首页 > 最新文献

Cellular Oncology最新文献

英文 中文
Prognostic significance of collagen signatures at breast tumor boundary obtained by combining multiphoton imaging and imaging analysis. 结合多光子成像和成像分析获得的乳腺肿瘤边界胶原特征的预后意义。
IF 6.6 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-02-01 Epub Date: 2023-08-22 DOI: 10.1007/s13402-023-00851-4
Xingxin Huang, Fangmeng Fu, Wenhui Guo, Deyong Kang, Xiahui Han, Liqin Zheng, Zhenlin Zhan, Chuan Wang, Qingyuan Zhang, Shu Wang, Shunwu Xu, Jianli Ma, Lida Qiu, Jianxin Chen, Lianhuang Li

Purpose: Collagen features in breast tumor microenvironment is closely associated with the prognosis of patients. We aim to explore the prognostic significance of collagen features at breast tumor border by combining multiphoton imaging and imaging analysis.

Methods: We used multiphoton microscopy (MPM) to label-freely image human breast tumor samples and then constructed an automatic classification model based on deep learning to identify collagen signatures from multiphoton images. We recognized three kinds of collagen signatures at tumor boundary (CSTB I-III) in a small-scale, and furthermore obtained a CSTB score for each patient based on the combined CSTB I-III by using the ridge regression analysis. The prognostic performance of CSTB score is assessed by the area under the receiver operating characteristic curve (AUC), Cox proportional hazard regression analysis, as well as Kaplan-Meier survival analysis.

Results: As an independent prognostic factor, statistical results reveal that the prognostic performance of CSTB score is better than that of the clinical model combining three independent prognostic indicators, molecular subtype, tumor size, and lymph nodal metastasis (AUC, Training dataset: 0.773 vs. 0.749; External validation: 0.753 vs. 0.724; HR, Training dataset: 4.18 vs. 3.92; External validation: 4.98 vs. 4.16), and as an auxiliary indicator, it can greatly improve the accuracy of prognostic prediction. And furthermore, a nomogram combining the CSTB score with the clinical model is established for prognosis prediction and clinical decision making.

Conclusion: This standardized and automated imaging prognosticator may convince pathologists to adopt it as a prognostic factor, thereby customizing more effective treatment plans for patients.

目的:乳腺肿瘤微环境中的胶原蛋白特征与患者的预后密切相关。我们旨在结合多光子成像和成像分析,探讨乳腺肿瘤边界胶原蛋白特征的预后意义:方法:我们使用多光子显微镜(MPM)对人类乳腺肿瘤样本进行无标记成像,然后构建了一个基于深度学习的自动分类模型,从多光子图像中识别胶原蛋白特征。我们在小范围内识别了肿瘤边界的三种胶原蛋白特征(CSTB I-III),并根据CSTB I-III的组合,通过脊回归分析得出了每位患者的CSTB评分。通过接收者操作特征曲线下面积(AUC)、Cox比例危险回归分析以及Kaplan-Meier生存分析评估CSTB评分的预后效果:统计结果显示,作为一个独立的预后因素,CSTB评分的预后效果优于结合分子亚型、肿瘤大小和淋巴结转移三个独立预后指标的临床模型(AUC,训练数据集:0.773 vs. 0.749; External validation:外部验证:0.753 vs. 0.724;HR,训练数据集:4.18 vs. 3.92):4.18 vs. 3.92; External validation:4.98 vs. 4.16),作为辅助指标,可以大大提高预后预测的准确性。此外,还建立了一个将CSTB评分与临床模型相结合的提名图,用于预后预测和临床决策:结论:这一标准化和自动化的影像预后指标可能会说服病理学家将其作为预后因素,从而为患者定制更有效的治疗方案。
{"title":"Prognostic significance of collagen signatures at breast tumor boundary obtained by combining multiphoton imaging and imaging analysis.","authors":"Xingxin Huang, Fangmeng Fu, Wenhui Guo, Deyong Kang, Xiahui Han, Liqin Zheng, Zhenlin Zhan, Chuan Wang, Qingyuan Zhang, Shu Wang, Shunwu Xu, Jianli Ma, Lida Qiu, Jianxin Chen, Lianhuang Li","doi":"10.1007/s13402-023-00851-4","DOIUrl":"10.1007/s13402-023-00851-4","url":null,"abstract":"<p><strong>Purpose: </strong>Collagen features in breast tumor microenvironment is closely associated with the prognosis of patients. We aim to explore the prognostic significance of collagen features at breast tumor border by combining multiphoton imaging and imaging analysis.</p><p><strong>Methods: </strong>We used multiphoton microscopy (MPM) to label-freely image human breast tumor samples and then constructed an automatic classification model based on deep learning to identify collagen signatures from multiphoton images. We recognized three kinds of collagen signatures at tumor boundary (CSTB I-III) in a small-scale, and furthermore obtained a CSTB score for each patient based on the combined CSTB I-III by using the ridge regression analysis. The prognostic performance of CSTB score is assessed by the area under the receiver operating characteristic curve (AUC), Cox proportional hazard regression analysis, as well as Kaplan-Meier survival analysis.</p><p><strong>Results: </strong>As an independent prognostic factor, statistical results reveal that the prognostic performance of CSTB score is better than that of the clinical model combining three independent prognostic indicators, molecular subtype, tumor size, and lymph nodal metastasis (AUC, Training dataset: 0.773 vs. 0.749; External validation: 0.753 vs. 0.724; HR, Training dataset: 4.18 vs. 3.92; External validation: 4.98 vs. 4.16), and as an auxiliary indicator, it can greatly improve the accuracy of prognostic prediction. And furthermore, a nomogram combining the CSTB score with the clinical model is established for prognosis prediction and clinical decision making.</p><p><strong>Conclusion: </strong>This standardized and automated imaging prognosticator may convince pathologists to adopt it as a prognostic factor, thereby customizing more effective treatment plans for patients.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"69-80"},"PeriodicalIF":6.6,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10039753","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Osteopontin secreted from obese adipocytes enhances angiogenesis and promotes progression of pancreatic ductal adenocarcinoma in obesity. 肥胖脂肪细胞分泌的Osteopontin能增强血管生成并促进肥胖症胰腺导管腺癌的发展。
IF 6.6 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-02-01 Epub Date: 2023-08-29 DOI: 10.1007/s13402-023-00865-y
Shigeki Fukusada, Takaya Shimura, Makoto Natsume, Ruriko Nishigaki, Yusuke Okuda, Hiroyasu Iwasaki, Naomi Sugimura, Mika Kitagawa, Takahito Katano, Mamoru Tanaka, Keiji Ozeki, Eiji Kubota, Kazuki Hayashi, Hiromi Kataoka

Purpose: Obesity is a risk factor and poor prognostic factor for pancreatic ductal adenocarcinoma (PDAC), but the underlying mechanisms remain unclear.

Methods: PDAC cells and obese visceral adipocytes (O-Ad) derived from mice and humans were used to analyze interactions between the two cell types, and human microvascular endothelial cells were used for angiogenesis assay. A xenograft mouse model with subcutaneously injected PDAC cells was used for animal studies. The relationship between visceral fat and prognosis was analyzed using resected tissues from PDAC patients with and without obesity.

Results: Conditioned media (CM) from O-Ad significantly increased PDAC cell growth and migration and angiogenic capacity in both human and mice cells, and blocking osteopontin (OPN) in O-Ad canceled O-Ad-induced effects in both mouse and human cells. In addition, O-Ad directly increased the migratory and tube-forming capacities of endothelial cells, while blocking OPN canceled these effects. O-Ad increased AKT phosphorylation and VEGFA expression in both PDAC and endothelial cells, and OPN inhibition in O-Ad canceled those O-Ad-induced effects. In the xenograft model, PDAC tumor volume was significantly increased in obese mice compared with lean mice, whereas blocking OPN significantly inhibited obesity-accelerated tumor growth. OPN expression in adipose tissues adjacent to human PDAC tumor was significantly higher in obese patients than in non-obese patients. In PDAC patients with obesity, high OPN expression in adipose tissues was significantly associated with poor prognosis.

Conclusion: Obese adipocytes trigger aggressive transformation in PDAC cells to induce PDAC progression and accelerate angiogenesis via OPN secretion.

目的:肥胖是胰腺导管腺癌(PDAC)的风险因素和不良预后因素,但其潜在机制仍不清楚:方法:使用小鼠和人类的 PDAC 细胞和肥胖内脏脂肪细胞(O-Ad)分析两种细胞类型之间的相互作用,并使用人类微血管内皮细胞进行血管生成检测。动物实验采用皮下注射 PDAC 细胞的异种移植小鼠模型。利用肥胖和非肥胖 PDAC 患者的切除组织分析了内脏脂肪与预后之间的关系:结果:O-Ad的条件培养基(CM)明显增加了PDAC细胞在人和小鼠细胞中的生长、迁移和血管生成能力。此外,O-Ad 还直接提高了内皮细胞的迁移能力和管形成能力,而阻断 OPN 则消除了这些影响。O-Ad 增加了 PDAC 和内皮细胞中的 AKT 磷酸化和 VEGFA 表达,而抑制 O-Ad 中的 OPN 可消除这些 O-Ad 诱导的效应。在异种移植模型中,与瘦小鼠相比,肥胖小鼠的PDAC肿瘤体积明显增大,而阻断OPN可明显抑制肥胖加速的肿瘤生长。肥胖患者邻近人类 PDAC 肿瘤的脂肪组织中 OPN 的表达明显高于非肥胖患者。在患有肥胖症的PDAC患者中,脂肪组织中OPN的高表达与预后不良明显相关:结论:肥胖脂肪细胞通过分泌 OPN 触发 PDAC 细胞的侵袭性转化,诱导 PDAC 进展并加速血管生成。
{"title":"Osteopontin secreted from obese adipocytes enhances angiogenesis and promotes progression of pancreatic ductal adenocarcinoma in obesity.","authors":"Shigeki Fukusada, Takaya Shimura, Makoto Natsume, Ruriko Nishigaki, Yusuke Okuda, Hiroyasu Iwasaki, Naomi Sugimura, Mika Kitagawa, Takahito Katano, Mamoru Tanaka, Keiji Ozeki, Eiji Kubota, Kazuki Hayashi, Hiromi Kataoka","doi":"10.1007/s13402-023-00865-y","DOIUrl":"10.1007/s13402-023-00865-y","url":null,"abstract":"<p><strong>Purpose: </strong>Obesity is a risk factor and poor prognostic factor for pancreatic ductal adenocarcinoma (PDAC), but the underlying mechanisms remain unclear.</p><p><strong>Methods: </strong>PDAC cells and obese visceral adipocytes (O-Ad) derived from mice and humans were used to analyze interactions between the two cell types, and human microvascular endothelial cells were used for angiogenesis assay. A xenograft mouse model with subcutaneously injected PDAC cells was used for animal studies. The relationship between visceral fat and prognosis was analyzed using resected tissues from PDAC patients with and without obesity.</p><p><strong>Results: </strong>Conditioned media (CM) from O-Ad significantly increased PDAC cell growth and migration and angiogenic capacity in both human and mice cells, and blocking osteopontin (OPN) in O-Ad canceled O-Ad-induced effects in both mouse and human cells. In addition, O-Ad directly increased the migratory and tube-forming capacities of endothelial cells, while blocking OPN canceled these effects. O-Ad increased AKT phosphorylation and VEGFA expression in both PDAC and endothelial cells, and OPN inhibition in O-Ad canceled those O-Ad-induced effects. In the xenograft model, PDAC tumor volume was significantly increased in obese mice compared with lean mice, whereas blocking OPN significantly inhibited obesity-accelerated tumor growth. OPN expression in adipose tissues adjacent to human PDAC tumor was significantly higher in obese patients than in non-obese patients. In PDAC patients with obesity, high OPN expression in adipose tissues was significantly associated with poor prognosis.</p><p><strong>Conclusion: </strong>Obese adipocytes trigger aggressive transformation in PDAC cells to induce PDAC progression and accelerate angiogenesis via OPN secretion.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"229-244"},"PeriodicalIF":6.6,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10102291","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
FGF19 promotes nasopharyngeal carcinoma progression by inducing angiogenesis via inhibiting TRIM21-mediated ANXA2 ubiquitination. FGF19通过抑制TRIM21介导的ANXA2泛素化诱导血管生成来促进鼻咽癌进展。
IF 6.6 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-02-01 Epub Date: 2023-10-02 DOI: 10.1007/s13402-023-00868-9
Si Shi, Qicheng Zhang, Kaiwen Zhang, Wenhui Chen, Haijing Xie, Si Pan, Ziyi Xue, Bo You, Jianmei Zhao, Yiwen You

Purpose: Nasopharyngeal carcinoma (NPC) has characteristics of high invasion and early metastasis. Most NPC patients present with locoregionally advanced illness when first diagnosed. Therefore, it is urgent to discover NPC biomarkers. Fibroblast growth Factor 19 (FGF19) plays a role in various physiological or pathological processes, including cancer. In this research, we discovered the importance of FGF19 in NPC, and clarified its role in tumour angiogenesis.

Methods: Western blotting, immunohistochemistry and ELISA were used to investigate FGF19 expression in NPC. Then we took CCK8, colony formation, Transwell and wound healing assays to identify the influence of FGF19 on NPC malignant behaviours. The proliferative and metastatic capacity of FGF19 were evaluated in nude mice and zebrafish. The role of FGF19 in angiogenesis was investigated by tube formation and Matrigel plug angiogenesis assays. We then evaluated the variation in Annexin A2(ANXA2) levels with the treatment of FGF19. Lastly, co-immunoprecipitation and ubiquitination assays were performed to identify the mechanisms involved.

Results: FGF19 levels were elevated in tissues and serum of NPC patients and were associated with poor clinical stages. High expression of FGF19 promoted NPC malignant behaviours. In particular, FGF19 expression was correlated with microvessel density in tissues and NPC-derived FGF19 could accelerate angiogenesis in vitro and in vivo. Mechanistically, FGF19 influenced ANXA2 expression to promote angiogenesis. Moreover, tripartite motif-containing 21(TRIM21) interacted with ANXA2 and was responsible for ANXA2 ubiquitination.

Conclusion: FGF19 promoted NPC angiogenesis by inhibiting TRIM21-mediated ANXA2 ubiquitination. It may serve as a noninvasive biomarker for NPC and provides new insights for therapy.

目的:鼻咽癌具有高侵袭性和早期转移的特点。大多数鼻咽癌患者在首次诊断时表现为局部晚期疾病。因此,发现NPC生物标志物是当务之急。成纤维细胞生长因子19(FGF19)在包括癌症在内的各种生理或病理过程中发挥作用。在本研究中,我们发现了FGF19在NPC中的重要性,并阐明了其在肿瘤血管生成中的作用。方法:采用蛋白质印迹法、免疫组织化学法和ELISA法检测FGF19在鼻咽癌中的表达。然后我们采用CCK8、集落形成、Transwell和伤口愈合试验来鉴定FGF19对NPC恶性行为的影响。在裸鼠和斑马鱼中评估FGF19的增殖和转移能力。FGF19在血管生成中的作用通过试管形成和Matrigel栓塞血管生成测定进行了研究。然后,我们评估了膜联蛋白A2(ANXA2)水平随FGF19治疗的变化。最后,进行免疫共沉淀和泛素化测定以确定所涉及的机制。结果:鼻咽癌患者组织和血清中FGF19水平升高,与临床分期差有关。FGF19的高表达促进了NPC的恶性行为。特别是,FGF19的表达与组织中的微血管密度相关,NPC衍生的FGF19可以在体外和体内加速血管生成。从机制上讲,FGF19影响ANXA2的表达以促进血管生成。此外,含有21的三元基序(TRIM21)与ANXA2相互作用,并负责ANXA2的泛素化。结论:FGF19通过抑制TRIM21介导的ANXA2泛素化而促进NPC血管生成。它可以作为NPC的非侵入性生物标志物,并为治疗提供新的见解。
{"title":"FGF19 promotes nasopharyngeal carcinoma progression by inducing angiogenesis via inhibiting TRIM21-mediated ANXA2 ubiquitination.","authors":"Si Shi, Qicheng Zhang, Kaiwen Zhang, Wenhui Chen, Haijing Xie, Si Pan, Ziyi Xue, Bo You, Jianmei Zhao, Yiwen You","doi":"10.1007/s13402-023-00868-9","DOIUrl":"10.1007/s13402-023-00868-9","url":null,"abstract":"<p><strong>Purpose: </strong>Nasopharyngeal carcinoma (NPC) has characteristics of high invasion and early metastasis. Most NPC patients present with locoregionally advanced illness when first diagnosed. Therefore, it is urgent to discover NPC biomarkers. Fibroblast growth Factor 19 (FGF19) plays a role in various physiological or pathological processes, including cancer. In this research, we discovered the importance of FGF19 in NPC, and clarified its role in tumour angiogenesis.</p><p><strong>Methods: </strong>Western blotting, immunohistochemistry and ELISA were used to investigate FGF19 expression in NPC. Then we took CCK8, colony formation, Transwell and wound healing assays to identify the influence of FGF19 on NPC malignant behaviours. The proliferative and metastatic capacity of FGF19 were evaluated in nude mice and zebrafish. The role of FGF19 in angiogenesis was investigated by tube formation and Matrigel plug angiogenesis assays. We then evaluated the variation in Annexin A2(ANXA2) levels with the treatment of FGF19. Lastly, co-immunoprecipitation and ubiquitination assays were performed to identify the mechanisms involved.</p><p><strong>Results: </strong>FGF19 levels were elevated in tissues and serum of NPC patients and were associated with poor clinical stages. High expression of FGF19 promoted NPC malignant behaviours. In particular, FGF19 expression was correlated with microvessel density in tissues and NPC-derived FGF19 could accelerate angiogenesis in vitro and in vivo. Mechanistically, FGF19 influenced ANXA2 expression to promote angiogenesis. Moreover, tripartite motif-containing 21(TRIM21) interacted with ANXA2 and was responsible for ANXA2 ubiquitination.</p><p><strong>Conclusion: </strong>FGF19 promoted NPC angiogenesis by inhibiting TRIM21-mediated ANXA2 ubiquitination. It may serve as a noninvasive biomarker for NPC and provides new insights for therapy.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"283-301"},"PeriodicalIF":6.6,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10899426/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41150010","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Endothelial-derived small extracellular vesicles support B-cell acute lymphoblastic leukemia development. 内皮细胞衍生的细胞外小泡支持B细胞急性淋巴细胞白血病的发展。
IF 6.6 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-02-01 Epub Date: 2023-09-26 DOI: 10.1007/s13402-023-00855-0
Dan Huang, Yamin Yuan, Liyuan Cao, Difan Zhang, Yu Jiang, Yaping Zhang, Chiqi Chen, Zhuo Yu, Li Xie, Yujuan Wei, Jiangbo Wan, Junke Zheng

Purpose: The bone marrow niche plays an important role in leukemia development. However, the contributions of different niche components to leukemia development and their underlying mechanisms remain largely unclear.

Method: Cre/LoxP-based conditional knockout technology was used to delete VPS33B or ANGPTL2 gene in niche cells. Murine B-ALL model was established by overexpressing the N-Myc oncogene in hematopoietic stem progenitor cells. The frequency of leukemia cells and immunophenotypic B220+ CD43+ LICs was detected by flow cytometry. SEVs was isolated by sequential centrifugation and mass spectrometry was performed to analyze the different components of SEVs. Immunoprecipitation and western blot were used to measure the interaction of VPS33B and ANGPTL2.

Results: Here, we showed that specific knockout of vascular protein sorting 33b (Vps33b) in endothelial cells (ECs), but not megakaryocytes or mesenchymal stem cells, resulted in a significant decrease in the secretion of small extracellular vesicles (SEVs) and a delay in the development of B-cell lymphoblastic leukemia (B-ALL). Vps33b knockdown endothelial cells contained much lower levels of SEVs that contained angiopoietin-like protein 2 (ANGPTL2) than the control cells. Importantly, conditional knockout of Angptl2 in ECs significantly delayed B-ALL progression. Moreover, C-terminal region of ANGPTL2 (aa247-471) could directly interact with Sec1-like domain 1 of VPS33B (aa1-aa146). We further demonstrated that the point mutations R399H and G402S in ANGPTL2 led to a dramatic decrease in the secretion of ANGPTL2-SEVs. We also showed that wild-type ANGPTL2-containing SEVs, but not mutant ANGPTL2-containing SEVs, significantly enhanced B-ALL development.

Conclusion: In summary, our findings indicate that the secretion of ANGPTL2-containing SEVs in ECs sustains the leukemogenic activities of B-ALL cells, which is fine-tuned by the direct interaction of VPS33B and ANGPTL2. These findings reveal that niche-specific SEVs play an important role in B-ALL development.

目的:骨髓生态位在白血病的发生发展中起着重要作用。然而,不同生态位成分对白血病发展的贡献及其潜在机制在很大程度上仍不清楚。方法:采用基于Cre/LoxP的条件敲除技术,对小生境细胞中的VPS33B或ANGPTL2基因进行敲除。通过在造血干祖细胞中过表达N-Myc癌基因建立小鼠B-ALL模型。流式细胞仪检测白血病细胞和免疫表型B220+CD43+LICs的频率。通过顺序离心分离SEV,并进行质谱分析来分析SEV的不同成分。免疫沉淀和蛋白质印迹法检测了VPS33B和ANGPTL2的相互作用。结果:我们发现,血管蛋白分选33b(VPS33B)在内皮细胞(EC)中的特异性敲除,而在巨核细胞或间充质干细胞中没有,导致细胞外小泡(SEVs)的分泌显著减少,并延迟B细胞淋巴细胞白血病(B-ALL)的发展。Vps33b敲低的内皮细胞含有比对照细胞低得多的含有血管生成素样蛋白2(ANGPTL2)的SEV水平。重要的是,ECs中Angptl2的条件性敲除显著延迟了B-ALL的进展。此外,ANGPTL2的C末端区域(aa247-471)可以直接与VPS33B的Sec1样结构域1(aa1-aa146)相互作用。我们进一步证明,ANGPTL2中的点突变R399H和G402S导致ANGPTL2 SEVs的分泌显著减少。我们还发现,含有SEVs的野生型ANGPTL2,而不是含有SEVss的突变体ANGPTL2显著增强了B-ALL的发育。结论:总之,我们的研究结果表明,内皮细胞中含有ANGPTL2的SEVs的分泌维持了B-ALL细胞的致白血病活性,而VPS33B和ANGPTL2之间的直接相互作用对其进行了微调。这些发现表明,小众特异性SEV在B-ALL的发展中起着重要作用。
{"title":"Endothelial-derived small extracellular vesicles support B-cell acute lymphoblastic leukemia development.","authors":"Dan Huang, Yamin Yuan, Liyuan Cao, Difan Zhang, Yu Jiang, Yaping Zhang, Chiqi Chen, Zhuo Yu, Li Xie, Yujuan Wei, Jiangbo Wan, Junke Zheng","doi":"10.1007/s13402-023-00855-0","DOIUrl":"10.1007/s13402-023-00855-0","url":null,"abstract":"<p><strong>Purpose: </strong>The bone marrow niche plays an important role in leukemia development. However, the contributions of different niche components to leukemia development and their underlying mechanisms remain largely unclear.</p><p><strong>Method: </strong>Cre/LoxP-based conditional knockout technology was used to delete VPS33B or ANGPTL2 gene in niche cells. Murine B-ALL model was established by overexpressing the N-Myc oncogene in hematopoietic stem progenitor cells. The frequency of leukemia cells and immunophenotypic B220<sup>+</sup> CD43<sup>+</sup> LICs was detected by flow cytometry. SEVs was isolated by sequential centrifugation and mass spectrometry was performed to analyze the different components of SEVs. Immunoprecipitation and western blot were used to measure the interaction of VPS33B and ANGPTL2.</p><p><strong>Results: </strong>Here, we showed that specific knockout of vascular protein sorting 33b (Vps33b) in endothelial cells (ECs), but not megakaryocytes or mesenchymal stem cells, resulted in a significant decrease in the secretion of small extracellular vesicles (SEVs) and a delay in the development of B-cell lymphoblastic leukemia (B-ALL). Vps33b knockdown endothelial cells contained much lower levels of SEVs that contained angiopoietin-like protein 2 (ANGPTL2) than the control cells. Importantly, conditional knockout of Angptl2 in ECs significantly delayed B-ALL progression. Moreover, C-terminal region of ANGPTL2 (aa247-471) could directly interact with Sec1-like domain 1 of VPS33B (aa1-aa146). We further demonstrated that the point mutations R399H and G402S in ANGPTL2 led to a dramatic decrease in the secretion of ANGPTL2-SEVs. We also showed that wild-type ANGPTL2-containing SEVs, but not mutant ANGPTL2-containing SEVs, significantly enhanced B-ALL development.</p><p><strong>Conclusion: </strong>In summary, our findings indicate that the secretion of ANGPTL2-containing SEVs in ECs sustains the leukemogenic activities of B-ALL cells, which is fine-tuned by the direct interaction of VPS33B and ANGPTL2. These findings reveal that niche-specific SEVs play an important role in B-ALL development.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"129-140"},"PeriodicalIF":6.6,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10899377/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41162157","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Immunosuppressive MFAP2+ cancer associated fibroblasts conferred unfavorable prognosis and therapeutic resistance in gastric cancer. 具有免疫抑制作用的 MFAP2+ 癌症相关成纤维细胞会导致胃癌的不良预后和耐药性。
IF 6.6 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-02-01 Epub Date: 2023-08-04 DOI: 10.1007/s13402-023-00849-y
Rongyuan Wei, Junquan Song, Xuanjun Liu, Shiying Huo, Chenchen Liu, Xiaowen Liu

Purpose: To explore the predictive merit of MFAP2+ cancer associated fibroblasts (CAFs) infiltration for clinical outcomes and adjuvant chemotherapy or immunotherapy responsiveness in gastric cancer (GC).

Methods: In this study, several independent cohorts were included respectively to dissect the relationship of clinical outcomes, therapeutic responses and tumor microenvironment with different MFAP2+ CAFs infiltration. Drug sensitivity analysis was conducted to predict the relationship between MFAP2+ CAFs infiltration and targeted drug response. Kaplan-Meier curves and the log-rank test were used to compare clinical outcomes of patients with different MFAP2+ CAFs infiltration.

Results: High MFAP2+ CAFs infiltration yielded inferior prognosis in terms of overall survival, progress free survival and recurrence free survival in GC. Patients with low MFAP2+ CAFs infiltration were more likely to gain benefit from adjuvant therapy. Moreover, low MFAP2+ CAFs infiltration could predict a promising response to immunotherapy in GC patients. MFAP2+ CAFs with immunosuppressive features were highly relevant to immune evasive contexture characterized by the dysfunction of CD8+ T cells. We found that MFAP2+ CAFs communicated with T cells, B cells and Macrophages through releasing macrophage migration inhibitor factor (MIF), which further suggested that MFAP2+ CAFs might promote therapeutic resistance through regulating T cells dysfunction and M2 macrophages polarization.

Conclusion: Immunosuppressive MFAP2+ CAFs constructed an immune evasive tumor microenvironment characterized by incapacitated immune effector cells, consequently predicting inferior clinical outcomes and response on adjuvant therapy and immunotherapy in patients with GC. The potential of immunosuppressive MFAP2+ CAFs as a therapeutic target for GC deserved thoroughly exploration.

目的:探讨MFAP2+癌相关成纤维细胞(CAFs)浸润对胃癌(GC)临床预后和辅助化疗或免疫治疗反应性的预测价值:本研究分别纳入了几个独立队列,以探讨临床预后、治疗反应和肿瘤微环境与不同MFAP2+ CAFs浸润的关系。通过药物敏感性分析来预测MFAP2+ CAFs浸润与靶向药物反应之间的关系。采用Kaplan-Meier曲线和对数秩检验比较不同MFAP2+ CAFs浸润患者的临床结果:结果:MFAP2+ CAFs浸润程度越高,GC患者的总生存期、无进展生存期和无复发生存期的预后越差。低MFAP2+ CAFs浸润的患者更有可能从辅助治疗中获益。此外,低MFAP2+ CAFs浸润可预测GC患者对免疫疗法的反应。具有免疫抑制特征的 MFAP2+ CAFs 与以 CD8+ T 细胞功能失调为特征的免疫逃避背景高度相关。我们发现,MFAP2+ CAFs通过释放巨噬细胞迁移抑制因子(MIF)与T细胞、B细胞和巨噬细胞沟通,这进一步表明MFAP2+ CAFs可能通过调节T细胞功能障碍和M2巨噬细胞极化来促进治疗抵抗:结论:免疫抑制性 MFAP2+ CAFs 构建了一个以免疫效应细胞失能为特征的免疫回避性肿瘤微环境,从而预示着 GC 患者的不良临床预后以及对辅助治疗和免疫治疗的反应。免疫抑制性 MFAP2+ CAFs 作为 GC 治疗靶点的潜力值得深入探讨。
{"title":"Immunosuppressive MFAP2<sup>+</sup> cancer associated fibroblasts conferred unfavorable prognosis and therapeutic resistance in gastric cancer.","authors":"Rongyuan Wei, Junquan Song, Xuanjun Liu, Shiying Huo, Chenchen Liu, Xiaowen Liu","doi":"10.1007/s13402-023-00849-y","DOIUrl":"10.1007/s13402-023-00849-y","url":null,"abstract":"<p><strong>Purpose: </strong>To explore the predictive merit of MFAP2<sup>+</sup> cancer associated fibroblasts (CAFs) infiltration for clinical outcomes and adjuvant chemotherapy or immunotherapy responsiveness in gastric cancer (GC).</p><p><strong>Methods: </strong>In this study, several independent cohorts were included respectively to dissect the relationship of clinical outcomes, therapeutic responses and tumor microenvironment with different MFAP2<sup>+</sup> CAFs infiltration. Drug sensitivity analysis was conducted to predict the relationship between MFAP2<sup>+</sup> CAFs infiltration and targeted drug response. Kaplan-Meier curves and the log-rank test were used to compare clinical outcomes of patients with different MFAP2<sup>+</sup> CAFs infiltration.</p><p><strong>Results: </strong>High MFAP2<sup>+</sup> CAFs infiltration yielded inferior prognosis in terms of overall survival, progress free survival and recurrence free survival in GC. Patients with low MFAP2<sup>+</sup> CAFs infiltration were more likely to gain benefit from adjuvant therapy. Moreover, low MFAP2<sup>+</sup> CAFs infiltration could predict a promising response to immunotherapy in GC patients. MFAP2<sup>+</sup> CAFs with immunosuppressive features were highly relevant to immune evasive contexture characterized by the dysfunction of CD8<sup>+</sup> T cells. We found that MFAP2<sup>+</sup> CAFs communicated with T cells, B cells and Macrophages through releasing macrophage migration inhibitor factor (MIF), which further suggested that MFAP2<sup>+</sup> CAFs might promote therapeutic resistance through regulating T cells dysfunction and M2 macrophages polarization.</p><p><strong>Conclusion: </strong>Immunosuppressive MFAP2<sup>+</sup> CAFs constructed an immune evasive tumor microenvironment characterized by incapacitated immune effector cells, consequently predicting inferior clinical outcomes and response on adjuvant therapy and immunotherapy in patients with GC. The potential of immunosuppressive MFAP2<sup>+</sup> CAFs as a therapeutic target for GC deserved thoroughly exploration.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"55-68"},"PeriodicalIF":6.6,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10291578","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Synergistic antitumor efficacy of gemcitabine and cisplatin to induce ferroptosis in pancreatic ductal adenocarcinoma via Sp1-SAT1-polyamine metabolism pathway. 吉西他滨和顺铂通过Sp1-SAT1-多胺代谢途径诱导胰腺导管腺癌铁变态反应的协同抗肿瘤效应
IF 6.6 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-02-01 Epub Date: 2023-09-09 DOI: 10.1007/s13402-023-00870-1
Wanhui Wei, Yuanyuan Lu, Qian Hu, Jinwen Yin, Youwei Wang, Heng Zhang, Qiu Zhao, Lan Liu

Purpose: The combination of cisplatin and gemcitabine-based chemotherapy has been recommended as a preferred regimen for pancreatic ductal adenocarcinoma (PDAC) patients with germline-based mutations. However, the underlying mechanism remains poorly elucidated. Therefore, our study aimed to explore the mechanistic basis of the cell-killing activity of gemcitabine plus cisplatin and identify potential therapeutic targets.

Methods: First, we explored the synergistic cytotoxic effects of gemcitabine and cisplatin on PDAC through in vitro and in vivo experiments. Then, we investigated ferroptosis-related biomarkers, to assess the impact of the combination therapy on ferroptosis. Using bioinformatics methods, we identified SAT1 as a potential key mediator of ferroptosis induced by gemcitabine and cisplatin. We tested the polyamine levels in PDAC cells by LC-MS after overexpressed or knocked down SAT1, and explored the role of polyamines in ferroptosis using exogenous supplementation. Finally, we explored the regulatory effect of Sp1 on SAT1 through ChIP-qPCR and dual-luciferase reporter assay.

Results: Gemcitabine plus cisplatin enhanced cell death and induced ferroptosis in PDAC. This combination upregulated SAT1 transcription by inhibiting Sp1. SAT1 activation promoted the catabolism of spermine and spermidine, leading to iron accumulation and lipid peroxide generation, ultimately resulting in ferroptosis.

Conclusions: In summary, our findings suggested the gemcitabine and cisplatin combination therapy induced ferroptosis in a GSH-independent manner in PDAC. The combined treatment inhibited Sp1 and upregulated SAT1 transcription, leading to the breakdown of spermine and spermidine. Therefore, targeting SAT1-induced polyamine metabolism may represent a promising therapeutic strategy for PDAC.

目的:顺铂和吉西他滨联合化疗已被推荐为基于基因突变的胰腺导管腺癌(PDAC)患者的首选方案。然而,其潜在机制仍未得到充分阐明。因此,我们的研究旨在探索吉西他滨加顺铂的细胞杀伤活性的机制基础,并确定潜在的治疗靶点:首先,我们通过体外和体内实验探讨了吉西他滨和顺铂对 PDAC 的协同细胞毒性作用。然后,我们研究了铁蛋白沉积相关的生物标志物,以评估联合疗法对铁蛋白沉积的影响。通过生物信息学方法,我们发现 SAT1 是吉西他滨和顺铂诱导铁突变的潜在关键介质。我们通过 LC-MS 检测了过表达或敲除 SAT1 后 PDAC 细胞中的多胺水平,并利用外源补充多胺的方法探讨了多胺在铁嗜性中的作用。最后,我们通过 ChIP-qPCR 和双荧光素酶报告实验探讨了 Sp1 对 SAT1 的调控作用:结果:吉西他滨加顺铂可增强 PDAC 的细胞死亡并诱导铁变态反应。这一组合通过抑制 Sp1 上调了 SAT1 的转录。SAT1 的激活促进了精胺和亚精胺的分解,导致铁积累和过氧化脂质的生成,最终导致铁变态反应:综上所述,我们的研究结果表明,吉西他滨和顺铂联合疗法能以GSH依赖性方式诱导PDAC发生铁变态反应。联合治疗抑制了 Sp1 并上调了 SAT1 的转录,导致精胺和亚精胺的分解。因此,靶向 SAT1 诱导的多胺代谢可能是治疗 PDAC 的一种有前景的策略。
{"title":"Synergistic antitumor efficacy of gemcitabine and cisplatin to induce ferroptosis in pancreatic ductal adenocarcinoma via Sp1-SAT1-polyamine metabolism pathway.","authors":"Wanhui Wei, Yuanyuan Lu, Qian Hu, Jinwen Yin, Youwei Wang, Heng Zhang, Qiu Zhao, Lan Liu","doi":"10.1007/s13402-023-00870-1","DOIUrl":"10.1007/s13402-023-00870-1","url":null,"abstract":"<p><strong>Purpose: </strong>The combination of cisplatin and gemcitabine-based chemotherapy has been recommended as a preferred regimen for pancreatic ductal adenocarcinoma (PDAC) patients with germline-based mutations. However, the underlying mechanism remains poorly elucidated. Therefore, our study aimed to explore the mechanistic basis of the cell-killing activity of gemcitabine plus cisplatin and identify potential therapeutic targets.</p><p><strong>Methods: </strong>First, we explored the synergistic cytotoxic effects of gemcitabine and cisplatin on PDAC through in vitro and in vivo experiments. Then, we investigated ferroptosis-related biomarkers, to assess the impact of the combination therapy on ferroptosis. Using bioinformatics methods, we identified SAT1 as a potential key mediator of ferroptosis induced by gemcitabine and cisplatin. We tested the polyamine levels in PDAC cells by LC-MS after overexpressed or knocked down SAT1, and explored the role of polyamines in ferroptosis using exogenous supplementation. Finally, we explored the regulatory effect of Sp1 on SAT1 through ChIP-qPCR and dual-luciferase reporter assay.</p><p><strong>Results: </strong>Gemcitabine plus cisplatin enhanced cell death and induced ferroptosis in PDAC. This combination upregulated SAT1 transcription by inhibiting Sp1. SAT1 activation promoted the catabolism of spermine and spermidine, leading to iron accumulation and lipid peroxide generation, ultimately resulting in ferroptosis.</p><p><strong>Conclusions: </strong>In summary, our findings suggested the gemcitabine and cisplatin combination therapy induced ferroptosis in a GSH-independent manner in PDAC. The combined treatment inhibited Sp1 and upregulated SAT1 transcription, leading to the breakdown of spermine and spermidine. Therefore, targeting SAT1-induced polyamine metabolism may represent a promising therapeutic strategy for PDAC.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"321-341"},"PeriodicalIF":6.6,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10188886","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting PUF60 prevents tumor progression by retarding mRNA decay of oxidative phosphorylation in ovarian cancer. 靶向 PUF60 可延缓卵巢癌氧化磷酸化 mRNA 的衰变,从而防止肿瘤进展。
IF 6.6 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-02-01 Epub Date: 2023-08-26 DOI: 10.1007/s13402-023-00859-w
Cancan Zhang, Xiaoge Ni, Chunlin Tao, Ziyang Zhou, Fengmian Wang, Fei Gu, Xiaoxiao Cui, Shuheng Jiang, Qing Li, Huan Lu, Dongxue Li, Zhiyong Wu, Rong Zhang

Purpose: Ovarian cancer (OC) is the leading cause of death from gynecological malignancies, and its etiology and pathogenesis are currently unclear. Recent studies have found that PUF60 overexpressed in various cancers. However, the exact function of PUF60 in global RNA processing and its role in OC has been unclear.

Methods: The expression of PUF60 and its relationship with clinical characteristics were analyzed by multiple database analysis and immunohistochemistry. Phenotypic effects of PUF60 on ovarian cancer cell proliferation and metastasis were examined by in vitro cell proliferation assay, migration assay, and in vivo xenograft models and lung metastasis models. RNA immunoprecipitation, seahorse analyses, RNA stability assay were used to study the effect of PUF60 on the stability of oxidative phosphorylation (OXPHOS)-related genes in OC.

Results: We report PUF60 is highly expressed in OC with frequent amplification of up to 33.9% and its upregulation predicts a poor prognosis. PUF60 promotes the proliferation and migration of OC cells both in vitro and in vivo. Mechanistically, we demonstrated that silencing of PUF60 enhanced the stability of mRNA transcripts involved in OXPHOS and decreased the formation of processing bodies (P-bodies), ultimately elevating the OXPHOS level.

Conclusion: Our study unveils a novel function of PUF60 in OC energy metabolism. Thus, PUF60 may serve as a novel target for the treatment of patients with OC.

目的:卵巢癌(OC)是妇科恶性肿瘤的主要死因,其病因和发病机制目前尚不清楚。最近的研究发现,PUF60 在多种癌症中过度表达。然而,PUF60在全局RNA处理中的确切功能及其在OC中的作用尚不清楚:方法:通过多重数据库分析和免疫组化分析了PUF60的表达及其与临床特征的关系。通过体外细胞增殖试验、迁移试验以及体内异种移植模型和肺转移模型,研究了 PUF60 对卵巢癌细胞增殖和转移的表型效应。通过RNA免疫沉淀、海马分析、RNA稳定性测定等方法研究了PUF60对OC中氧化磷酸化(OXPHOS)相关基因稳定性的影响:结果:我们发现PUF60在OC中高表达,扩增率高达33.9%,其上调预示着预后不良。PUF60 可促进体外和体内 OC 细胞的增殖和迁移。从机理上讲,我们证实沉默 PUF60 可增强参与 OXPHOS 的 mRNA 转录本的稳定性,减少加工体(P-bodies)的形成,最终提高 OXPHOS 水平:我们的研究揭示了 PUF60 在 OC 能量代谢中的新功能。结论:我们的研究揭示了 PUF60 在 OC 能量代谢中的新功能,因此,PUF60 可作为治疗 OC 患者的新靶点。
{"title":"Targeting PUF60 prevents tumor progression by retarding mRNA decay of oxidative phosphorylation in ovarian cancer.","authors":"Cancan Zhang, Xiaoge Ni, Chunlin Tao, Ziyang Zhou, Fengmian Wang, Fei Gu, Xiaoxiao Cui, Shuheng Jiang, Qing Li, Huan Lu, Dongxue Li, Zhiyong Wu, Rong Zhang","doi":"10.1007/s13402-023-00859-w","DOIUrl":"10.1007/s13402-023-00859-w","url":null,"abstract":"<p><strong>Purpose: </strong>Ovarian cancer (OC) is the leading cause of death from gynecological malignancies, and its etiology and pathogenesis are currently unclear. Recent studies have found that PUF60 overexpressed in various cancers. However, the exact function of PUF60 in global RNA processing and its role in OC has been unclear.</p><p><strong>Methods: </strong>The expression of PUF60 and its relationship with clinical characteristics were analyzed by multiple database analysis and immunohistochemistry. Phenotypic effects of PUF60 on ovarian cancer cell proliferation and metastasis were examined by in vitro cell proliferation assay, migration assay, and in vivo xenograft models and lung metastasis models. RNA immunoprecipitation, seahorse analyses, RNA stability assay were used to study the effect of PUF60 on the stability of oxidative phosphorylation (OXPHOS)-related genes in OC.</p><p><strong>Results: </strong>We report PUF60 is highly expressed in OC with frequent amplification of up to 33.9% and its upregulation predicts a poor prognosis. PUF60 promotes the proliferation and migration of OC cells both in vitro and in vivo. Mechanistically, we demonstrated that silencing of PUF60 enhanced the stability of mRNA transcripts involved in OXPHOS and decreased the formation of processing bodies (P-bodies), ultimately elevating the OXPHOS level.</p><p><strong>Conclusion: </strong>Our study unveils a novel function of PUF60 in OC energy metabolism. Thus, PUF60 may serve as a novel target for the treatment of patients with OC.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"157-174"},"PeriodicalIF":6.6,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10899302/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10078596","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Overexpressed FAM111B degrades GSDMA to promote esophageal cancer tumorigenesis and cisplatin resistance. 过表达的 FAM111B 能降解 GSDMA,从而促进食管癌肿瘤发生和顺铂耐药。
IF 6.6 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-02-01 Epub Date: 2023-09-06 DOI: 10.1007/s13402-023-00871-0
Haiqin Wang, Haohui Wang, Jiajing Chen, Pian Liu, Xiaoxiong Xiao

Background: Chemotherapeutic agents such as cisplatin are commonly used in patients with clinically unresectable or recurrent esophageal cancer (ESCA). However, patients often develop resistance to cisplatin, which in turn leads to a poor prognosis. Studies have shown that FAM111B may be involved in the development of tumors as an oncogene or tumor suppressor gene. However, the pathological role and corresponding mechanism of FAM111B in ESCA are still unclear.

Methods: The GEPIA web tool, ENCORI Pan-Cancer Analysis Platform and UALCAN-TCGA database were used to study the expression of FAM111B in ESCA. CCK-8, angiogenesis, Transwell and xenograft assays were applied to explore the biological function of FAM111B in ESCA. Western blot, RT-qPCR, and RNA-seq analyses were applied to study the FAM111B/GSDMA axis in the progression of ESCA cells. CCK-8 and xenograft assays were used to study the role of the FAM111B/GSDMA axis in determining the sensitivity of ESCA to cisplatin.

Results: Our results demonstrated that FAM111B is highly expressed in ESCA tissues compared to normal tissues. We showed that FAM111B promotes the progression of ESCC cells by binding to GSDMA and that the trypsin protease domain is essential for the activity of FAM111B. Furthermore, we showed that the FAM111B/GSDMA axis regulates cisplatin sensitivity in ESCA.

Conclusions: Overall, we identified a novel FAM111B/GSDMA axis regulating ESCA tumorigenesis and chemosensitivity, at least in ESCC cells.

背景:顺铂等化疗药物常用于临床上无法切除或复发的食管癌(ESCA)患者。然而,患者往往会对顺铂产生耐药性,进而导致预后不良。研究表明,FAM111B 可能作为癌基因或抑癌基因参与肿瘤的发生发展。然而,FAM111B在ESCA中的病理作用和相应机制仍不清楚:方法:利用GEPIA网络工具、ENCORI泛癌分析平台和UALCAN-TCGA数据库研究FAM111B在ESCA中的表达。应用 CCK-8、血管生成、Transwell 和异种移植试验探讨 FAM111B 在 ESCA 中的生物学功能。应用Western印迹、RT-qPCR和RNA-seq分析研究FAM111B/GSDMA轴在ESCA细胞进展中的作用。CCK-8和异种移植试验用于研究FAM111B/GSDMA轴在决定ESCA对顺铂敏感性中的作用:结果:我们的研究结果表明,与正常组织相比,FAM111B在ESCA组织中高表达。结果:我们的研究结果表明,与正常组织相比,FAM111B在ESCCA组织中高表达。我们还发现,FAM111B通过与GSDMA结合促进ESCC细胞的进展,而胰蛋白酶蛋白酶结构域对FAM111B的活性至关重要。此外,我们还发现FAM111B/GSDMA轴调节ESCCA对顺铂的敏感性:总之,我们发现了一种新型的 FAM111B/GSDMA 轴,它至少在 ESCC 细胞中调控着 ESCA 的肿瘤发生和化疗敏感性。
{"title":"Overexpressed FAM111B degrades GSDMA to promote esophageal cancer tumorigenesis and cisplatin resistance.","authors":"Haiqin Wang, Haohui Wang, Jiajing Chen, Pian Liu, Xiaoxiong Xiao","doi":"10.1007/s13402-023-00871-0","DOIUrl":"10.1007/s13402-023-00871-0","url":null,"abstract":"<p><strong>Background: </strong>Chemotherapeutic agents such as cisplatin are commonly used in patients with clinically unresectable or recurrent esophageal cancer (ESCA). However, patients often develop resistance to cisplatin, which in turn leads to a poor prognosis. Studies have shown that FAM111B may be involved in the development of tumors as an oncogene or tumor suppressor gene. However, the pathological role and corresponding mechanism of FAM111B in ESCA are still unclear.</p><p><strong>Methods: </strong>The GEPIA web tool, ENCORI Pan-Cancer Analysis Platform and UALCAN-TCGA database were used to study the expression of FAM111B in ESCA. CCK-8, angiogenesis, Transwell and xenograft assays were applied to explore the biological function of FAM111B in ESCA. Western blot, RT-qPCR, and RNA-seq analyses were applied to study the FAM111B/GSDMA axis in the progression of ESCA cells. CCK-8 and xenograft assays were used to study the role of the FAM111B/GSDMA axis in determining the sensitivity of ESCA to cisplatin.</p><p><strong>Results: </strong>Our results demonstrated that FAM111B is highly expressed in ESCA tissues compared to normal tissues. We showed that FAM111B promotes the progression of ESCC cells by binding to GSDMA and that the trypsin protease domain is essential for the activity of FAM111B. Furthermore, we showed that the FAM111B/GSDMA axis regulates cisplatin sensitivity in ESCA.</p><p><strong>Conclusions: </strong>Overall, we identified a novel FAM111B/GSDMA axis regulating ESCA tumorigenesis and chemosensitivity, at least in ESCC cells.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"343-359"},"PeriodicalIF":6.6,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10534802","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Ferritinophagy induced ferroptosis in the management of cancer. 癌症治疗中的噬铁蛋白诱导铁变态反应。
IF 6.6 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-02-01 Epub Date: 2023-09-15 DOI: 10.1007/s13402-023-00858-x
Yi-Chen Liu, Yi-Ting Gong, Qing-Yan Sun, Bei Wang, Yue Yan, Yi-Xu Chen, Li-Jun Zhang, Wei-Dong Zhang, Xin Luan

Background: Ferroptosis, a newly form of regulated cell death (RCD), is characterized by iron dyshomeostasis and unrestricted lipid peroxidation. Emerging evidence depicts a pivotal role for ferroptosis in driving some pathological processes, especially in cancer. Triggering ferroptosis can suppress tumor growth and induce an anti-tumor immune response, denoting the therapeutic promises for targeting ferroptosis in the management of cancer. As an autophagic phenomenon, ferritinophagy is critical to induce ferroptosis by degradation of ferritin to release intracellular free iron. Recently, a great deal of effort has gone into designing and developing anti-cancer strategies based on targeting ferritinophagy to induce ferroptosis.

Conclusion: This review delineates the regulatory mechanism of ferritinophagy firstly and summarizes the role of ferritinophagy-induced ferroptosis in cancer. Moreover, the strategies targeting ferritinophagy to induce ferroptosis are highlighted to unveil the therapeutic value of ferritinophagy as a target to manage cancer. Finally, the future research directions on how to cope with the challenges in developing ferritinophagy promoters into clinical therapeutics are discussed.

背景:铁变态反应是一种新的调节性细胞死亡(RCD)形式,其特点是铁失衡和不受限制的脂质过氧化。新的证据表明,铁变态反应在某些病理过程中起着关键作用,尤其是在癌症中。触发铁变态反应可抑制肿瘤生长并诱导抗肿瘤免疫反应,这表明针对铁变态反应的癌症治疗大有可为。作为一种自噬现象,噬铁蛋白是通过降解铁蛋白释放细胞内游离铁诱导铁变态反应的关键。近来,人们在设计和开发基于靶向噬铁蛋白诱导铁变态反应的抗癌策略方面做出了大量努力:本综述首先阐述了噬铁蛋白的调控机制,并总结了噬铁蛋白诱导的铁变态反应在癌症中的作用。此外,还重点介绍了针对噬铁蛋白诱导铁变态反应的策略,以揭示噬铁蛋白作为治疗癌症靶点的治疗价值。最后,还讨论了未来的研究方向,即如何应对将嗜铁蛋白促进剂开发成临床疗法所面临的挑战。
{"title":"Ferritinophagy induced ferroptosis in the management of cancer.","authors":"Yi-Chen Liu, Yi-Ting Gong, Qing-Yan Sun, Bei Wang, Yue Yan, Yi-Xu Chen, Li-Jun Zhang, Wei-Dong Zhang, Xin Luan","doi":"10.1007/s13402-023-00858-x","DOIUrl":"10.1007/s13402-023-00858-x","url":null,"abstract":"<p><strong>Background: </strong>Ferroptosis, a newly form of regulated cell death (RCD), is characterized by iron dyshomeostasis and unrestricted lipid peroxidation. Emerging evidence depicts a pivotal role for ferroptosis in driving some pathological processes, especially in cancer. Triggering ferroptosis can suppress tumor growth and induce an anti-tumor immune response, denoting the therapeutic promises for targeting ferroptosis in the management of cancer. As an autophagic phenomenon, ferritinophagy is critical to induce ferroptosis by degradation of ferritin to release intracellular free iron. Recently, a great deal of effort has gone into designing and developing anti-cancer strategies based on targeting ferritinophagy to induce ferroptosis.</p><p><strong>Conclusion: </strong>This review delineates the regulatory mechanism of ferritinophagy firstly and summarizes the role of ferritinophagy-induced ferroptosis in cancer. Moreover, the strategies targeting ferritinophagy to induce ferroptosis are highlighted to unveil the therapeutic value of ferritinophagy as a target to manage cancer. Finally, the future research directions on how to cope with the challenges in developing ferritinophagy promoters into clinical therapeutics are discussed.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"19-35"},"PeriodicalIF":6.6,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10592229","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting ONECUT3 blocks glycolytic metabolism and potentiates anti-PD-1 therapy in pancreatic cancer. 靶向 ONECUT3 可阻断糖酵解代谢并增强胰腺癌的抗 PD-1 治疗。
IF 6.6 2区 医学 Q2 CELL BIOLOGY Pub Date : 2024-02-01 Epub Date: 2023-08-22 DOI: 10.1007/s13402-023-00852-3
Peng-Cheng Chen, Yong Ning, Hui Li, Jin-Gen Su, Jiang-Bo Shen, Qing-Chun Feng, Shu-Heng Jiang, Pei-Dong Shi, Run-Sheng Guo

Background: Reprogramming glucose metabolism, also known as the Warburg effect (aerobic glycolysis), is a hallmark of cancers. Increased tumor glycolysis not only favors rapid cancer cell proliferation but reprograms the immune microenvironment to enable tumor progression. The transcriptional factor ONECUT3 plays key roles in the development of the liver and pancreas, however, limited is known about its oncogenic roles, particularly metabolic reprogramming.

Methods: Immunohistochemistry and Western blotting are applied to determine the expression pattern of ONECUT3 and its clinical relevance in pancreatic ductal adenocarcinoma (PDAC). Knockdown and overexpression strategies are employed to determine the in vitro and in vivo functions of ONECUT3. Chromatin immunoprecipitation, luciferase reporter assay, and gene set enrichment analysis are used to decipher the molecular mechanisms.

Results: The glycolytic metabolism is inversely associated with T-cell infiltration in PDAC. ONECUT3 is identified as a key regulator for PDAC glycolysis and CD8+ T-cell infiltration. Genetic silencing of ONECUT3 inhibits cell proliferation, promotes cell apoptosis, and reduces glycolytic metabolism as evidenced by glucose uptake, lactate production, and extracellular acidification rate. Opposite effects of ONECUT3 are observed in overexpression studies. ONECUT3 enhances aerobic glycolysis via transcriptional regulation of PDK1. Targeting ONECUT3 effectively suppresses tumor growth, increases CD8+ T-cell infiltration, and potentiates anti-PD-1 therapy in PDAC. Pharmacological inhibition of PDK1 also shows a synergistic effect with anti-PD-1 therapy. In clinical setting, ONECUT3 is closely associated with PDK1 expression and T-cell infiltration in PDAC and acts as an independent prognostic factor.

Conclusions: Our study reveals a previous unprecedented regulatory role of ONECUT3 in PDAC glycolysis and provides in vivo evidence that increased glycolysis is linked to an immunosuppressive microenvironment. Moreover, targeting ONECUT3-PDK1 axis may serve as a promising therapeutic approach for the treatment of PDAC.

背景:葡萄糖代谢重编程,也称为沃伯格效应(有氧糖酵解),是癌症的一个特征。肿瘤糖酵解的增加不仅有利于癌细胞的快速增殖,还能重编程免疫微环境,使肿瘤得以进展。转录因子ONECUT3在肝脏和胰腺的发育过程中发挥着关键作用,然而,人们对它的致癌作用,尤其是代谢重编程作用了解有限:方法:应用免疫组化和 Western 印迹技术确定 ONECUT3 的表达模式及其在胰腺导管腺癌(PDAC)中的临床意义。采用基因敲除和过表达策略确定 ONECUT3 的体外和体内功能。染色质免疫共沉淀、荧光素酶报告分析和基因组富集分析被用来破译其分子机制:结果:糖代谢与 PDAC 中的 T 细胞浸润成反比。结果:糖酵解代谢与 PDAC 的 T 细胞浸润成反比,ONECUT3 是 PDAC 糖酵解和 CD8+ T 细胞浸润的关键调节因子。基因沉默ONECUT3可抑制细胞增殖、促进细胞凋亡并降低糖酵解代谢,葡萄糖摄取、乳酸生成和细胞外酸化率都证明了这一点。在过表达研究中观察到 ONECUT3 的相反作用。ONECUT3 通过转录调控 PDK1 增强有氧糖酵解。靶向 ONECUT3 能有效抑制肿瘤生长,增加 CD8+ T 细胞浸润,并增强 PDAC 的抗 PD-1 治疗效果。药理抑制 PDK1 还能与抗 PD-1 疗法产生协同效应。在临床环境中,ONECUT3与PDAC中PDK1的表达和T细胞浸润密切相关,是一个独立的预后因素:我们的研究揭示了 ONECUT3 在 PDAC 糖酵解中前所未有的调控作用,并提供了糖酵解增加与免疫抑制微环境相关的体内证据。此外,靶向 ONECUT3-PDK1 轴可能是治疗 PDAC 的一种有前景的治疗方法。
{"title":"Targeting ONECUT3 blocks glycolytic metabolism and potentiates anti-PD-1 therapy in pancreatic cancer.","authors":"Peng-Cheng Chen, Yong Ning, Hui Li, Jin-Gen Su, Jiang-Bo Shen, Qing-Chun Feng, Shu-Heng Jiang, Pei-Dong Shi, Run-Sheng Guo","doi":"10.1007/s13402-023-00852-3","DOIUrl":"10.1007/s13402-023-00852-3","url":null,"abstract":"<p><strong>Background: </strong>Reprogramming glucose metabolism, also known as the Warburg effect (aerobic glycolysis), is a hallmark of cancers. Increased tumor glycolysis not only favors rapid cancer cell proliferation but reprograms the immune microenvironment to enable tumor progression. The transcriptional factor ONECUT3 plays key roles in the development of the liver and pancreas, however, limited is known about its oncogenic roles, particularly metabolic reprogramming.</p><p><strong>Methods: </strong>Immunohistochemistry and Western blotting are applied to determine the expression pattern of ONECUT3 and its clinical relevance in pancreatic ductal adenocarcinoma (PDAC). Knockdown and overexpression strategies are employed to determine the in vitro and in vivo functions of ONECUT3. Chromatin immunoprecipitation, luciferase reporter assay, and gene set enrichment analysis are used to decipher the molecular mechanisms.</p><p><strong>Results: </strong>The glycolytic metabolism is inversely associated with T-cell infiltration in PDAC. ONECUT3 is identified as a key regulator for PDAC glycolysis and CD8<sup>+</sup> T-cell infiltration. Genetic silencing of ONECUT3 inhibits cell proliferation, promotes cell apoptosis, and reduces glycolytic metabolism as evidenced by glucose uptake, lactate production, and extracellular acidification rate. Opposite effects of ONECUT3 are observed in overexpression studies. ONECUT3 enhances aerobic glycolysis via transcriptional regulation of PDK1. Targeting ONECUT3 effectively suppresses tumor growth, increases CD8<sup>+</sup> T-cell infiltration, and potentiates anti-PD-1 therapy in PDAC. Pharmacological inhibition of PDK1 also shows a synergistic effect with anti-PD-1 therapy. In clinical setting, ONECUT3 is closely associated with PDK1 expression and T-cell infiltration in PDAC and acts as an independent prognostic factor.</p><p><strong>Conclusions: </strong>Our study reveals a previous unprecedented regulatory role of ONECUT3 in PDAC glycolysis and provides in vivo evidence that increased glycolysis is linked to an immunosuppressive microenvironment. Moreover, targeting ONECUT3-PDK1 axis may serve as a promising therapeutic approach for the treatment of PDAC.</p>","PeriodicalId":49223,"journal":{"name":"Cellular Oncology","volume":" ","pages":"81-96"},"PeriodicalIF":6.6,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"10039754","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Cellular Oncology
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1