数据,或可提取真阳性、假阳性、真阴性和假阴性的数量,并明确定义了病毒DNA的检测方法。荟萃分析是根据流行病学观察性研究荟萃分析(MOOSE)报告指南进行的。该荟萃分析旨在评估 ddPCR 检测ctHPVDNA 和 TTMV 的灵敏度、特异性和准确性,以确定其在临床环境中随访 HPV-OPSCC 的有效性:纳入荟萃分析的 12 项研究共对 1311 名患者进行了分析(ctHPVDNA 评估了 398 名患者,TTMV-HPVDNA 评估了 913 名患者)。汇总灵敏度和特异度分别为86%(95% CI:78%-91%)和96%(95% CI:91%-99%);阴性和阳性似然比分别为0.072(95% CI:0.057-0.093)和24.7(95% CI:6.5-93.2);汇总DOR为371.66(95% CI:179.1-918)。曲线下面积(AUC)为 0.81(95% CI,0.67-0.91)。通过液体活检鉴定细胞游离DNA可能会提早发现HPV+OPSCC患者的复发。目前,液体活检方案尚需标准化,液体活检还不能用于临床。未来,将多种临床、放射学和实验室数据联系起来的多维综合方法将有助于为HPV-OPSCC的随访制定最佳的随访策略。
{"title":"The landscape of circulating tumor HPV DNA and TTMV-HPVDNA for surveillance of HPV-oropharyngeal carcinoma: systematic review and meta-analysis.","authors":"Flaminia Campo, Oreste Iocca, Francesca Paolini, Valentina Manciocco, Silvia Moretto, Armando De Virgilio, Claudio Moretti, Antonello Vidiri, Aldo Venuti, Paolo Bossi, Giovanni Blandino, Raul Pellini","doi":"10.1186/s13046-024-03137-1","DOIUrl":"10.1186/s13046-024-03137-1","url":null,"abstract":"<p><strong>Background: </strong>Human papilloma virus (HPV) related cancers of the oropharynx are rapidly increasing in incidence and may soon represent the majority of all head and neck cancers. Improved monitoring and surveillance methods are thus an urgent need in public health.</p><p><strong>Main text: </strong>The goal is to highlight the current potential and limitations of liquid biopsy through a meta analytic study on ctHPVDNA and TTMV-HPVDNA. It was performed a Literature search on articles published until December 2023 using three different databases: MEDLINE, Embase, and Cochrane Library. Studies that evaluated post-treatment ctHPVDNA and TTMV-HPVDNA in patients with HPV + OPSCC, studies reporting complete data on the diagnostic accuracy in recurrence, or in which the number of true positives, false positives, true negatives, and false negatives was extractable, and methods of detection of viral DNA clearly defined. The meta-analysis was conducted following the Meta-analysis Of Observational Studies in Epidemiology (MOOSE) reporting guidelines. The aim of this meta-analysis was to evaluate the sensitivity, specificity, and accuracy of ctHPVDNA and TTMV by ddPCR to define its efficacy in clinical setting for the follow up of HPV-OPSCC.</p><p><strong>Conclusion: </strong>The 12 studies included in the meta-analysis provided a total of 1311 patients for the analysis (398 valuated with ctHPVDNA and 913 with TTMV-HPVDNA). Pooled sensitivity and specificity were 86% (95% CI: 78%-91%) and 96% (95% CI: 91%-99%), respectively; negative and positive likelihood ratios were 0.072 (95% CI: 0.057-0.093) and 24.7 (95% CI: 6.5-93.2), respectively; pooled DOR was 371.66 (95% CI: 179.1-918). The area under the curve (AUC) was 0.81 (95% CI, 0.67-0.91). Liquid biopsy for the identification of cell free DNA might identify earlier recurrence in HPV + OPSCC patients. At the present time, liquid biopsy protocol needs to be standardized and liquid biopsy cannot yet be used in clinical setting. In the future, a multidimensional integrated approach which links multiple clinical, radiological, and laboratory data will contribute to obtain the best follow-up strategies for the follow-up of HPV-OPSCC.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"215"},"PeriodicalIF":11.4,"publicationDate":"2024-08-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11297591/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141879678","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
Tumor-derived GLI1 promotes remodeling of the immune tumor microenvironment in melanoma. 肿瘤衍生的 GLI1 促进了黑色素瘤免疫肿瘤微环境的重塑。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-02 DOI: 10.1186/s13046-024-03138-0
Alessandro Giammona, Chiara De Vellis, Enrica Crivaro, Luisa Maresca, Roberta Amoriello, Federica Ricci, Giulia Anichini, Silvia Pietrobono, David R Pease, Martin E Fernandez-Zapico, Clara Ballerini, Barbara Stecca

Background: Melanoma progression is based on a close interaction between cancer cells and immune cells in the tumor microenvironment (TME). Thus, a better understanding of the mechanisms controlling TME dynamics and composition will help improve the management of this dismal disease. Work from our and other groups has reported the requirement of an active Hedgehog-GLI (HH-GLI) signaling for melanoma growth and stemness. However, the role of the downstream GLI1 transcription factor in melanoma TME remains largely unexplored.

Methods: The immune-modulatory activity of GLI1 was evaluated in a syngeneic B16F10 melanoma mouse model assessing immune populations by flow cytometry. Murine polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) were differentiated from bone marrow cells and their immunosuppressive ability was assessed by inhibition of T cells. Conditioned media (CM) from GLI1-overexpressing mouse melanoma cells was used to culture PMN-MDSCs, and the effects of CM were evaluated by Transwell invasion assay and T cell inhibition. Cytokine array analysis, qPCR and chromatin immunoprecipitation were performed to explore the regulation of CX3CL1 expression by GLI1. Human monocyte-derived dendritic cells (moDCs) were cultured in CM from GLI1-silenced patient-derived melanoma cells to assess their activation and recruitment. Blocking antibodies anti-CX3CL1, anti-CCL7 and anti-CXCL8 were used for in vitro functional assays.

Results: Melanoma cell-intrinsic activation of GLI1 promotes changes in the infiltration of immune cells, leading to accumulation of immunosuppressive PMN-MDSCs and regulatory T cells, and to decreased infiltration of dendric cells (DCs), CD8 + and CD4 + T cells in the TME. In addition, we show that ectopic expression of GLI1 in melanoma cells enables PMN-MDSC expansion and recruitment, and increases their ability to inhibit T cells. The chemokine CX3CL1, a direct transcriptional target of GLI1, contributes to PMN-MDSC expansion and recruitment. Finally, silencing of GLI1 in patient-derived melanoma cells promotes the activation of human monocyte-derived dendritic cells (moDCs), increasing cytoskeleton remodeling and invasion ability. This phenotype is partially prevented by blocking the chemokine CCL7, but not CXCL8.

Conclusion: Our findings highlight the relevance of tumor-derived GLI1 in promoting an immune-suppressive TME, which allows melanoma cells to evade the immune system, and pave the way for the design of new combination treatments targeting GLI1.

背景:黑色素瘤的发展基于肿瘤微环境(TME)中癌细胞与免疫细胞之间的密切相互作用。因此,更好地了解控制肿瘤微环境动态和组成的机制将有助于改善这种令人沮丧的疾病的治疗。我们和其他研究小组的研究报告显示,黑色素瘤的生长和干性需要活跃的刺猬-GLI(HH-GLI)信号传导。然而,下游的 GLI1 转录因子在黑色素瘤 TME 中的作用在很大程度上仍未得到探索:方法:通过流式细胞术评估免疫群体,在合成B16F10黑色素瘤小鼠模型中评估了GLI1的免疫调节活性。从骨髓细胞中分化出小鼠多形核骨髓源性抑制细胞(PMN-MDSCs),并通过抑制T细胞评估其免疫抑制能力。用GLI1表达的小鼠黑色素瘤细胞的条件培养基(CM)培养PMN-MDSCs,并通过Transwell侵袭实验和T细胞抑制实验评估CM的效果。通过细胞因子阵列分析、qPCR 和染色质免疫沉淀来探讨 GLI1 对 CX3CL1 表达的调控。用GLI1沉默的患者衍生黑色素瘤细胞的CM培养人单核细胞衍生树突状细胞(moDCs),以评估它们的活化和募集。体外功能测试使用了阻断抗体抗-CX3CL1、抗-CCL7和抗-CXCL8:结果:黑色素瘤细胞内在激活GLI1会促进免疫细胞浸润的变化,导致免疫抑制性PMN-MDSCs和调节性T细胞的聚集,并减少树突细胞(DCs)、CD8 +和CD4 + T细胞在TME中的浸润。此外,我们还发现,GLI1 在黑色素瘤细胞中的异位表达能使 PMN-MDSC 扩增和募集,并增强其抑制 T 细胞的能力。趋化因子 CX3CL1 是 GLI1 的直接转录靶标,它有助于 PMN-MDSC 的扩增和募集。最后,在患者衍生的黑色素瘤细胞中沉默 GLI1 会促进人类单核细胞衍生树突状细胞(moDCs)的活化,增加细胞骨架重塑和侵袭能力。通过阻断趋化因子 CCL7(而非 CXCL8)可部分防止这种表型:我们的研究结果凸显了肿瘤衍生的 GLI1 在促进免疫抑制性 TME 方面的相关性,这种 TME 使黑色素瘤细胞得以逃避免疫系统,并为设计针对 GLI1 的新型联合疗法铺平了道路。
{"title":"Tumor-derived GLI1 promotes remodeling of the immune tumor microenvironment in melanoma.","authors":"Alessandro Giammona, Chiara De Vellis, Enrica Crivaro, Luisa Maresca, Roberta Amoriello, Federica Ricci, Giulia Anichini, Silvia Pietrobono, David R Pease, Martin E Fernandez-Zapico, Clara Ballerini, Barbara Stecca","doi":"10.1186/s13046-024-03138-0","DOIUrl":"10.1186/s13046-024-03138-0","url":null,"abstract":"<p><strong>Background: </strong>Melanoma progression is based on a close interaction between cancer cells and immune cells in the tumor microenvironment (TME). Thus, a better understanding of the mechanisms controlling TME dynamics and composition will help improve the management of this dismal disease. Work from our and other groups has reported the requirement of an active Hedgehog-GLI (HH-GLI) signaling for melanoma growth and stemness. However, the role of the downstream GLI1 transcription factor in melanoma TME remains largely unexplored.</p><p><strong>Methods: </strong>The immune-modulatory activity of GLI1 was evaluated in a syngeneic B16F10 melanoma mouse model assessing immune populations by flow cytometry. Murine polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) were differentiated from bone marrow cells and their immunosuppressive ability was assessed by inhibition of T cells. Conditioned media (CM) from GLI1-overexpressing mouse melanoma cells was used to culture PMN-MDSCs, and the effects of CM were evaluated by Transwell invasion assay and T cell inhibition. Cytokine array analysis, qPCR and chromatin immunoprecipitation were performed to explore the regulation of CX3CL1 expression by GLI1. Human monocyte-derived dendritic cells (moDCs) were cultured in CM from GLI1-silenced patient-derived melanoma cells to assess their activation and recruitment. Blocking antibodies anti-CX3CL1, anti-CCL7 and anti-CXCL8 were used for in vitro functional assays.</p><p><strong>Results: </strong>Melanoma cell-intrinsic activation of GLI1 promotes changes in the infiltration of immune cells, leading to accumulation of immunosuppressive PMN-MDSCs and regulatory T cells, and to decreased infiltration of dendric cells (DCs), CD8 + and CD4 + T cells in the TME. In addition, we show that ectopic expression of GLI1 in melanoma cells enables PMN-MDSC expansion and recruitment, and increases their ability to inhibit T cells. The chemokine CX3CL1, a direct transcriptional target of GLI1, contributes to PMN-MDSC expansion and recruitment. Finally, silencing of GLI1 in patient-derived melanoma cells promotes the activation of human monocyte-derived dendritic cells (moDCs), increasing cytoskeleton remodeling and invasion ability. This phenotype is partially prevented by blocking the chemokine CCL7, but not CXCL8.</p><p><strong>Conclusion: </strong>Our findings highlight the relevance of tumor-derived GLI1 in promoting an immune-suppressive TME, which allows melanoma cells to evade the immune system, and pave the way for the design of new combination treatments targeting GLI1.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"214"},"PeriodicalIF":11.4,"publicationDate":"2024-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11295348/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141876557","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
FGL1: a novel biomarker and target for non-small cell lung cancer, promoting tumor progression and metastasis through KDM4A/STAT3 transcription mechanism. FGL1:非小细胞肺癌的新型生物标记物和靶点,通过 KDM4A/STAT3 转录机制促进肿瘤进展和转移。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-08-01 DOI: 10.1186/s13046-024-03140-6
Tian Yao Liu, Jin Shan Yan, Xin Li, Lu Xu, Jun Li Hao, Su Ya Zhao, Qi Lin Hu, Fang Jian Na, He Ming Li, Yue Zhao, Ming Fang Zhao

Non-small cell lung cancer (NSCLC) is characterized by a high incidence rate and poor prognosis worldwide. A deeper insight into the pathogenesis of NSCLC and identification of novel therapeutic targets are essential to improve the prognosis of NSCLC. In this study, we revealed that fibrinogen-like protein 1 (FGL1) promotes proliferation, migration, and invasion of NSCLC cells. Mechanistically, we found that Stat3 acts as a transcription factor and can be recruited to the FGL1 promoter, enhancing FGL1 promoter activity. Lysine-specific demethylase 4A (KDM4A) interacts with Stat3 and facilitates the removal of methyl groups from H3K9me3, thereby enhancing Stat3-mediated transcription of FGL1. Furthermore, we observed that Stat3 and KDM4A promote NSCLC cell proliferation, migration, and invasion partly by upregulating FGL1 expression. Additionally, the expression of FGL1 was significantly higher in cancer tissues (n = 90) than in adjacent non-cancerous tissues (n = 90). Furthermore, patients with high FGL1 expression had a shorter overall survival (OS) compared to those with low FGL1 expression. We measured the expression levels of FGL1 on circulating tumor cells (CTCs) in 65 patients and found that patients with a dynamic decrease in FGL1 expression on CTCs exhibited a better therapeutic response. These findings suggest that the dynamic changes in FGL1 expression can serve as a potential biomarker for predicting treatment efficacy in NSCLC. Overall, this study revealed the significant role and regulatory mechanisms of FGL1 in the development of NSCLC, suggesting its potential as a therapeutic target for patients with NSCLC. Future studies should provide more personalized and effective treatment options for patients with NSCLC to improve clinical outcomes.

非小细胞肺癌(NSCLC)在全球具有发病率高、预后差的特点。要改善非小细胞肺癌的预后,深入了解非小细胞肺癌的发病机制并确定新的治疗靶点至关重要。在这项研究中,我们发现纤维蛋白原样蛋白1(FGL1)能促进NSCLC细胞的增殖、迁移和侵袭。从机理上讲,我们发现Stat3作为一种转录因子,可以被招募到FGL1启动子上,从而增强FGL1启动子的活性。赖氨酸特异性去甲基化酶 4A(KDM4A)与 Stat3 相互作用,促进 H3K9me3 中甲基的去除,从而增强 Stat3 介导的 FGL1 转录。此外,我们观察到 Stat3 和 KDM4A 部分通过上调 FGL1 的表达促进 NSCLC 细胞的增殖、迁移和侵袭。此外,癌症组织(n = 90)中 FGL1 的表达明显高于邻近的非癌症组织(n = 90)。此外,与 FGL1 低表达的患者相比,FGL1 高表达的患者总生存期(OS)更短。我们测量了65名患者循环肿瘤细胞(CTCs)上的FGL1表达水平,发现CTCs上FGL1表达动态下降的患者表现出更好的治疗反应。这些发现表明,FGL1表达的动态变化可作为预测NSCLC疗效的潜在生物标记物。总之,这项研究揭示了FGL1在NSCLC发病过程中的重要作用和调控机制,表明它有可能成为NSCLC患者的治疗靶点。未来的研究应为NSCLC患者提供更多个性化的有效治疗方案,以改善临床疗效。
{"title":"FGL1: a novel biomarker and target for non-small cell lung cancer, promoting tumor progression and metastasis through KDM4A/STAT3 transcription mechanism.","authors":"Tian Yao Liu, Jin Shan Yan, Xin Li, Lu Xu, Jun Li Hao, Su Ya Zhao, Qi Lin Hu, Fang Jian Na, He Ming Li, Yue Zhao, Ming Fang Zhao","doi":"10.1186/s13046-024-03140-6","DOIUrl":"10.1186/s13046-024-03140-6","url":null,"abstract":"<p><p>Non-small cell lung cancer (NSCLC) is characterized by a high incidence rate and poor prognosis worldwide. A deeper insight into the pathogenesis of NSCLC and identification of novel therapeutic targets are essential to improve the prognosis of NSCLC. In this study, we revealed that fibrinogen-like protein 1 (FGL1) promotes proliferation, migration, and invasion of NSCLC cells. Mechanistically, we found that Stat3 acts as a transcription factor and can be recruited to the FGL1 promoter, enhancing FGL1 promoter activity. Lysine-specific demethylase 4A (KDM4A) interacts with Stat3 and facilitates the removal of methyl groups from H3K9me3, thereby enhancing Stat3-mediated transcription of FGL1. Furthermore, we observed that Stat3 and KDM4A promote NSCLC cell proliferation, migration, and invasion partly by upregulating FGL1 expression. Additionally, the expression of FGL1 was significantly higher in cancer tissues (n = 90) than in adjacent non-cancerous tissues (n = 90). Furthermore, patients with high FGL1 expression had a shorter overall survival (OS) compared to those with low FGL1 expression. We measured the expression levels of FGL1 on circulating tumor cells (CTCs) in 65 patients and found that patients with a dynamic decrease in FGL1 expression on CTCs exhibited a better therapeutic response. These findings suggest that the dynamic changes in FGL1 expression can serve as a potential biomarker for predicting treatment efficacy in NSCLC. Overall, this study revealed the significant role and regulatory mechanisms of FGL1 in the development of NSCLC, suggesting its potential as a therapeutic target for patients with NSCLC. Future studies should provide more personalized and effective treatment options for patients with NSCLC to improve clinical outcomes.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"213"},"PeriodicalIF":11.4,"publicationDate":"2024-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11293164/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141861507","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
Inhibiting interferon-γ induced cancer intrinsic TNFRSF14 elevation restrains the malignant progression of glioblastoma. 抑制干扰素-γ诱导的癌症内在TNFRSF14的升高可抑制胶质母细胞瘤的恶性发展。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-07-31 DOI: 10.1186/s13046-024-03131-7
Yunhe Han, Cunyi Zou, Tianqi Liu, Wen Cheng, Peng Cheng, Anhua Wu

Background: Prolonged interferon-γ signaling activation induces cancer resistance to therapeutics, especially immunotherapy. However, the detailed mechanisms are not well characterized. In present study, we explored cancer intrinsic resistant mechanisms employing for evading immune checkpoint blockade (ICB) and searched for key immune checkpoints contributing to the constitution of suppressive immune microenvironment of glioblastoma (GBM).

Methods: We screened key immune checkpoint (IC) associated with IFN signaling activation in GBM according to integrated transcriptomic profiling on the ICs. Expression analysis and functional assays revealed that malignant cells elevated the key IC, TNFRSF14 expression under IFN-γ stimulation, which enhanced their proliferation and in vivo tumorigenicity. Therapeutic efficiency of TNFRSF14 disruption in GBM was evaluated with in vitro and in vivo functional assays, including immunofluorescence, transwell, RT-qPCR, flow cytometry, mass cytometry, and mice preclinical GBM models. Moreover, the improvement of TNFRSF14 blockade on the efficacy of PD-L1 treatment was examined in mice intracranial xenograft bearing models.

Results: TNFRSF14, a previously poorly characterized IC, was disclosed as a checkpoint with malignant intrinsic elevation closely associated with type II not type I IFN signaling activation in GBM. Anti-PD-L1 treatment induces compensatory TNFRSF14 elevation, while enhancing IFN-γ production. TNFRSF14 phosphorylates FAK at Y397 and consequently activates NF-κB, which not only strengthens the tumorigenicity of GBM cells, but also enhances TAMs recruitment through elevating CXCL1/CXCL5 secretion from GBM cells. TNFRSF14 ablation reduces the tumorigenicity of GBM cells, reshapes the immunosuppressive microenvironment, and enhances therapeutic efficacy of anti-PD-L1 in mouse orthotopic GBM model.

Conclusion: Our findings highlight a malignant TNFRSF14/FAK axis as a potential target to blunt cancer-intrinsic resistance to ICB treatment, which may help improve the therapeutic efficiency of immunotherapy in malignancies.

背景:长时间的干扰素-γ 信号激活会诱导癌症对治疗药物,尤其是免疫疗法产生抗药性。然而,其详细机制还不十分明确。在本研究中,我们探索了癌症内在的抗药性机制,这些机制用于逃避免疫检查点阻断(ICB),并寻找导致胶质母细胞瘤(GBM)抑制性免疫微环境形成的关键免疫检查点:方法:我们根据ICs的综合转录组图谱筛选了与IFN信号激活相关的GBM关键免疫检查点(IC)。表达分析和功能测试显示,恶性细胞在IFN-γ刺激下升高了关键免疫检查点TNFRSF14的表达,这增强了它们的增殖和体内致瘤性。通过免疫荧光、transwell、RT-qPCR、流式细胞仪、质谱仪和小鼠临床前GBM模型等体外和体内功能测试,评估了破坏TNFRSF14对GBM的治疗效果。此外,还在小鼠颅内异种移植模型中检测了TNFRSF14阻断对PD-L1治疗效果的改善作用:结果:TNFRSF14是一种以前特征不明显的IC,它是一种检查点,其恶性内在升高与GBM中II型而非IFN信号激活密切相关。抗 PD-L1 治疗可诱导 TNFRSF14 的代偿性升高,同时增强 IFN-γ 的产生。TNFRSF14 可使 Y397 处的 FAK 磷酸化,从而激活 NF-κB,这不仅会增强 GBM 细胞的致瘤性,还会通过提高 GBM 细胞的 CXCL1/CXCL5 分泌来增强 TAMs 募集。TNFRSF14的消融降低了GBM细胞的致瘤性,重塑了免疫抑制微环境,提高了抗PD-L1在小鼠正位GBM模型中的疗效:我们的研究结果凸显了恶性肿瘤 TNFRSF14/FAK 轴是削弱癌症对 ICB 治疗内在耐药性的潜在靶点,这可能有助于提高免疫疗法在恶性肿瘤中的治疗效率。
{"title":"Inhibiting interferon-γ induced cancer intrinsic TNFRSF14 elevation restrains the malignant progression of glioblastoma.","authors":"Yunhe Han, Cunyi Zou, Tianqi Liu, Wen Cheng, Peng Cheng, Anhua Wu","doi":"10.1186/s13046-024-03131-7","DOIUrl":"10.1186/s13046-024-03131-7","url":null,"abstract":"<p><strong>Background: </strong>Prolonged interferon-γ signaling activation induces cancer resistance to therapeutics, especially immunotherapy. However, the detailed mechanisms are not well characterized. In present study, we explored cancer intrinsic resistant mechanisms employing for evading immune checkpoint blockade (ICB) and searched for key immune checkpoints contributing to the constitution of suppressive immune microenvironment of glioblastoma (GBM).</p><p><strong>Methods: </strong>We screened key immune checkpoint (IC) associated with IFN signaling activation in GBM according to integrated transcriptomic profiling on the ICs. Expression analysis and functional assays revealed that malignant cells elevated the key IC, TNFRSF14 expression under IFN-γ stimulation, which enhanced their proliferation and in vivo tumorigenicity. Therapeutic efficiency of TNFRSF14 disruption in GBM was evaluated with in vitro and in vivo functional assays, including immunofluorescence, transwell, RT-qPCR, flow cytometry, mass cytometry, and mice preclinical GBM models. Moreover, the improvement of TNFRSF14 blockade on the efficacy of PD-L1 treatment was examined in mice intracranial xenograft bearing models.</p><p><strong>Results: </strong>TNFRSF14, a previously poorly characterized IC, was disclosed as a checkpoint with malignant intrinsic elevation closely associated with type II not type I IFN signaling activation in GBM. Anti-PD-L1 treatment induces compensatory TNFRSF14 elevation, while enhancing IFN-γ production. TNFRSF14 phosphorylates FAK at Y397 and consequently activates NF-κB, which not only strengthens the tumorigenicity of GBM cells, but also enhances TAMs recruitment through elevating CXCL1/CXCL5 secretion from GBM cells. TNFRSF14 ablation reduces the tumorigenicity of GBM cells, reshapes the immunosuppressive microenvironment, and enhances therapeutic efficacy of anti-PD-L1 in mouse orthotopic GBM model.</p><p><strong>Conclusion: </strong>Our findings highlight a malignant TNFRSF14/FAK axis as a potential target to blunt cancer-intrinsic resistance to ICB treatment, which may help improve the therapeutic efficiency of immunotherapy in malignancies.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"212"},"PeriodicalIF":11.4,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11289992/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141861508","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
IGF1R signaling induces epithelial-mesenchymal plasticity via ITGAV in cutaneous carcinoma. IGF1R 信号通过 ITGAV 在皮肤癌中诱导上皮-间质可塑性。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-07-29 DOI: 10.1186/s13046-024-03119-3
Marta Lopez-Cerda, Laura Lorenzo-Sanz, Victoria da Silva-Diz, Sandra Llop, Rosa M Penin, Josep Oriol Bermejo, Richard de Goeij-de Haas, Sander R Piersma, Thang V Pham, Connie R Jimenez, Juan Martin-Liberal, Purificación Muñoz

Background: Early cutaneous squamous cell carcinomas (cSCCs) generally show epithelial differentiation features and good prognosis, whereas advanced cSCCs present mesenchymal traits associated with tumor relapse, metastasis, and poor survival. Currently, the mechanisms involved in cSCC progression are unclear, and the established markers are suboptimal for accurately predicting the clinical course of the disease.

Methods: Using a mouse model of cSCC progression, expression microarray analysis, immunofluorescence and flow cytometry assays, we have identified a prognostic biomarker of tumor relapse, which has been evaluated in a cohort of cSCC patient samples. Phosphoproteomic analysis have revealed signaling pathways induced in epithelial plastic cancer cells that promote epithelial-mesenchymal plasticity (EMP) and tumor progression. These pathways have been validated by genetic and pharmacological inhibition assays.

Results: We show that the emergence of epithelial cancer cells expressing integrin αV (ITGAV) promotes cSCC progression to a mesenchymal state. Consistently, ITGAV expression allows the identification of patients at risk of cSCC relapse above the currently employed clinical histopathological parameters. We also demonstrate that activation of insulin-like growth factor-1 receptor (IGF1R) pathway in epithelial cancer cells is necessary to induce EMP and mesenchymal state acquisition in response to tumor microenvironment-derived factors, while promoting ITGAV expression. Likewise, ITGAV knockdown in epithelial plastic cancer cells also blocks EMP acquisition, generating epithelial tumors.

Conclusions: Our results demonstrate that ITGAV is a prognostic biomarker of relapse in cSCCs that would allow improved patient stratification. ITGAV also collaborates with IGF1R to induce EMP in epithelial cancer cells and promotes cSCC progression, revealing a potential therapeutic strategy to block the generation of advanced mesenchymal cSCCs.

背景:早期皮肤鳞状细胞癌(cSCC)一般表现为上皮分化特征,预后良好,而晚期cSCC则表现为间质特征,与肿瘤复发、转移和生存率低有关。目前,参与 cSCC 进展的机制尚不清楚,已有的标记物也无法准确预测疾病的临床过程:方法:我们利用小鼠 cSCC 进展模型、表达微阵列分析、免疫荧光和流式细胞术检测,确定了肿瘤复发的预后生物标志物,并在一组 cSCC 患者样本中进行了评估。磷蛋白组学分析揭示了上皮可塑性癌细胞中诱导的信号通路,这些通路促进了上皮-间质可塑性(EMP)和肿瘤的进展。这些通路已通过基因和药理抑制实验得到验证:结果:我们发现,表达整合素αV(ITGAV)的上皮癌细胞的出现促进了cSCC向间充质状态的进展。同样,ITGAV 的表达可以识别有复发风险的 cSCC 患者,其风险高于目前使用的临床组织病理学参数。我们还证明,上皮癌细胞中胰岛素样生长因子-1 受体(IGF1R)通路的激活是诱导 EMP 和间质状态获得的必要条件,以应对肿瘤微环境衍生因子,同时促进 ITGAV 的表达。同样,ITGAV在上皮整形癌细胞中的敲除也会阻止EMP的获得,从而产生上皮肿瘤:我们的研究结果表明,ITGAV是cSCC复发的预后生物标志物,可改善患者分层。ITGAV还与IGF1R合作诱导上皮癌细胞中的EMP,并促进cSCC的进展,从而揭示了阻止晚期间质型cSCC生成的潜在治疗策略。
{"title":"IGF1R signaling induces epithelial-mesenchymal plasticity via ITGAV in cutaneous carcinoma.","authors":"Marta Lopez-Cerda, Laura Lorenzo-Sanz, Victoria da Silva-Diz, Sandra Llop, Rosa M Penin, Josep Oriol Bermejo, Richard de Goeij-de Haas, Sander R Piersma, Thang V Pham, Connie R Jimenez, Juan Martin-Liberal, Purificación Muñoz","doi":"10.1186/s13046-024-03119-3","DOIUrl":"10.1186/s13046-024-03119-3","url":null,"abstract":"<p><strong>Background: </strong>Early cutaneous squamous cell carcinomas (cSCCs) generally show epithelial differentiation features and good prognosis, whereas advanced cSCCs present mesenchymal traits associated with tumor relapse, metastasis, and poor survival. Currently, the mechanisms involved in cSCC progression are unclear, and the established markers are suboptimal for accurately predicting the clinical course of the disease.</p><p><strong>Methods: </strong>Using a mouse model of cSCC progression, expression microarray analysis, immunofluorescence and flow cytometry assays, we have identified a prognostic biomarker of tumor relapse, which has been evaluated in a cohort of cSCC patient samples. Phosphoproteomic analysis have revealed signaling pathways induced in epithelial plastic cancer cells that promote epithelial-mesenchymal plasticity (EMP) and tumor progression. These pathways have been validated by genetic and pharmacological inhibition assays.</p><p><strong>Results: </strong>We show that the emergence of epithelial cancer cells expressing integrin αV (ITGAV) promotes cSCC progression to a mesenchymal state. Consistently, ITGAV expression allows the identification of patients at risk of cSCC relapse above the currently employed clinical histopathological parameters. We also demonstrate that activation of insulin-like growth factor-1 receptor (IGF1R) pathway in epithelial cancer cells is necessary to induce EMP and mesenchymal state acquisition in response to tumor microenvironment-derived factors, while promoting ITGAV expression. Likewise, ITGAV knockdown in epithelial plastic cancer cells also blocks EMP acquisition, generating epithelial tumors.</p><p><strong>Conclusions: </strong>Our results demonstrate that ITGAV is a prognostic biomarker of relapse in cSCCs that would allow improved patient stratification. ITGAV also collaborates with IGF1R to induce EMP in epithelial cancer cells and promotes cSCC progression, revealing a potential therapeutic strategy to block the generation of advanced mesenchymal cSCCs.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"211"},"PeriodicalIF":11.4,"publicationDate":"2024-07-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11285232/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141793952","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
Pericytes recruited by CCL28 promote vascular normalization after anti-angiogenesis therapy through RA/RXRA/ANGPT1 pathway in lung adenocarcinoma. CCL28招募的周细胞通过RA/RXRA/AGPT1途径促进肺腺癌抗血管生成治疗后的血管正常化。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-07-29 DOI: 10.1186/s13046-024-03135-3
Ying Chen, Zhiyong Zhang, Fan Pan, Pengfei Li, Weiping Yao, Yuxi Chen, Lei Xiong, Tingting Wang, Yan Li, Guichun Huang

Background: It has been proposed that anti-angiogenesis therapy could induce tumor "vascular normalization" and further enhance the efficacy of chemotherapy, radiotherapy, target therapy, and immunotherapy for nearly twenty years. However, the detailed molecular mechanism of this phenomenon is still obscure.

Method: Overexpression and knockout of CCL28 in human lung adenocarcinoma cell line A549 and murine lung adenocarcinoma cell line LLC, respectively, were utilized to establish mouse models. Single-cell sequencing was performed to analyze the proportion of different cell clusters and metabolic changes in the tumor microenvironment (TME). Immunofluorescence and multiplex immunohistochemistry were conducted in murine tumor tissues and clinical biopsy samples to assess the percentage of pericytes coverage. Primary pericytes were isolated from lung adenocarcinoma tumor tissues using magnetic-activated cell sorting (MACS). These pericytes were then treated with recombinant human CCL28 protein, followed by transwell migration assays and RNA sequencing analysis. Changes in the secretome and metabolome were examined, and verification of retinoic acid metabolism alterations in pericytes was conducted using quantitative real-time PCR, western blotting, and LC-MS technology. Chromatin immunoprecipitation followed by quantitative PCR (ChIP-qPCR) was employed to validate the transcriptional regulatory ability and affinity of RXRα to specific sites at the ANGPT1 promoter.

Results: Our study showed that after undergoing anti-angiogenesis treatment, the tumor exhibited a state of ischemia and hypoxia, leading to an upregulation in the expression of CCL28 in hypoxic lung adenocarcinoma cells by the hypoxia-sensitive transcription factor CEBPB. Increased CCL28 could promote tumor vascular normalization through recruiting and metabolic reprogramming pericytes in the tumor microenvironment. Mechanistically, CCL28 modified the retinoic acid (RA) metabolism and increased ANGPT1 expression via RXRα in pericytes, thereby enhancing the stability of endothelial cells.

Conclusion: We reported the details of the molecular mechanisms of "vascular normalization" after anti-angiogenesis therapy for the first time. Our work might provide a prospective molecular marker for guiding the clinical arrangement of combination therapy between anti-angiogenesis treatment and other therapies.

背景:近二十年来,抗血管生成治疗可诱导肿瘤 "血管正常化",进一步提高化疗、放疗、靶向治疗和免疫治疗的疗效。然而,这一现象的详细分子机制仍然模糊不清:方法:分别在人肺腺癌细胞株 A549 和鼠肺腺癌细胞株 LLC 中过表达和敲除 CCL28,建立小鼠模型。通过单细胞测序分析不同细胞群的比例和肿瘤微环境(TME)的代谢变化。对小鼠肿瘤组织和临床活检样本进行免疫荧光和多重免疫组化,以评估周细胞的覆盖率。利用磁激活细胞分选技术(MACS)从肺腺癌肿瘤组织中分离出原发性周细胞。然后用重组人 CCL28 蛋白处理这些周细胞,接着进行跨孔迁移试验和 RNA 测序分析。研究人员检测了分泌组和代谢组的变化,并利用定量实时 PCR、Western 印迹和 LC-MS 技术验证了周细胞中维甲酸代谢的改变。采用染色质免疫共沉淀后定量 PCR(ChIP-qPCR)技术验证了 RXRα 对 ANGPT1 启动子特定位点的转录调控能力和亲和力:我们的研究表明,在接受抗血管生成治疗后,肿瘤呈现缺血缺氧状态,缺氧敏感转录因子CEBPB导致CCL28在缺氧肺腺癌细胞中表达上调。CCL28的增加可通过招募和代谢重编程肿瘤微环境中的周细胞,促进肿瘤血管正常化。从机制上讲,CCL28改变了视黄酸(RA)的代谢,并通过RXRα增加了周细胞中ANGPT1的表达,从而增强了内皮细胞的稳定性:我们首次报道了抗血管生成治疗后 "血管正常化 "的分子机制细节。结论:我们首次报道了抗血管生成治疗后 "血管正常化 "的分子机制细节,为指导临床安排抗血管生成治疗与其他疗法的联合治疗提供了前瞻性分子标记。
{"title":"Pericytes recruited by CCL28 promote vascular normalization after anti-angiogenesis therapy through RA/RXRA/ANGPT1 pathway in lung adenocarcinoma.","authors":"Ying Chen, Zhiyong Zhang, Fan Pan, Pengfei Li, Weiping Yao, Yuxi Chen, Lei Xiong, Tingting Wang, Yan Li, Guichun Huang","doi":"10.1186/s13046-024-03135-3","DOIUrl":"10.1186/s13046-024-03135-3","url":null,"abstract":"<p><strong>Background: </strong>It has been proposed that anti-angiogenesis therapy could induce tumor \"vascular normalization\" and further enhance the efficacy of chemotherapy, radiotherapy, target therapy, and immunotherapy for nearly twenty years. However, the detailed molecular mechanism of this phenomenon is still obscure.</p><p><strong>Method: </strong>Overexpression and knockout of CCL28 in human lung adenocarcinoma cell line A549 and murine lung adenocarcinoma cell line LLC, respectively, were utilized to establish mouse models. Single-cell sequencing was performed to analyze the proportion of different cell clusters and metabolic changes in the tumor microenvironment (TME). Immunofluorescence and multiplex immunohistochemistry were conducted in murine tumor tissues and clinical biopsy samples to assess the percentage of pericytes coverage. Primary pericytes were isolated from lung adenocarcinoma tumor tissues using magnetic-activated cell sorting (MACS). These pericytes were then treated with recombinant human CCL28 protein, followed by transwell migration assays and RNA sequencing analysis. Changes in the secretome and metabolome were examined, and verification of retinoic acid metabolism alterations in pericytes was conducted using quantitative real-time PCR, western blotting, and LC-MS technology. Chromatin immunoprecipitation followed by quantitative PCR (ChIP-qPCR) was employed to validate the transcriptional regulatory ability and affinity of RXRα to specific sites at the ANGPT1 promoter.</p><p><strong>Results: </strong>Our study showed that after undergoing anti-angiogenesis treatment, the tumor exhibited a state of ischemia and hypoxia, leading to an upregulation in the expression of CCL28 in hypoxic lung adenocarcinoma cells by the hypoxia-sensitive transcription factor CEBPB. Increased CCL28 could promote tumor vascular normalization through recruiting and metabolic reprogramming pericytes in the tumor microenvironment. Mechanistically, CCL28 modified the retinoic acid (RA) metabolism and increased ANGPT1 expression via RXRα in pericytes, thereby enhancing the stability of endothelial cells.</p><p><strong>Conclusion: </strong>We reported the details of the molecular mechanisms of \"vascular normalization\" after anti-angiogenesis therapy for the first time. Our work might provide a prospective molecular marker for guiding the clinical arrangement of combination therapy between anti-angiogenesis treatment and other therapies.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"210"},"PeriodicalIF":11.4,"publicationDate":"2024-07-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11285179/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141793953","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
IL-33 stimulates the anticancer activities of eosinophils through extracellular vesicle-driven reprogramming of tumor cells. IL-33 通过细胞外囊泡驱动的肿瘤细胞重编程,刺激嗜酸性粒细胞的抗癌活性。
IF 11.4 1区 医学 Q1 ONCOLOGY Pub Date : 2024-07-27 DOI: 10.1186/s13046-024-03129-1
Adriana Rosa Gambardella, Caterina Antonucci, Cristiana Zanetti, Francesco Noto, Sara Andreone, Davide Vacca, Valentina Pellerito, Chiara Sicignano, Giuseppe Parrottino, Valentina Tirelli, Antonella Tinari, Mario Falchi, Adele De Ninno, Luca Businaro, Stefania Loffredo, Gilda Varricchi, Claudio Tripodo, Claudia Afferni, Isabella Parolini, Fabrizio Mattei, Giovanna Schiavoni

Immune cell-derived extracellular vesicles (EV) affect tumor progression and hold promise for therapeutic applications. Eosinophils are major effectors in Th2-related pathologies recently implied in cancer. Here, we evaluated the anti-tumor activities of eosinophil-derived EV following activation with the alarmin IL-33. We demonstrate that IL-33-activated mouse and human eosinophils produce higher quantities of EV with respect to eosinophils stimulated with IL-5. Following incorporation of EV from IL-33-activated eosinophils (Eo33-EV), but not EV from IL-5-treated eosinophils (Eo5-EV), mouse and human tumor cells increased the expression of cyclin-dependent kinase inhibitor (CDKI)-related genes resulting in cell cycle arrest in G0/G1, reduced proliferation and inhibited tumor spheroid formation. Moreover, tumor cells incorporating Eo33-EV acquired an epithelial-like phenotype characterized by E-Cadherin up-regulation, N-Cadherin downregulation, reduced cell elongation and migratory extent in vitro, and impaired capacity to metastasize to lungs when injected in syngeneic mice. RNA sequencing revealed distinct mRNA signatures in Eo33-EV and Eo5-EV with increased presence of tumor suppressor genes and enrichment in pathways related to epithelial phenotypes and negative regulation of cellular processes in Eo33-EV compared to Eo5-EV. Our studies underscore novel IL-33-stimulated anticancer activities of eosinophils through EV-mediated reprogramming of tumor cells opening perspectives on the use of eosinophil-derived EV in cancer therapy.

免疫细胞衍生的细胞外囊泡 (EV) 会影响肿瘤的发展,并有望用于治疗。嗜酸性粒细胞是 Th2 相关病症的主要效应因子,最近在癌症中也有所体现。在这里,我们评估了嗜酸性粒细胞衍生的EV在被警戒素IL-33激活后的抗肿瘤活性。我们证明,IL-33 激活的小鼠和人类嗜酸性粒细胞与 IL-5 刺激的嗜酸性粒细胞相比,能产生更多的 EV。小鼠和人类肿瘤细胞吸收了 IL-33 激活的嗜酸性粒细胞产生的 EV(Eo33-EV)而非 IL-5 处理的嗜酸性粒细胞产生的 EV(Eo5-EV)后,细胞周期依赖性激酶抑制剂(CDKI)相关基因的表达增加,导致细胞周期停滞在 G0/G1,增殖减少并抑制了肿瘤球体的形成。此外,含有Eo33-EV的肿瘤细胞获得了上皮样表型,其特征是E-Cadherin上调、N-Cadherin下调、体外细胞伸长和迁移程度降低,以及在注射给合成小鼠时向肺部转移的能力减弱。RNA测序显示,Eo33-EV和Eo5-EV中的mRNA特征不同,与Eo5-EV相比,Eo33-EV中肿瘤抑制基因增多,与上皮表型和细胞过程负调控相关的通路丰富。我们的研究强调了嗜酸性粒细胞通过EV介导的肿瘤细胞重编程而产生的新型IL-33刺激抗癌活性,为在癌症治疗中使用嗜酸性粒细胞衍生的EV开辟了前景。
{"title":"IL-33 stimulates the anticancer activities of eosinophils through extracellular vesicle-driven reprogramming of tumor cells.","authors":"Adriana Rosa Gambardella, Caterina Antonucci, Cristiana Zanetti, Francesco Noto, Sara Andreone, Davide Vacca, Valentina Pellerito, Chiara Sicignano, Giuseppe Parrottino, Valentina Tirelli, Antonella Tinari, Mario Falchi, Adele De Ninno, Luca Businaro, Stefania Loffredo, Gilda Varricchi, Claudio Tripodo, Claudia Afferni, Isabella Parolini, Fabrizio Mattei, Giovanna Schiavoni","doi":"10.1186/s13046-024-03129-1","DOIUrl":"10.1186/s13046-024-03129-1","url":null,"abstract":"<p><p>Immune cell-derived extracellular vesicles (EV) affect tumor progression and hold promise for therapeutic applications. Eosinophils are major effectors in Th2-related pathologies recently implied in cancer. Here, we evaluated the anti-tumor activities of eosinophil-derived EV following activation with the alarmin IL-33. We demonstrate that IL-33-activated mouse and human eosinophils produce higher quantities of EV with respect to eosinophils stimulated with IL-5. Following incorporation of EV from IL-33-activated eosinophils (Eo33-EV), but not EV from IL-5-treated eosinophils (Eo5-EV), mouse and human tumor cells increased the expression of cyclin-dependent kinase inhibitor (CDKI)-related genes resulting in cell cycle arrest in G0/G1, reduced proliferation and inhibited tumor spheroid formation. Moreover, tumor cells incorporating Eo33-EV acquired an epithelial-like phenotype characterized by E-Cadherin up-regulation, N-Cadherin downregulation, reduced cell elongation and migratory extent in vitro, and impaired capacity to metastasize to lungs when injected in syngeneic mice. RNA sequencing revealed distinct mRNA signatures in Eo33-EV and Eo5-EV with increased presence of tumor suppressor genes and enrichment in pathways related to epithelial phenotypes and negative regulation of cellular processes in Eo33-EV compared to Eo5-EV. Our studies underscore novel IL-33-stimulated anticancer activities of eosinophils through EV-mediated reprogramming of tumor cells opening perspectives on the use of eosinophil-derived EV in cancer therapy.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":"43 1","pages":"209"},"PeriodicalIF":11.4,"publicationDate":"2024-07-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11282757/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141767961","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