首页 > 最新文献

Oncology Research最新文献

英文 中文
CES1 is associated with cisplatin resistance and poor prognosis of head and neck squamous cell carcinoma. CES1 与头颈部鳞状细胞癌的顺铂耐药性和不良预后有关。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-11-13 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.052244
Chuan Jiang, Chunlei Liu, X I Yao, Jingya Su, Wei Lu, Zhengbo Wei, Ying Xie

Background: Head and neck squamous cell carcinoma (HNSCC) is a prevalent form of cancer globally, with chemoresistance posing a major challenge in treatment outcomes. The efficacy of the commonly used chemotherapeutic agent, cisplatin, is diminished in patients with poor prognoses.

Methods: Various bioinformatics databases were utilized to examine Carboxylesterase 1 (CES1) gene expression, clinicopathologic features, patient survival analysis, and gene function. An organoid model of HNSCC was established, along with the induction of drug-resistant HNSCC in the organoid model. CES1 expression was assessed using qRT-PCR and Western Blot, and differential markers were identified through transcriptome sequencing. Knockdown and overexpression models of CES1 were created in SCC-9 and patient-derived organoid (PDO) cells using shRNA and lentivirus to investigate the tumor biology and cisplatin resistance associated with CES1.

Results: Research in bioinformatics has uncovered a strong correlation between the expression level of CES1 and the prognosis of HNSCC. The data suggests a significant link between CES1 expression and tobacco smoking. RNA-sequencing revealed a notable increase in CES1 expression in HNSCC-PDOcis-R cells compared to the parental PDO cells. Subsequently, we performed in vitro studies by HNSCC-PDO and SCC-9 and found that CES1-overexpressing cells exhibited reduced sensitivity to cisplatin and stronger tumor malignant biological behavior compared with CES1-knockdown cells.

Conclusion: The observed association between CES1 expression and tobacco smoking implies a potential influence of smoking on the efficacy of cisplatin-based chemotherapy in HNSCC through the regulation of CES1 expression.

背景:头颈部鳞状细胞癌(HNSCC)是一种全球流行的癌症,其化疗耐药性是治疗效果的一大挑战。常用化疗药物顺铂在预后不良的患者中疗效减弱:方法:利用各种生物信息学数据库研究羧基酯酶1(CES1)基因表达、临床病理特征、患者生存分析和基因功能。建立了HNSCC的类器官模型,并在类器官模型中诱导出耐药的HNSCC。利用 qRT-PCR 和 Western Blot 评估了 CES1 的表达,并通过转录组测序确定了差异标记。利用 shRNA 和慢病毒在 SCC-9 和患者衍生类器官(PDO)细胞中创建了 CES1 的敲除和过表达模型,以研究与 CES1 相关的肿瘤生物学和顺铂耐药性:生物信息学研究发现,CES1 的表达水平与 HNSCC 的预后密切相关。数据表明,CES1的表达与吸烟有重要关系。RNA测序显示,与亲本PDO细胞相比,HNSCC-PDOcis-R细胞中CES1的表达明显增加。随后,我们用 HNSCC-PDO 和 SCC-9 进行了体外研究,发现与 CES1 敲除的细胞相比,CES1 表达的细胞对顺铂的敏感性降低,肿瘤恶性生物学行为更强:结论:观察到的CES1表达与吸烟之间的关联意味着,吸烟可能会通过调控CES1的表达影响HNSCC中以顺铂为基础的化疗的疗效。
{"title":"CES1 is associated with cisplatin resistance and poor prognosis of head and neck squamous cell carcinoma.","authors":"Chuan Jiang, Chunlei Liu, X I Yao, Jingya Su, Wei Lu, Zhengbo Wei, Ying Xie","doi":"10.32604/or.2024.052244","DOIUrl":"https://doi.org/10.32604/or.2024.052244","url":null,"abstract":"<p><strong>Background: </strong>Head and neck squamous cell carcinoma (HNSCC) is a prevalent form of cancer globally, with chemoresistance posing a major challenge in treatment outcomes. The efficacy of the commonly used chemotherapeutic agent, cisplatin, is diminished in patients with poor prognoses.</p><p><strong>Methods: </strong>Various bioinformatics databases were utilized to examine Carboxylesterase 1 (CES1) gene expression, clinicopathologic features, patient survival analysis, and gene function. An organoid model of HNSCC was established, along with the induction of drug-resistant HNSCC in the organoid model. CES1 expression was assessed using qRT-PCR and Western Blot, and differential markers were identified through transcriptome sequencing. Knockdown and overexpression models of CES1 were created in SCC-9 and patient-derived organoid (PDO) cells using shRNA and lentivirus to investigate the tumor biology and cisplatin resistance associated with CES1.</p><p><strong>Results: </strong>Research in bioinformatics has uncovered a strong correlation between the expression level of CES1 and the prognosis of HNSCC. The data suggests a significant link between CES1 expression and tobacco smoking. RNA-sequencing revealed a notable increase in CES1 expression in HNSCC-PDO<sup>cis-R</sup> cells compared to the parental PDO cells. Subsequently, we performed <i>in vitro</i> studies by HNSCC-PDO and SCC-9 and found that CES1-overexpressing cells exhibited reduced sensitivity to cisplatin and stronger tumor malignant biological behavior compared with CES1-knockdown cells.</p><p><strong>Conclusion: </strong>The observed association between CES1 expression and tobacco smoking implies a potential influence of smoking on the efficacy of cisplatin-based chemotherapy in HNSCC through the regulation of CES1 expression.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"32 12","pages":"1935-1948"},"PeriodicalIF":2.0,"publicationDate":"2024-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11576922/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142687755","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Using Multi-Omics Analysis to Explore Diagnostic Tool and Optimize Drug Therapy Selection for Patients with Glioma Based on Cross-Talk Gene Signature. 利用多指标分析探索胶质瘤诊断工具并优化基于交叉基因特征的药物治疗选择
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-11-13 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.046191
Yushi Yang, Chujiao Hu, Shan Lei, Xin Bao, Zhirui Zeng, Wenpeng Cao

Background: The heterogeneity of prognosis and treatment benefits among patients with gliomas is due to tumor microenvironment characteristics. However, biomarkers that reflect microenvironmental characteristics and predict the prognosis of gliomas are limited. Therefore, we aimed to develop a model that can effectively predict prognosis, differentiate microenvironment signatures, and optimize drug selection for patients with glioma.

Materials and methods: The CIBERSORT algorithm, bulk sequencing analysis, and single-cell RNA (scRNA) analysis were employed to identify significant cross-talk genes between M2 macrophages and cancer cells in glioma tissues. A predictive model was constructed based on cross-talk gene expression, and its effect on prognosis, recurrence prediction, and microenvironment characteristics was validated in multiple cohorts. The effect of the predictive model on drug selection was evaluated using the OncoPredict algorithm and relevant cellular biology experiments.

Results: A high abundance of M2 macrophages in glioma tissues indicates poor prognosis, and cross-talk between macrophages and cancer cells plays a crucial role in shaping the tumor microenvironment. Eight genes involved in the cross-talk between macrophages and cancer cells were identified. Among them, periostin (POSTN), chitinase 3 like 1 (CHI3L1), serum amyloid A1 (SAA1), and matrix metallopeptidase 9 (MMP9) were selected to construct a predictive model. The developed model demonstrated significant efficacy in distinguishing patient prognosis, recurrent cases, and characteristics of high inflammation, hypoxia, and immunosuppression. Furthermore, this model can serve as a valuable tool for guiding the use of trametinib.

Conclusions: In summary, this study provides a comprehensive understanding of the interplay between M2 macrophages and cancer cells in glioma; utilizes a cross-talk gene signature to develop a predictive model that can predict the differentiation of patient prognosis, recurrence instances, and microenvironment characteristics; and aids in optimizing the application of trametinib in glioma patients.

背景:胶质瘤患者的预后和治疗获益的异质性是由肿瘤微环境特征造成的。然而,能反映胶质瘤微环境特征并预测其预后的生物标志物非常有限。因此,我们旨在开发一种能有效预测预后、区分微环境特征并优化胶质瘤患者药物选择的模型:采用CIBERSORT算法、批量测序分析和单细胞RNA(scRNA)分析来确定胶质瘤组织中M2巨噬细胞和癌细胞之间的重要交叉基因。根据交叉对话基因的表达构建了预测模型,并在多个队列中验证了该模型对预后、复发预测和微环境特征的影响。利用OncoPredict算法和相关的细胞生物学实验评估了预测模型对药物选择的影响:结果:胶质瘤组织中M2巨噬细胞的高丰度预示着预后不良,巨噬细胞和癌细胞之间的交叉对话在肿瘤微环境的形成中起着至关重要的作用。研究发现了 8 个参与巨噬细胞和癌细胞之间交叉对话的基因。在这些基因中,我们选择了骨膜增生蛋白(POSTN)、几丁质酶 3 like 1(CHI3L1)、血清淀粉样蛋白 A1(SAA1)和基质金属肽酶 9(MMP9)来构建预测模型。所建立的模型在区分患者预后、复发病例以及高炎症、缺氧和免疫抑制等特征方面具有显著疗效。此外,该模型还可作为指导使用曲美替尼的重要工具:综上所述,本研究全面了解了胶质瘤中M2巨噬细胞与癌细胞之间的相互作用;利用交叉对话基因特征建立了一个预测模型,可预测患者预后、复发情况和微环境特征的分化;有助于优化曲美替尼在胶质瘤患者中的应用。
{"title":"Using Multi-Omics Analysis to Explore Diagnostic Tool and Optimize Drug Therapy Selection for Patients with Glioma Based on Cross-Talk Gene Signature.","authors":"Yushi Yang, Chujiao Hu, Shan Lei, Xin Bao, Zhirui Zeng, Wenpeng Cao","doi":"10.32604/or.2024.046191","DOIUrl":"https://doi.org/10.32604/or.2024.046191","url":null,"abstract":"<p><strong>Background: </strong>The heterogeneity of prognosis and treatment benefits among patients with gliomas is due to tumor microenvironment characteristics. However, biomarkers that reflect microenvironmental characteristics and predict the prognosis of gliomas are limited. Therefore, we aimed to develop a model that can effectively predict prognosis, differentiate microenvironment signatures, and optimize drug selection for patients with glioma.</p><p><strong>Materials and methods: </strong>The CIBERSORT algorithm, bulk sequencing analysis, and single-cell RNA (scRNA) analysis were employed to identify significant cross-talk genes between M2 macrophages and cancer cells in glioma tissues. A predictive model was constructed based on cross-talk gene expression, and its effect on prognosis, recurrence prediction, and microenvironment characteristics was validated in multiple cohorts. The effect of the predictive model on drug selection was evaluated using the OncoPredict algorithm and relevant cellular biology experiments.</p><p><strong>Results: </strong>A high abundance of M2 macrophages in glioma tissues indicates poor prognosis, and cross-talk between macrophages and cancer cells plays a crucial role in shaping the tumor microenvironment. Eight genes involved in the cross-talk between macrophages and cancer cells were identified. Among them, periostin (<i>POSTN</i>), chitinase 3 like 1 (<i>CHI3L1</i>), serum amyloid A1 (<i>SAA1</i>), and matrix metallopeptidase 9 (<i>MMP9</i>) were selected to construct a predictive model. The developed model demonstrated significant efficacy in distinguishing patient prognosis, recurrent cases, and characteristics of high inflammation, hypoxia, and immunosuppression. Furthermore, this model can serve as a valuable tool for guiding the use of trametinib.</p><p><strong>Conclusions: </strong>In summary, this study provides a comprehensive understanding of the interplay between M2 macrophages and cancer cells in glioma; utilizes a cross-talk gene signature to develop a predictive model that can predict the differentiation of patient prognosis, recurrence instances, and microenvironment characteristics; and aids in optimizing the application of trametinib in glioma patients.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"32 12","pages":"1921-1934"},"PeriodicalIF":2.0,"publicationDate":"2024-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11576925/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142687913","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
LncRNA AFAP1-AS1 exhibits oncogenic characteristics and promotes gemcitabine-resistance of cervical cancer cells through miR-7-5p/EGFR axis. LncRNA AFAP1-AS1 具有致癌特性,并通过 miR-7-5p/EGFR 轴促进宫颈癌细胞对吉西他滨的耐药性。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-11-13 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.044547
Chaoqun Wang, Ting Zhang, Chaohe Zhang

Background: Drug resistance is the main factor contributing to cancer recurrence and poor prognosis. Exploration of drug resistance-related mechanisms and effective therapeutic targets are the aim of molecular targeted therapy. In our study, the role of long non-coding RNA (lncRNA) AFAP1-AS1 in gemcitabine resistance and related mechanisms were explored in cervical cancer cells.

Methods: Gemcitabine-resistant cervical cancer cell lines HT-3-Gem and SW756-Gem were constructed using the gemcitabine concentration gradient method. The overall survival rates and recurrence-free survival rates were evaluated by Kaplan-Meier analysis. The interaction was verified through a Dual-luciferase reporter gene assay and a Biotinylated RNA pull-down assay. Cell proliferation ability was assessed through methyl-thiazolyl-tetrazolium (MTT), soft agar, and colony formation experiments. Cell cycle and apoptosis were detected by flow cytometry.

Results: Up-regulation of AFAP1-AS1 in cervical cancer predicted a poor prognosis. Besides, patients in the gemcitabine-resistance group had higher levels of AFAP1-AS1 than the gemcitabine-sensitive group. AFAP1-AS1 promoted tumor growth and induced gemcitabine tolerance of cervical cancer cells. In addition, AFAP1-AS1 mediated epidermal growth factor receptor (EGFR) expression by serving as a molecular sponge for microRNA-7a-5p (miR-7-5p). This present study also proved that the knockdown of EGFR or overexpression of miR-7a-5p abolished the accelerative role of AFAP1-AS1 overexpression in cancer progression and gemcitabine tolerance.

Conclusions: In general, the AFAP1-AS1/miR-7-5p/EGFR axis was tightly related to the progression and gemcitabine tolerance of cervical cancer, providing potential targets for the management of cervical cancer.

背景:耐药性是导致癌症复发和预后不良的主要因素。探索耐药相关机制和有效治疗靶点是分子靶向治疗的目标。本研究探讨了宫颈癌细胞中长非编码 RNA(lncRNA)AFAP1-AS1 在吉西他滨耐药中的作用及相关机制:方法:采用吉西他滨浓度梯度法构建了吉西他滨耐药的宫颈癌细胞系HT-3-Gem和SW756-Gem。采用 Kaplan-Meier 分析法评估了总生存率和无复发生存率。通过双荧光素酶报告基因试验和生物素化 RNA 拉取试验验证了相互作用。细胞增殖能力通过甲基-噻唑基-四唑啉(MTT)、软琼脂和菌落形成实验进行评估。流式细胞术检测细胞周期和细胞凋亡:结果:AFAP1-AS1在宫颈癌中的上调预示着预后不良。此外,吉西他滨耐药组患者的AFAP1-AS1水平高于吉西他滨敏感组。AFAP1-AS1能促进肿瘤生长,诱导宫颈癌细胞对吉西他滨耐受。此外,AFAP1-AS1通过充当microRNA-7a-5p(miR-7-5p)的分子海绵,介导表皮生长因子受体(EGFR)的表达。本研究还证明,敲除表皮生长因子受体或过表达 miR-7a-5p 可消除 AFAP1-AS1 过表达在癌症进展和吉西他滨耐受中的加速作用:总的来说,AFAP1-AS1/miR-7-5p/EGFR轴与宫颈癌的进展和吉西他滨耐受性密切相关,为宫颈癌的治疗提供了潜在靶点。
{"title":"LncRNA AFAP1-AS1 exhibits oncogenic characteristics and promotes gemcitabine-resistance of cervical cancer cells through miR-7-5p/EGFR axis.","authors":"Chaoqun Wang, Ting Zhang, Chaohe Zhang","doi":"10.32604/or.2024.044547","DOIUrl":"https://doi.org/10.32604/or.2024.044547","url":null,"abstract":"<p><strong>Background: </strong>Drug resistance is the main factor contributing to cancer recurrence and poor prognosis. Exploration of drug resistance-related mechanisms and effective therapeutic targets are the aim of molecular targeted therapy. In our study, the role of long non-coding RNA (lncRNA) AFAP1-AS1 in gemcitabine resistance and related mechanisms were explored in cervical cancer cells.</p><p><strong>Methods: </strong>Gemcitabine-resistant cervical cancer cell lines HT-3-Gem and SW756-Gem were constructed using the gemcitabine concentration gradient method. The overall survival rates and recurrence-free survival rates were evaluated by Kaplan-Meier analysis. The interaction was verified through a Dual-luciferase reporter gene assay and a Biotinylated RNA pull-down assay. Cell proliferation ability was assessed through methyl-thiazolyl-tetrazolium (MTT), soft agar, and colony formation experiments. Cell cycle and apoptosis were detected by flow cytometry.</p><p><strong>Results: </strong>Up-regulation of AFAP1-AS1 in cervical cancer predicted a poor prognosis. Besides, patients in the gemcitabine-resistance group had higher levels of AFAP1-AS1 than the gemcitabine-sensitive group. AFAP1-AS1 promoted tumor growth and induced gemcitabine tolerance of cervical cancer cells. In addition, AFAP1-AS1 mediated epidermal growth factor receptor (EGFR) expression by serving as a molecular sponge for microRNA-7a-5p (miR-7-5p). This present study also proved that the knockdown of EGFR or overexpression of miR-7a-5p abolished the accelerative role of AFAP1-AS1 overexpression in cancer progression and gemcitabine tolerance.</p><p><strong>Conclusions: </strong>In general, the AFAP1-AS1/miR-7-5p/EGFR axis was tightly related to the progression and gemcitabine tolerance of cervical cancer, providing potential targets for the management of cervical cancer.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"32 12","pages":"1867-1879"},"PeriodicalIF":2.0,"publicationDate":"2024-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11576921/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142687903","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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 superiority of PMFs on reversing drug resistance of colon cancer and the effect on aerobic glycolysis-ROS-autophagy signaling axis. PMFs 在逆转结肠癌耐药性方面的优势以及对有氧糖酵解-ROS-自噬信号轴的影响。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-11-13 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.048778
Yuqin Yin, Y U Wu, Hongliang Huang, Yingying Duan, Zhongwen Yuan, Lihui Cao, Jinjin Ying, Yongheng Zhou, Senling Feng

Background: Polymethoxylated flavones (PMFs) are compounds present in citrus peels and other Rutaceae plants, which exhibit diverse biological activities, including robust antitumor and antioxidant effects. However, the mechanism of PMFs in reversing drug resistance to colon cancer remains unknown. In the present study, we aimed to investigate the potential connection between the aerobic glycolysis-ROS-autophagy signaling axis and the reversal of PTX resistance in colon cancer by PMFs.

Methods: MTT Cell viability assay and colony formation assay were used to investigate the effect of PMFs combined with PTX in reversing HCT8/T cell resistance ex vivo; the mRNA and protein levels of the target were detected by SDS-PAGE (sodium dodecyl sulfate-polyacrylamide gel electrophoresis), quantitative real-time fluorescence polymerase chain reaction (qRT-PCR) and Western blot protein immunoblotting (WB); An HCT8/T cell xenograft model was established to investigate the MDR reversal activity of PMFs in vivo; The extracellular acidification rate (ECAR) and the oxygen consumption rate (OCR) were detected to assess the cellular oxygen consumption rate and glycolytic process.

Results: HCT8/T cells demonstrated significant resistance to PTX, up-regulating the expression levels of ABCB1 mRNA, P-gp, LC3-I, and LC3-II protein, and increasing intracellular reactive oxygen species (ROS) content. PMFs mainly contain two active ingredients, nobiletin, and tangeretin, which were able to reverse drug resistance in HCT8/T cells in a concentration-dependent manner. PMFs exhibited high tolerance in the HCT8/T nude mouse model while increasing the sensitivity of PTX-resistant cells and suppressing tumor growth significantly. PMFs combined with PTX reduced extracellular acidification rate (ECAR) and oxygen consumption rate (OCR) in HCT8/T cells. Additionally, PMFs reduced intracellular ROS content, down-regulated the expression levels of autophagy-related proteins LC3-I, LC3-II, Beclin1, and ATG7, and significantly reduced the number of autophagosomes in HCT8/T cells.

Conclusions: The present study demonstrated that PMFs could potentially reverse PTX resistance in colon cancer by regulating the aerobic glycolysis-ROS-autophagy signaling axis, which indicated that PMFs would be potential potentiators for future chemotherapeutic agents in colon cancer.

背景:聚甲氧基黄酮(PMFs)是存在于柑橘皮和其他芸香科植物中的化合物,具有多种生物活性,包括强大的抗肿瘤和抗氧化作用。然而,PMFs 在逆转结肠癌耐药性方面的作用机制尚不清楚。在本研究中,我们旨在探讨有氧糖酵解-ROS-自噬信号轴与PMFs逆转结肠癌PTX耐药性之间的潜在联系:方法:采用MTT细胞活力检测和菌落形成检测研究PMFs联合PTX对HCT8/T细胞体内耐药性的逆转作用;采用SDS-PAGE(十二烷基硫酸钠-聚丙烯酰胺凝胶电泳)、实时荧光定量聚合酶链式反应(qRT-PCR)和Western blot蛋白免疫印迹(WB)检测靶标的mRNA和蛋白水平;建立HCT8/T细胞异种移植模型,研究PMFs在体内的MDR逆转活性;检测细胞外酸化率(ECAR)和耗氧量(OCR),评估细胞耗氧量和糖酵解过程。结果HCT8/T细胞对PTX表现出明显的耐药性,上调了ABCB1 mRNA、P-gp、LC3-I和LC3-II蛋白的表达水平,增加了细胞内活性氧(ROS)含量。PMFs主要含有两种活性成分:金霉素和桔皮苷,它们能够以浓度依赖性的方式逆转HCT8/T细胞的耐药性。PMFs在HCT8/T裸鼠模型中表现出较高的耐受性,同时提高了PTX耐药细胞的敏感性,并显著抑制了肿瘤的生长。PMFs 与 PTX 联用可降低 HCT8/T 细胞的细胞外酸化率(ECAR)和耗氧量(OCR)。此外,PMFs还能降低细胞内ROS含量,下调自噬相关蛋白LC3-I、LC3-II、Beclin1和ATG7的表达水平,并显著减少HCT8/T细胞中自噬体的数量:本研究表明,PMFs可通过调节有氧糖酵解-ROS-自噬信号轴逆转结肠癌对PTX的耐药性,这表明PMFs将成为未来结肠癌化疗药物的潜在增效剂。
{"title":"The superiority of PMFs on reversing drug resistance of colon cancer and the effect on aerobic glycolysis-ROS-autophagy signaling axis.","authors":"Yuqin Yin, Y U Wu, Hongliang Huang, Yingying Duan, Zhongwen Yuan, Lihui Cao, Jinjin Ying, Yongheng Zhou, Senling Feng","doi":"10.32604/or.2024.048778","DOIUrl":"https://doi.org/10.32604/or.2024.048778","url":null,"abstract":"<p><strong>Background: </strong>Polymethoxylated flavones (PMFs) are compounds present in citrus peels and other Rutaceae plants, which exhibit diverse biological activities, including robust antitumor and antioxidant effects. However, the mechanism of PMFs in reversing drug resistance to colon cancer remains unknown. In the present study, we aimed to investigate the potential connection between the aerobic glycolysis-ROS-autophagy signaling axis and the reversal of PTX resistance in colon cancer by PMFs.</p><p><strong>Methods: </strong>MTT Cell viability assay and colony formation assay were used to investigate the effect of PMFs combined with PTX in reversing HCT8/T cell resistance <i>ex vivo</i>; the mRNA and protein levels of the target were detected by SDS-PAGE (sodium dodecyl sulfate-polyacrylamide gel electrophoresis), quantitative real-time fluorescence polymerase chain reaction (qRT-PCR) and Western blot protein immunoblotting (WB); An HCT8/T cell xenograft model was established to investigate the MDR reversal activity of PMFs <i>in vivo</i>; The extracellular acidification rate (ECAR) and the oxygen consumption rate (OCR) were detected to assess the cellular oxygen consumption rate and glycolytic process.</p><p><strong>Results: </strong>HCT8/T cells demonstrated significant resistance to PTX, up-regulating the expression levels of ABCB1 mRNA, P-gp, LC3-I, and LC3-II protein, and increasing intracellular reactive oxygen species (ROS) content. PMFs mainly contain two active ingredients, nobiletin, and tangeretin, which were able to reverse drug resistance in HCT8/T cells in a concentration-dependent manner. PMFs exhibited high tolerance in the HCT8/T nude mouse model while increasing the sensitivity of PTX-resistant cells and suppressing tumor growth significantly. PMFs combined with PTX reduced extracellular acidification rate (ECAR) and oxygen consumption rate (OCR) in HCT8/T cells. Additionally, PMFs reduced intracellular ROS content, down-regulated the expression levels of autophagy-related proteins LC3-I, LC3-II, Beclin1, and ATG7, and significantly reduced the number of autophagosomes in HCT8/T cells.</p><p><strong>Conclusions: </strong>The present study demonstrated that PMFs could potentially reverse PTX resistance in colon cancer by regulating the aerobic glycolysis-ROS-autophagy signaling axis, which indicated that PMFs would be potential potentiators for future chemotherapeutic agents in colon cancer.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"32 12","pages":"1891-1902"},"PeriodicalIF":2.0,"publicationDate":"2024-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11576955/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142687908","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
A comprehensive and systematic analysis of Dihydrolipoamide S-acetyltransferase (DLAT) as a novel prognostic biomarker in pan-cancer and glioma. 二氢脂酰胺 S-乙酰转移酶(DLAT)作为泛癌和胶质瘤新型预后生物标记物的全面系统分析。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-11-13 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.048138
Hui Zhou, Zhengyu Yu, Jing Xu, Zhongwang Wang, Yali Tao, Jinjin Wang, Peipei Yang, Jinrong Yang, Ting Niu

Background: Dihydrolipoamide S-acetyltransferase (DLAT) is a subunit of the pyruvate dehydrogenase complex (PDC), a rate-limiting enzyme complex, that can participate in either glycolysis or the tricarboxylic acid cycle (TCA). However, the pathogenesis is not fully understood. We aimed to perform a more systematic and comprehensive analysis of DLAT in the occurrence and progression of tumors, and to investigate its function in patients' prognosis and immunotherapy.

Methods: The differential expression, diagnosis, prognosis, genetic and epigenetic alterations, tumor microenvironment, stemness, immune infiltration cells, function enrichment, single-cell analysis, and drug response across cancers were conducted based on multiple computational tools. Additionally, we validated its carcinogenic effect and possible mechanism in glioma cells.

Results: We exhibited that DLAT expression was increased in most tumors, especially in glioma, and affected the survival of tumor patients. DLAT was related to RNA modification genes, DNA methylation, immune infiltration, and immune infiltration cells, including CD4+ T cells, CD8+ T cells, Tregs, and cancer-associated fibroblasts. Single-cell analysis displayed that DLAT might regulate cancer by mediating angiogenesis, inflammation, and stemness. Enrichment analysis revealed that DLAT might take part in the cell cycle pathway. Increased expression of DLAT leads tumor cells to be more resistant to many kinds of compounds, including PI3Kβ inhibitors, PKC inhibitors, HSP90 inhibitors, and MEK inhibitors. In addition, glioma cells with DLAT silence inhibited proliferation, migration, and invasion ability, and promoted cell apoptosis.

Conclusion: We conducted a comprehensive analysis of DLAT in the occurrence and progression of tumors, and its possible functions and mechanisms. DLAT is a potential diagnostic, prognostic, and immunotherapeutic biomarker for cancer patients.

背景:二氢脂酰胺 S-乙酰转移酶(DLAT)是丙酮酸脱氢酶复合物(PDC)的一个亚基,PDC 是一种限速酶复合物,可参与糖酵解或三羧酸循环(TCA)。然而,其发病机制尚未完全明了。我们旨在对 DLAT 在肿瘤发生和发展过程中的作用进行更系统、更全面的分析,并研究其在患者预后和免疫治疗中的功能:方法:基于多种计算工具,对不同癌症的差异表达、诊断、预后、遗传和表观遗传学改变、肿瘤微环境、干细胞、免疫浸润细胞、功能富集、单细胞分析和药物反应进行了研究。此外,我们还验证了它在胶质瘤细胞中的致癌作用和可能的机制:结果:我们发现 DLAT 在大多数肿瘤中表达增加,尤其是在胶质瘤中,并影响肿瘤患者的生存。DLAT 与 RNA 修饰基因、DNA 甲基化、免疫浸润以及免疫浸润细胞(包括 CD4+ T 细胞、CD8+ T 细胞、Tregs 和癌症相关成纤维细胞)有关。单细胞分析显示,DLAT可能通过介导血管生成、炎症和干细胞来调控癌症。富集分析显示,DLAT可能参与了细胞周期途径。DLAT表达的增加会导致肿瘤细胞对多种化合物(包括PI3Kβ抑制剂、PKC抑制剂、HSP90抑制剂和MEK抑制剂)产生抗药性。此外,DLAT沉默的胶质瘤细胞具有抑制增殖、迁移和侵袭能力,促进细胞凋亡的作用:我们全面分析了DLAT在肿瘤发生和发展中的作用及其可能的功能和机制。DLAT是一种潜在的癌症诊断、预后和免疫治疗生物标记物。
{"title":"A comprehensive and systematic analysis of Dihydrolipoamide S-acetyltransferase <i>(DLAT)</i> as a novel prognostic biomarker in pan-cancer and glioma.","authors":"Hui Zhou, Zhengyu Yu, Jing Xu, Zhongwang Wang, Yali Tao, Jinjin Wang, Peipei Yang, Jinrong Yang, Ting Niu","doi":"10.32604/or.2024.048138","DOIUrl":"https://doi.org/10.32604/or.2024.048138","url":null,"abstract":"<p><strong>Background: </strong>Dihydrolipoamide S-acetyltransferase (<i>DLAT</i>) is a subunit of the pyruvate dehydrogenase complex (PDC), a rate-limiting enzyme complex, that can participate in either glycolysis or the tricarboxylic acid cycle (TCA). However, the pathogenesis is not fully understood. We aimed to perform a more systematic and comprehensive analysis of <i>DLAT</i> in the occurrence and progression of tumors, and to investigate its function in patients' prognosis and immunotherapy.</p><p><strong>Methods: </strong>The differential expression, diagnosis, prognosis, genetic and epigenetic alterations, tumor microenvironment, stemness, immune infiltration cells, function enrichment, single-cell analysis, and drug response across cancers were conducted based on multiple computational tools. Additionally, we validated its carcinogenic effect and possible mechanism in glioma cells.</p><p><strong>Results: </strong>We exhibited that <i>DLAT</i> expression was increased in most tumors, especially in glioma, and affected the survival of tumor patients. <i>DLAT</i> was related to RNA modification genes, DNA methylation, immune infiltration, and immune infiltration cells, including CD4+ T cells, CD8+ T cells, Tregs, and cancer-associated fibroblasts. Single-cell analysis displayed that <i>DLAT</i> might regulate cancer by mediating angiogenesis, inflammation, and stemness. Enrichment analysis revealed that <i>DLAT</i> might take part in the cell cycle pathway. Increased expression of <i>DLAT</i> leads tumor cells to be more resistant to many kinds of compounds, including PI3Kβ inhibitors, PKC inhibitors, HSP90 inhibitors, and MEK inhibitors. In addition, glioma cells with <i>DLAT</i> silence inhibited proliferation, migration, and invasion ability, and promoted cell apoptosis.</p><p><strong>Conclusion: </strong>We conducted a comprehensive analysis of <i>DLAT</i> in the occurrence and progression of tumors, and its possible functions and mechanisms. <i>DLAT</i> is a potential diagnostic, prognostic, and immunotherapeutic biomarker for cancer patients.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"32 12","pages":"1903-1919"},"PeriodicalIF":2.0,"publicationDate":"2024-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11576973/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142687769","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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 M2 macrophage-related genes for establishing a prognostic model in pancreatic cancer: FCGR3A as key gene. 鉴定与 M2 巨噬细胞相关的基因以建立胰腺癌预后模型:FCGR3A是关键基因
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-11-13 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.055286
Zhen Wang, Jun Fu, Saisai Zhu, Haodong Tang, Kui Shi, Jihua Yang, Meng Wang, Mengge Wu, Dunfeng Qi

Background: Pancreatic ductal adenocarcinoma (PDAC) has a rich and complex tumor immune microenvironment (TIME). M2 macrophages are among the most extensively infiltrated immune cells in the TIME and are necessary for the growth and migration of cancers. However, the mechanisms and targets mediating M2 macrophage infiltration in pancreatic cancer remain elusive.

Methods: The M2 macrophage infiltration score of patients was assessed using the xCell algorithm. Using weighted gene co-expression network analysis (WGCNA), module genes associated with M2 macrophages were identified, and a predictive model was designed. The variations in immunological cell patterns, cancer mutations, and enrichment pathways between the cohorts with the high- and low-risk were examined. Additionally, the expression of FCGR3A and RNASE2, as well as their association with M2 macrophages were evaluated using the HPA, TNMplot, and GEPIA2 databases and verified by tissue immunofluorescence staining. Moreover, in vitro cell experiments were conducted, where FCGR3A was knocked down in pancreatic cancer cells using siRNA to analyze its effects on M2 macrophage infiltration, tumor proliferation, and metastasis.

Results: The prognosis of patients in high-risk and low-risk groups was successfully distinguished using a prognostic risk score model of M2 macrophage-related genes (p = 0.024). Between the high- and low-risk cohorts, there have been notable variations in immune cell infiltration patterns, tumor mutations, and biological functions. The risk score was linked to the manifestation of prevalent immunological checkpoints, immunological scores, and stroma values (all p < 0.05). In vitro experiments and tissue immunofluorescence staining revealed that FCGR3A can promote the infiltration or polarization of M2 macrophages and enhance tumor proliferation and migration.

Conclusions: In this study, an M2 macrophage-related pancreatic cancer risk score model was established, and found that FCGR3A was correlated with tumor formation, metastasis, and M2 macrophage infiltration.

背景:胰腺导管腺癌(PDAC)具有丰富而复杂的肿瘤免疫微环境(TIME)。M2 巨噬细胞是 TIME 中浸润最广泛的免疫细胞之一,是癌症生长和迁移所必需的。然而,胰腺癌中介导 M2 巨噬细胞浸润的机制和靶点仍未确定:方法:使用 xCell 算法评估患者的 M2 巨噬细胞浸润评分。利用加权基因共表达网络分析(WGCNA),确定了与 M2 巨噬细胞相关的模块基因,并设计了一个预测模型。研究了高风险和低风险队列之间在免疫细胞模式、癌症突变和富集途径方面的差异。此外,还利用 HPA、TNMplot 和 GEPIA2 数据库评估了 FCGR3A 和 RNASE2 的表达及其与 M2 巨噬细胞的关联,并通过组织免疫荧光染色进行了验证。此外,还进行了体外细胞实验,使用 siRNA 敲除胰腺癌细胞中的 FCGR3A,分析其对 M2 巨噬细胞浸润、肿瘤增殖和转移的影响:结果:利用M2巨噬细胞相关基因的预后风险评分模型,成功区分了高危和低危组患者的预后(p = 0.024)。高危和低危组之间在免疫细胞浸润模式、肿瘤突变和生物功能方面存在显著差异。风险评分与流行的免疫学检查点表现、免疫学评分和基质值有关(均 p <0.05)。体外实验和组织免疫荧光染色显示,FCGR3A能促进M2巨噬细胞的浸润或极化,并增强肿瘤的增殖和迁移:本研究建立了M2巨噬细胞相关胰腺癌风险评分模型,发现FCGR3A与肿瘤形成、转移和M2巨噬细胞浸润相关。
{"title":"Identification of M2 macrophage-related genes for establishing a prognostic model in pancreatic cancer: <i>FCGR3A</i> as key gene.","authors":"Zhen Wang, Jun Fu, Saisai Zhu, Haodong Tang, Kui Shi, Jihua Yang, Meng Wang, Mengge Wu, Dunfeng Qi","doi":"10.32604/or.2024.055286","DOIUrl":"https://doi.org/10.32604/or.2024.055286","url":null,"abstract":"<p><strong>Background: </strong>Pancreatic ductal adenocarcinoma (PDAC) has a rich and complex tumor immune microenvironment (TIME). M2 macrophages are among the most extensively infiltrated immune cells in the TIME and are necessary for the growth and migration of cancers. However, the mechanisms and targets mediating M2 macrophage infiltration in pancreatic cancer remain elusive.</p><p><strong>Methods: </strong>The M2 macrophage infiltration score of patients was assessed using the xCell algorithm. Using weighted gene co-expression network analysis (WGCNA), module genes associated with M2 macrophages were identified, and a predictive model was designed. The variations in immunological cell patterns, cancer mutations, and enrichment pathways between the cohorts with the high- and low-risk were examined. Additionally, the expression of FCGR3A and RNASE2, as well as their association with M2 macrophages were evaluated using the HPA, TNMplot, and GEPIA2 databases and verified by tissue immunofluorescence staining. Moreover, <i>in vitro</i> cell experiments were conducted, where FCGR3A was knocked down in pancreatic cancer cells using siRNA to analyze its effects on M2 macrophage infiltration, tumor proliferation, and metastasis.</p><p><strong>Results: </strong>The prognosis of patients in high-risk and low-risk groups was successfully distinguished using a prognostic risk score model of M2 macrophage-related genes (<i>p</i> = 0.024). Between the high- and low-risk cohorts, there have been notable variations in immune cell infiltration patterns, tumor mutations, and biological functions. The risk score was linked to the manifestation of prevalent immunological checkpoints, immunological scores, and stroma values (all <i>p</i> < 0.05). <i>In vitro</i> experiments and tissue immunofluorescence staining revealed that FCGR3A can promote the infiltration or polarization of M2 macrophages and enhance tumor proliferation and migration.</p><p><strong>Conclusions: </strong>In this study, an M2 macrophage-related pancreatic cancer risk score model was established, and found that FCGR3A was correlated with tumor formation, metastasis, and M2 macrophage infiltration.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"32 12","pages":"1851-1866"},"PeriodicalIF":2.0,"publicationDate":"2024-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11576954/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142687851","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Caffeic acid hinders the proliferation and migration through inhibition of IL-6 mediated JAK-STAT-3 signaling axis in human prostate cancer. 咖啡酸通过抑制 IL-6 介导的 JAK-STAT-3 信号轴阻碍人类前列腺癌的增殖和迁移。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-11-13 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.048007
Yuan Yin, Zhengyin Wang, Yujie Hu, Jia Wang, Y I Wang, Qun Lu

Background: Caffeic acid (CA) is considered a promising phytochemical that has inhibited numerous cancer cell proliferation. Therefore, it is gaining increasing attention due to its safe and pharmacological applications. In this study, we investigated the role of CA in inhibiting the Interleukin-6 (IL-6)/Janus kinase (JAK)/Signal transducer and activator of transcription-3 (STAT-3) mediated suppression of the proliferation signaling in human prostate cancer cells.

Materials and methods: The role of CA in proliferation and colony formation abilities was studied using 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) assay and colony formation assays. Tumour cell death and cell cycle arrest were identified using flow cytometry techniques. CA treatment-associated protein expression of mitogen-activated protein kinase (MAPK) families, IL-6/JAK/STAT-3, proliferation, and apoptosis protein expressions in PC-3 and LNCaP cell lines were measured using Western blot investigation.

Results: We have obtained that treatment with CA inhibits prostate cancer cells (PC-3 and LNCaP) proliferation and induces reactive oxygen species (ROS), cell cycle arrest, and apoptosis cell death in a concentration-dependent manner. Moreover, CA treatment alleviates the expression phosphorylated form of MAPK families, i.e., extracellular signal-regulated kinase 1 (ERK1), c-Jun N-terminal kinase (JNK), and p38 in PC-3 cells. IL-6 mediated JAK/STAT3 expressions regulate the proliferation and antiapoptosis that leads to prostate cancer metastasis and migration. Therefore, to mitigate the expression of IL-6/JAK/STAT-3 is considered an important target for the treatment of prostate cancer. In this study, we have observed that CA inhibits the expression of IL-6, JAK1, and phosphorylated STAT-3 in both PC-3 and LNCaP cells. Due to the inhibitory effect of IL-6/JAK/STAT-3, it resulted in decreased expression of cyclin-D1, cyclin-D2, and CDK1 in both PC-3 cells. In addition, CA induces apoptosis by enhancing the expression of Bax and caspase-3; and decreased expression of Bcl-2 in prostate cancer cells.

Conclusions: Thus, CA might act as a therapeutical application against prostate cancer by targeting the IL-6/JAK/STAT3 signaling axis.

背景:咖啡酸(CA)被认为是一种很有前景的植物化学物质,能抑制多种癌细胞增殖。因此,咖啡酸因其安全性和药理应用而受到越来越多的关注。在这项研究中,我们探讨了 CA 在抑制白细胞介素-6(IL-6)/Janus 激酶(JAK)/信号转导和激活剂转录-3(STAT-3)介导的人前列腺癌细胞增殖信号转导中的作用:采用 3-[4,5-二甲基噻唑-2-基]-2,5-二苯基溴化四唑(MTT)试验和集落形成试验研究了 CA 在增殖和集落形成能力中的作用。使用流式细胞仪技术确定肿瘤细胞死亡和细胞周期停滞。采用 Western 印迹法检测了 PC-3 和 LNCaP 细胞系中与 CA 处理相关的丝裂原活化蛋白激酶(MAPK)家族、IL-6/JAK/STAT-3 蛋白表达、增殖和凋亡蛋白表达:结果:我们发现,CA 能抑制前列腺癌细胞(PC-3 和 LNCaP)的增殖,并能诱导活性氧(ROS)、细胞周期停滞和细胞凋亡。此外,CA 还能减轻 PC-3 细胞中 MAPK 家族(即细胞外信号调节激酶 1(ERK1)、c-Jun N 端激酶(JNK)和 p38)磷酸化形式的表达。IL-6 介导的 JAK/STAT3 表达调控增殖和抗凋亡,从而导致前列腺癌转移和迁移。因此,减轻 IL-6/JAK/STAT-3 的表达被认为是治疗前列腺癌的一个重要靶点。在这项研究中,我们观察到 CA 可抑制 PC-3 和 LNCaP 细胞中 IL-6、JAK1 和磷酸化 STAT-3 的表达。由于IL-6/JAK/STAT-3的抑制作用,导致PC-3细胞中细胞周期蛋白-D1、细胞周期蛋白-D2和CDK1的表达减少。此外,CA还能通过增强前列腺癌细胞中Bax和caspase-3的表达以及降低Bcl-2的表达来诱导细胞凋亡:因此,CA 可通过靶向 IL-6/JAK/STAT3 信号轴来治疗前列腺癌。
{"title":"Caffeic acid hinders the proliferation and migration through inhibition of IL-6 mediated JAK-STAT-3 signaling axis in human prostate cancer.","authors":"Yuan Yin, Zhengyin Wang, Yujie Hu, Jia Wang, Y I Wang, Qun Lu","doi":"10.32604/or.2024.048007","DOIUrl":"https://doi.org/10.32604/or.2024.048007","url":null,"abstract":"<p><strong>Background: </strong>Caffeic acid (CA) is considered a promising phytochemical that has inhibited numerous cancer cell proliferation. Therefore, it is gaining increasing attention due to its safe and pharmacological applications. In this study, we investigated the role of CA in inhibiting the Interleukin-6 (IL-6)/Janus kinase (JAK)/Signal transducer and activator of transcription-3 (STAT-3) mediated suppression of the proliferation signaling in human prostate cancer cells.</p><p><strong>Materials and methods: </strong>The role of CA in proliferation and colony formation abilities was studied using 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) assay and colony formation assays. Tumour cell death and cell cycle arrest were identified using flow cytometry techniques. CA treatment-associated protein expression of mitogen-activated protein kinase (MAPK) families, IL-6/JAK/STAT-3, proliferation, and apoptosis protein expressions in PC-3 and LNCaP cell lines were measured using Western blot investigation.</p><p><strong>Results: </strong>We have obtained that treatment with CA inhibits prostate cancer cells (PC-3 and LNCaP) proliferation and induces reactive oxygen species (ROS), cell cycle arrest, and apoptosis cell death in a concentration-dependent manner. Moreover, CA treatment alleviates the expression phosphorylated form of MAPK families, i.e., extracellular signal-regulated kinase 1 (ERK1), c-Jun N-terminal kinase (JNK), and p38 in PC-3 cells. IL-6 mediated JAK/STAT3 expressions regulate the proliferation and antiapoptosis that leads to prostate cancer metastasis and migration. Therefore, to mitigate the expression of IL-6/JAK/STAT-3 is considered an important target for the treatment of prostate cancer. In this study, we have observed that CA inhibits the expression of IL-6, JAK1, and phosphorylated STAT-3 in both PC-3 and LNCaP cells. Due to the inhibitory effect of IL-6/JAK/STAT-3, it resulted in decreased expression of cyclin-D1, cyclin-D2, and CDK1 in both PC-3 cells. In addition, CA induces apoptosis by enhancing the expression of Bax and caspase-3; and decreased expression of Bcl-2 in prostate cancer cells.</p><p><strong>Conclusions: </strong>Thus, CA might act as a therapeutical application against prostate cancer by targeting the IL-6/JAK/STAT3 signaling axis.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"32 12","pages":"1881-1890"},"PeriodicalIF":2.0,"publicationDate":"2024-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11576972/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142687746","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Exploring the therapeutic potential of precision T-Cell Receptors (TCRs) in targeting KRAS G12D cancer through in vitro development. 通过体外开发探索精准 T 细胞受体 (TCR) 针对 KRAS G12D 癌症的治疗潜力。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-11-13 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.056565
Weitao Zheng, Dong Jiang, Songen Chen, Meiling Wu, Baoqi Yan, Jiahui Zhai, Yunqiang Shi, Bin Xie, Xingwang Xie, Kanghong Hu, Wenxue Ma

Objectives: The Kirsten rat sarcoma virus (KRAS) G12D oncogenic mutation poses a significant challenge in treating solid tumors due to the lack of specific and effective therapeutic interventions. This study aims to explore innovative approaches in T cell receptor (TCR) engineering and characterization to target the KRAS G12D7-16 mutation, providing potential strategies for overcoming this therapeutic challenge.

Methods: In this innovative study, we engineered and characterized two T cell receptors (TCRs), KDA11-01 and KDA11-02 with high affinity for the KRAS G12D7-16 mutation. These TCRs were isolated from tumor-infiltrating lymphocytes (TILs) derived from tumor tissues of patients with the KRAS G12D mutation. We assessed their specificity and anti-tumor activity in vitro using various cancer cell lines.

Results: KDA11-01 and KDA11-02 demonstrated exceptional specificity for the HLA-A*11:01-restricted KRAS G12D7-16 epitope, significantly inducing IFN-γ release and eliminating tumor cells without cross-reactivity or alloreactivity.

Conclusions: The successful development of KDA11-01 and KDA11-02 introduces a novel and precise TCR-based therapeutic strategy against KRAS G12D mutation, showing potential for significant advancements in cancer immunotherapy.

目的:柯氏大鼠肉瘤病毒(KRAS)G12D致癌突变是治疗实体瘤的重大挑战,因为缺乏特异性的有效治疗干预措施。本研究旨在探索针对 KRAS G12D7-16 突变的 T 细胞受体(TCR)工程和表征的创新方法,为克服这一治疗难题提供潜在策略:在这项创新性研究中,我们设计并鉴定了两种对 KRAS G12D7-16 突变具有高亲和力的 T 细胞受体 (TCR):KDA11-01 和 KDA11-02。这些TCR是从KRAS G12D突变患者的肿瘤组织中提取的肿瘤浸润淋巴细胞(TIL)中分离出来的。我们利用各种癌细胞系对它们的特异性和体外抗肿瘤活性进行了评估:结果:KDA11-01和KDA11-02对HLA-A*11:01限制的KRAS G12D7-16表位表现出了极高的特异性,能显著诱导IFN-γ的释放并清除肿瘤细胞,且无交叉反应或异体反应:KDA11-01和KDA11-02的成功开发为基于TCR的针对KRAS G12D突变的新型精准治疗策略提供了可能,有望在癌症免疫治疗领域取得重大进展。
{"title":"Exploring the therapeutic potential of precision T-Cell Receptors (TCRs) in targeting KRAS G12D cancer through <i>in vitro</i> development.","authors":"Weitao Zheng, Dong Jiang, Songen Chen, Meiling Wu, Baoqi Yan, Jiahui Zhai, Yunqiang Shi, Bin Xie, Xingwang Xie, Kanghong Hu, Wenxue Ma","doi":"10.32604/or.2024.056565","DOIUrl":"https://doi.org/10.32604/or.2024.056565","url":null,"abstract":"<p><strong>Objectives: </strong>The Kirsten rat sarcoma virus (KRAS) G12D oncogenic mutation poses a significant challenge in treating solid tumors due to the lack of specific and effective therapeutic interventions. This study aims to explore innovative approaches in T cell receptor (TCR) engineering and characterization to target the KRAS G12D<sub>7-16</sub> mutation, providing potential strategies for overcoming this therapeutic challenge.</p><p><strong>Methods: </strong>In this innovative study, we engineered and characterized two T cell receptors (TCRs), KDA11-01 and KDA11-02 with high affinity for the KRAS G12D<sub>7-16</sub> mutation. These TCRs were isolated from tumor-infiltrating lymphocytes (TILs) derived from tumor tissues of patients with the KRAS G12D mutation. We assessed their specificity and anti-tumor activity <i>in vitro</i> using various cancer cell lines.</p><p><strong>Results: </strong>KDA11-01 and KDA11-02 demonstrated exceptional specificity for the HLA-A*11:01-restricted KRAS G12D<sub>7-16</sub> epitope, significantly inducing IFN-γ release and eliminating tumor cells without cross-reactivity or alloreactivity.</p><p><strong>Conclusions: </strong>The successful development of KDA11-01 and KDA11-02 introduces a novel and precise TCR-based therapeutic strategy against KRAS G12D mutation, showing potential for significant advancements in cancer immunotherapy.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"32 12","pages":"1837-1850"},"PeriodicalIF":2.0,"publicationDate":"2024-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11576958/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142687777","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
Liver-directed therapies for fibrolamellar carcinoma: A single-center experience. 纤维母细胞癌的肝脏导向疗法:单中心经验。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-11-13 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.052985
Sam Son, Akshaar Brahmbhatt, Ken Zhao, Brett Marinelli, James Harding, William Jarnagin, Ghassan K Abou-Alfa, Hooman Yarmohammadi

Background: This article aims to present the single-institution outcomes of patients with Fibrolamellar Carcinoma (FLC) treated with liver-directed therapies (LDT).

Methods: In this single-center retrospective study, all patients diagnosed with FLC who underwent LDT were identified. Between July 2012 and July 2023, six patients were identified. One patient was excluded due to bleeding. Demographic and clinical parameters were recorded. Complications within 30 days of the LDT were evaluated. Radiological treatment responses at 1, 6, and 12 months were assessed per mRECIST.

Results: A total of five patients, which included three females and two males, were reviewed. Three patients were treated with transarterial hepatic embolization (TAE; n = 3), transarterial radioembolization (TARE; n = 1), and combined TAE + radiofrequency ablation (n = 1). The objective response rate at one month was 80% [CR = 2 (40%), PR = 2 (40%), and SD = 1 (20%)]. At 12 months (n = 4), two patients demonstrated CR (50%) and two demonstrated PR (50%). Overall survival from LDT at five years was 50%. There was no 30-day mortality among this group of patients or any adverse event attributable to the LDT.

Conclusion: TAE, TARE, and ablation are safe and effective therapeutic options for FLC. Based on this study and previously published case reports, ablation and TARE yielded the most favorable results.

背景:本文旨在介绍单个机构对纤维母细胞瘤(FLC)患者进行肝脏导向疗法(LDT)治疗的结果:在这项单中心回顾性研究中,确定了所有确诊为FLC并接受LDT治疗的患者。2012年7月至2023年7月期间,共确定了6名患者。其中一名患者因出血而被排除。记录了人口统计学和临床参数。评估了 LDT 术后 30 天内的并发症。根据 mRECIST 评估了 1、6 和 12 个月的放射治疗反应:共审查了五名患者,其中包括三名女性和两名男性。三名患者接受了经动脉肝栓塞(TAE;n = 3)、经动脉放射栓塞(TARE;n = 1)和TAE + 射频消融联合治疗(n = 1)。一个月的客观反应率为 80% [CR = 2 (40%)、PR = 2 (40%)、SD = 1 (20%)]。12 个月时(n = 4),两名患者表现为 CR(50%),两名表现为 PR(50%)。LDT五年总生存率为50%。这组患者中没有 30 天死亡病例,也没有发生任何可归因于 LDT 的不良事件:结论:TAE、TARE 和消融术是 FLC 安全有效的治疗方案。结论:TAE、TARE 和消融术是治疗 FLC 的安全有效的治疗方案。根据本研究和之前发表的病例报告,消融术和 TARE 取得了最理想的效果。
{"title":"Liver-directed therapies for fibrolamellar carcinoma: A single-center experience.","authors":"Sam Son, Akshaar Brahmbhatt, Ken Zhao, Brett Marinelli, James Harding, William Jarnagin, Ghassan K Abou-Alfa, Hooman Yarmohammadi","doi":"10.32604/or.2024.052985","DOIUrl":"https://doi.org/10.32604/or.2024.052985","url":null,"abstract":"<p><strong>Background: </strong>This article aims to present the single-institution outcomes of patients with Fibrolamellar Carcinoma (FLC) treated with liver-directed therapies (LDT).</p><p><strong>Methods: </strong>In this single-center retrospective study, all patients diagnosed with FLC who underwent LDT were identified. Between July 2012 and July 2023, six patients were identified. One patient was excluded due to bleeding. Demographic and clinical parameters were recorded. Complications within 30 days of the LDT were evaluated. Radiological treatment responses at 1, 6, and 12 months were assessed per mRECIST.</p><p><strong>Results: </strong>A total of five patients, which included three females and two males, were reviewed. Three patients were treated with transarterial hepatic embolization (TAE; n = 3), transarterial radioembolization (TARE; n = 1), and combined TAE + radiofrequency ablation (n = 1). The objective response rate at one month was 80% [CR = 2 (40%), PR = 2 (40%), and SD = 1 (20%)]. At 12 months (n = 4), two patients demonstrated CR (50%) and two demonstrated PR (50%). Overall survival from LDT at five years was 50%. There was no 30-day mortality among this group of patients or any adverse event attributable to the LDT.</p><p><strong>Conclusion: </strong>TAE, TARE, and ablation are safe and effective therapeutic options for FLC. Based on this study and previously published case reports, ablation and TARE yielded the most favorable results.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"32 12","pages":"1831-1836"},"PeriodicalIF":2.0,"publicationDate":"2024-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11576953/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142687852","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"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
hsa-miR-181a-5p inhibits glioblastoma development via the MAPK pathway: in-silico and in-vitro study. hsa-miR-181a-5p通过MAPK通路抑制胶质母细胞瘤的发展:体内和体外研究
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-11-13 eCollection Date: 2024-01-01 DOI: 10.32604/or.2024.051569
Mahdi Abdoli Shadbad, Behzad Baradaran

Background: Glioblastoma remains a highly invasive primary brain malignancy with an undesirable prognosis. Growing evidence has shed light on the importance of microRNAs (miRs), as small non-coding RNAs, in tumor development and progression. The present study leverages the in-silico and in-vitro techniques to investigate the significance of hsa-miR-181a-5p and the underlying hsa-miR-181a-5p-meidated signaling pathway in glioblastoma development.

Methods: Bioinformatic studies were performed on GSE158284, GSE108474 (REMBRANDT study), TCGA-GTEx, CCLE, GeneMANIA, Reactome, WikiPathways, KEGG, miRDB, and microT-CDS to identify the significance of hsa-miR-181a-5p and its underlying target. Afterward, the U373 cell line was selected and transfected with hsa-miR-181a-5p mimics, and the cell viability, clonogenicity, migration, mRNA expression, apoptosis, and cell cycle were studied using the MTT assay, colony formation test, migration assay, qRT-PCR, and flow cytometry respectively.

Results: hsa-miR-181a-5p expression is decreased in glioblastoma samples. The in-silico results have shown that hsa-miR-181a-5p could regulate the MAPK pathway by targeting AKT3. The experimental assays have shown that hsa-miR-181a-5p decreases the migration of glioblastoma cells, arrests the cell cycle, and increases the apoptosis rate. Besides downregulating MMP9 and upregulating BAX, hsa-miR-181a-5p downregulates MET, MAP2K1, MAPK1, MAPK3, and AKT3 expression in U373 cells. The in-vitro results were consistent with in-silico results regarding the regulatory effect of hsa-miR-181a-5p on the MAPK pathway, leading to tumor suppression in glioblastoma.

Conclusions: hsa-miR-181a-5p inhibits glioblastoma development partially by regulating the signaling factors of the MAPK pathway.

背景:胶质母细胞瘤是一种侵袭性很强的原发性脑恶性肿瘤,预后不良。越来越多的证据表明,作为小型非编码 RNA 的微 RNA(miRs)在肿瘤发生和发展中的重要性。本研究利用体内和体外技术研究了hsa-miR-181a-5p和hsa-miR-181a-5p-meidated信号通路在胶质母细胞瘤发展中的意义:对GSE158284、GSE108474(REMBRANDT研究)、TCGA-GTEx、CCLE、GeneMANIA、Reactome、WikiPathways、KEGG、miRDB和microT-CDS进行生物信息学研究,以确定hsa-miR-181a-5p及其潜在靶点的意义。结果显示:hsa-miR-181a-5p在胶质母细胞瘤样本中的表达下降。结果表明:hsa-miR-181a-5p 在胶质母细胞瘤样本中的表达量下降,体内研究结果表明,hsa-miR-181a-5p 可通过靶向 AKT3 调节 MAPK 通路。实验结果表明,hsa-miR-181a-5p 能降低胶质母细胞瘤细胞的迁移、阻滞细胞周期并提高细胞凋亡率。除了下调 MMP9 和上调 BAX 外,hsa-miR-181a-5p 还能下调 U373 细胞中 MET、MAP2K1、MAPK1、MAPK3 和 AKT3 的表达。体外实验结果与体内实验结果一致,即 hsa-miR-181a-5p 对 MAPK 通路具有调控作用,从而抑制胶质母细胞瘤的发展。
{"title":"hsa-miR-181a-5p inhibits glioblastoma development via the MAPK pathway: <i>in-silico</i> and <i>in-vitro</i> study.","authors":"Mahdi Abdoli Shadbad, Behzad Baradaran","doi":"10.32604/or.2024.051569","DOIUrl":"https://doi.org/10.32604/or.2024.051569","url":null,"abstract":"<p><strong>Background: </strong>Glioblastoma remains a highly invasive primary brain malignancy with an undesirable prognosis. Growing evidence has shed light on the importance of microRNAs (miRs), as small non-coding RNAs, in tumor development and progression. The present study leverages the <i>in-silico</i> and <i>in-vitro</i> techniques to investigate the significance of hsa-miR-181a-5p and the underlying hsa-miR-181a-5p-meidated signaling pathway in glioblastoma development.</p><p><strong>Methods: </strong>Bioinformatic studies were performed on GSE158284, GSE108474 (REMBRANDT study), TCGA-GTEx, CCLE, GeneMANIA, Reactome, WikiPathways, KEGG, miRDB, and microT-CDS to identify the significance of hsa-miR-181a-5p and its underlying target. Afterward, the U373 cell line was selected and transfected with hsa-miR-181a-5p mimics, and the cell viability, clonogenicity, migration, mRNA expression, apoptosis, and cell cycle were studied using the MTT assay, colony formation test, migration assay, qRT-PCR, and flow cytometry respectively.</p><p><strong>Results: </strong>hsa-miR-181a-5p expression is decreased in glioblastoma samples. The <i>in-silico</i> results have shown that hsa-miR-181a-5p could regulate the MAPK pathway by targeting <i>AKT3</i>. The experimental assays have shown that hsa-miR-181a-5p decreases the migration of glioblastoma cells, arrests the cell cycle, and increases the apoptosis rate. Besides downregulating <i>MMP9</i> and upregulating <i>BAX</i>, hsa-miR-181a-5p downregulates <i>MET</i>, <i>MAP2K1</i>, <i>MAPK1</i>, <i>MAPK3</i>, and <i>AKT3</i> expression in U373 cells. The <i>in-vitro</i> results were consistent with <i>in-silico</i> results regarding the regulatory effect of hsa-miR-181a-5p on the MAPK pathway, leading to tumor suppression in glioblastoma.</p><p><strong>Conclusions: </strong>hsa-miR-181a-5p inhibits glioblastoma development partially by regulating the signaling factors of the MAPK pathway.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"32 12","pages":"1949-1958"},"PeriodicalIF":2.0,"publicationDate":"2024-11-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11576920/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142687841","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Oncology Research
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1