Journal of Experimental & Clinical Cancer Research最新文献

筛选
英文 中文
Molecular understanding and clinical aspects of tumor-associated macrophages in the immunotherapy of renal cell carcinoma. 肿瘤相关巨噬细胞在肾细胞癌免疫疗法中的分子认识和临床应用。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2024-08-22 DOI: 10.1186/s13046-024-03164-y
Han Liu, Zongwei Lv, Gong Zhang, Zhenhong Yan, Song Bai, Dan Dong, Kefeng Wang
{"title":"Molecular understanding and clinical aspects of tumor-associated macrophages in the immunotherapy of renal cell carcinoma.","authors":"Han Liu, Zongwei Lv, Gong Zhang, Zhenhong Yan, Song Bai, Dan Dong, Kefeng Wang","doi":"10.1186/s13046-024-03164-y","DOIUrl":"10.1186/s13046-024-03164-y","url":null,"abstract":"<p><p>Renal cell carcinoma (RCC) is one of the most common tumors that afflicts the urinary system, accounting for 90-95% of kidney cancer cases. Although its incidence has increased over the past decades, its pathogenesis is still unclear. Tumor-associated macrophages (TAMs) are the most prominent immune cells in the tumor microenvironment (TME), comprising more than 50% of the tumor volume. By interacting with cancer cells, TAMs can be polarized into two distinct phenotypes, M1-type and M2-type TAMs. In the TME, M2-type TAMs, which are known to promote tumorigenesis, are more abundant than M1-type TAMs, which are known to suppress tumor growth. This ratio of M1 to M2 TAMs can create an immunosuppressive environment that contributes to tumor cell progression and survival. This review focused on the role of TAMs in RCC, including their polarization, impacts on tumor proliferation, angiogenesis, invasion, migration, drug resistance, and immunosuppression. In addition, we discussed the potential of targeting TAMs for clinical therapy in RCC. A deeper understanding of the molecular biology of TAMs is essential for exploring innovative therapeutic strategies for the treatment of RCC.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":null,"pages":null},"PeriodicalIF":11.4,"publicationDate":"2024-08-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11340075/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142019404","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
Connecting the dots: investigating the link between environmental, genetic, and epigenetic influences in metabolomic alterations in oral squamous cell carcinoma. 连接点:研究口腔鳞状细胞癌代谢组学改变中环境、遗传和表观遗传影响之间的联系。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2024-08-21 DOI: 10.1186/s13046-024-03141-5
Ishita Gupta, Fariba Badrzadeh, Yuri Tsentalovich, Daria A Gaykalova
{"title":"Connecting the dots: investigating the link between environmental, genetic, and epigenetic influences in metabolomic alterations in oral squamous cell carcinoma.","authors":"Ishita Gupta, Fariba Badrzadeh, Yuri Tsentalovich, Daria A Gaykalova","doi":"10.1186/s13046-024-03141-5","DOIUrl":"10.1186/s13046-024-03141-5","url":null,"abstract":"<p><p>Oral squamous cell carcinoma (OSCC) accounts for around 90% of all oral cancers and is the eighth most common cancer worldwide. Despite progress in managing OSCC, the overall prognosis remains poor, with a survival rate of around 50-60%, largely due to tumor size and recurrence. The challenges of late-stage diagnosis and limitations in current methods emphasize the urgent need for less invasive techniques to enable early detection and treatment, crucial for improving outcomes in this aggressive form of oral cancer. Research is currently aimed at unraveling tumor-specific metabolite profiles to identify candidate biomarkers as well as discover underlying pathways involved in the onset and progression of cancer that could be used as new targets for diagnostic and therapeutic purposes. Metabolomics is an advanced technological approach to identify metabolites in different sample types (biological fluids and tissues). Since OSCC promotes metabolic reprogramming influenced by a combination of genetic predisposition and environmental factors, including tobacco and alcohol consumption, and viral infections, the identification of distinct metabolites through screening may aid in the diagnosis of this condition. Moreover, studies have shown the use of metabolites during the catalysis of epigenetic modification, indicating a link between epigenetics and metabolism. In this review, we will focus on the link between environmental, genetic, and epigenetic influences in metabolomic alterations in OSCC. In addition, we will discuss therapeutic targets of tumor metabolism, which may prevent oral tumor growth, metastasis, and drug resistance.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":null,"pages":null},"PeriodicalIF":11.4,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11337877/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142019444","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 DNA repair pathway as a therapeutic target to synergize with trastuzumab deruxtecan in HER2-targeted antibody-drug conjugate-resistant HER2-overexpressing breast cancer. 将DNA修复途径作为治疗靶点,与曲妥珠单抗德鲁司坦协同治疗HER2靶向抗体-药物共轭物耐药的HER2表达缺失乳腺癌。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2024-08-21 DOI: 10.1186/s13046-024-03143-3
Jangsoon Lee, Kumiko Kida, Jiwon Koh, Huey Liu, Ganiraju C Manyam, Young Jin Gi, Dileep R Rampa, Asha S Multani, Jing Wang, Gitanjali Jayachandran, Dae-Won Lee, James M Reuben, Aysegul Sahin, Lei Huo, Debu Tripathy, Seock-Ah Im, Naoto T Ueno
{"title":"The DNA repair pathway as a therapeutic target to synergize with trastuzumab deruxtecan in HER2-targeted antibody-drug conjugate-resistant HER2-overexpressing breast cancer.","authors":"Jangsoon Lee, Kumiko Kida, Jiwon Koh, Huey Liu, Ganiraju C Manyam, Young Jin Gi, Dileep R Rampa, Asha S Multani, Jing Wang, Gitanjali Jayachandran, Dae-Won Lee, James M Reuben, Aysegul Sahin, Lei Huo, Debu Tripathy, Seock-Ah Im, Naoto T Ueno","doi":"10.1186/s13046-024-03143-3","DOIUrl":"10.1186/s13046-024-03143-3","url":null,"abstract":"<p><strong>Background: </strong>Anti-HER2 therapies, including the HER2 antibody-drug conjugates (ADCs) trastuzumab emtansine (T-DM1) and trastuzumab deruxtecan (T-DXd), have led to improved survival outcomes in patients with HER2-overexpressing (HER2+) metastatic breast cancer. However, intrinsic or acquired resistance to anti-HER2-based therapies remains a clinical challenge in these patients, as there is no standard of care following disease progression. The purpose of this study was to elucidate the mechanisms of resistance to T-DM1 and T-DXd in HER2+ BC patients and preclinical models and identify targets whose inhibition enhances the antitumor activity of T-DXd in HER2-directed ADC-resistant HER2+ breast cancer in vitro and in vivo.</p><p><strong>Methods: </strong>Targeted DNA and whole transcriptome sequencing were performed in breast cancer patient tissue samples to investigate genetic aberrations that arose after anti-HER2 therapy. We generated T-DM1 and T-DXd-resistant HER2+ breast cancer cell lines. To elucidate their resistance mechanisms and to identify potential synergistic kinase targets for enhancing the efficacy of T-DXd, we used fluorescence in situ hybridization, droplet digital PCR, Western blotting, whole-genome sequencing, cDNA microarray, and synthetic lethal kinome RNA interference screening. In addition, cell viability, colony formation, and xenograft assays were used to determine the synergistic antitumor effect of T-DXd combinations.</p><p><strong>Results: </strong>We found reduced HER2 expression in patients and amplified DNA repair-related genes in patients after anti-HER2 therapy. Reduced ERBB2 gene amplification in HER2-directed ADC-resistant HER2+ breast cancer cell lines was through DNA damage and epigenetic mechanisms. In HER2-directed ADC-resistant HER2+ breast cancer cell lines, our non-biased RNA interference screening identified the DNA repair pathway as a potential target within the canonical pathways to enhance the efficacy of T-DXd. We validated that the combination of T-DXd with ataxia telangiectasia and Rad3-related inhibitor, elimusertib, led to significant breast cancer cell death in vitro (P < 0.01) and in vivo (P < 0.01) compared to single agents.</p><p><strong>Conclusions: </strong>The DNA repair pathways contribute to HER2-directed ADC resistance. Our data justify exploring the combination treatment of T-DXd with DNA repair-targeting drugs to treat HER2-directed ADC-resistant HER2+ breast cancer in clinical trials.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":null,"pages":null},"PeriodicalIF":11.4,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11337831/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142009858","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
Circulating tumour DNA dynamics predict recurrence in stage III melanoma patients receiving neoadjuvant immunotherapy. 循环肿瘤 DNA 动态预测接受新辅助免疫疗法的 III 期黑色素瘤患者的复发。
IF 11.4 1区 医学
Journal of Experimental & Clinical Cancer Research Pub Date : 2024-08-21 DOI: 10.1186/s13046-024-03153-1
Wei Yen Chan, Jenny H Lee, Ashleigh Stewart, Russell J Diefenbach, Maria Gonzalez, Alexander M Menzies, Christian Blank, Richard A Scolyer, Georgina V Long, Helen Rizos
{"title":"Circulating tumour DNA dynamics predict recurrence in stage III melanoma patients receiving neoadjuvant immunotherapy.","authors":"Wei Yen Chan, Jenny H Lee, Ashleigh Stewart, Russell J Diefenbach, Maria Gonzalez, Alexander M Menzies, Christian Blank, Richard A Scolyer, Georgina V Long, Helen Rizos","doi":"10.1186/s13046-024-03153-1","DOIUrl":"10.1186/s13046-024-03153-1","url":null,"abstract":"<p><strong>Background: </strong>Neoadjuvant therapy improves recurrence-free survival (RFS) in resectable stage III cutaneous melanoma. However, accurately predicting individual recurrence risk remains a significant challenge. We investigated circulating tumour DNA (ctDNA) as a biomarker for recurrence in measurable stage IIIB/C melanoma patients undergoing neoadjuvant immunotherapy.</p><p><strong>Methods: </strong>Plasma samples were collected pre-neoadjuvant treatment, pre-surgery and/or six weeks post-surgery from 40 patients enrolled in the OpACIN-neo and PRADO clinical trials. Patients received two cycles of ipilimumab (anti-CTLA-4) and nivolumab (anti-PD-1) before surgery. Cell free DNA (cfDNA) underwent unbiased pre-amplification followed by tumour-informed mutation detection using droplet digital polymerase chain reaction (ddPCR) with the Bio-Rad QX600 PCR system.</p><p><strong>Results: </strong>Pre-treatment ctDNA was detectable in 19/40 (48%) patients. Among these, 17/19 (89%) zero-converted within six weeks of surgery and none recurred. Positive ctDNA post-surgery (N = 4), irrespective of pre-treatment ctDNA status, was 100% predictive of recurrence (sensitivity 44%, specificity 100%). Furthermore, ctDNA cleared prior to surgery in 7/9 (78%) patients who did not recur, warranting further investigation into ctDNA-guided surgical management.</p><p><strong>Conclusion: </strong>Post-surgery ctDNA positivity and zero-conversion are highly predictive of recurrence, offering a window for personalised modification of adjuvant therapy.</p>","PeriodicalId":50199,"journal":{"name":"Journal of Experimental & Clinical Cancer Research","volume":null,"pages":null},"PeriodicalIF":11.4,"publicationDate":"2024-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11337884/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142019443","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
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
Journal of Experimental & Clinical Cancer Research 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
{"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":null,"pages":null},"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
Journal of Experimental & Clinical Cancer Research 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
{"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":null,"pages":null},"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
Journal of Experimental & Clinical Cancer Research 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
{"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":null,"pages":null},"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
Journal of Experimental & Clinical Cancer Research 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
{"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":null,"pages":null},"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
Journal of Experimental & Clinical Cancer Research 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
{"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":null,"pages":null},"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
Journal of Experimental & Clinical Cancer Research 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
{"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":null,"pages":null},"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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信