首页 > 最新文献

Translational cancer research最新文献

英文 中文
Construction and validation of prognostic model for colorectal mucinous adenocarcinoma patients and identification of a new prognosis related gene FAM174B. 构建并验证结直肠粘液腺癌患者的预后模型,发现新的预后相关基因FAM174B
IF 1.5 4区 医学 Q4 ONCOLOGY Pub Date : 2024-10-31 Epub Date: 2024-10-29 DOI: 10.21037/tcr-24-347
Xiangwen Tan, Qing Fang, Yunhua Xu, Shuxiang Li, Jinyi Yuan, Kunming Xu, Xiguang Chen, Guang Fu, Yarui Liu, Qiulin Huang, Xiuda Peng, Shuai Xiao

Background: Mucinous adenocarcinoma (MAC) is a peculiar histological subtype of colorectal cancer (CRC) with distinct medical, disease-related, and genetic characteristics. The prognosis of MAC is generally poorer less favorable compared to non-specific adenocarcinoma (AC), but the prognostic indicator of MAC is rare. Therefore, this study aims to identify potential biomarkers and construct a prognostic model to better predict patient outcomes in MAC.

Methods: We conducted differential genes expression investigation, weighted gene co-expression network analysis (WGCNA), and least absolute shrinkage and selection operator (LASSO)-Cox regression model using RNA sequencing (RNA-seq) data from The Cancer Genome Atlas (TCGA) to pinpoint hub genes. Then, the hub genes were used to construct a prognostic model for MAC. Kaplan-Meier survival, receiver operating characteristic (ROC), and Cox regression analysis were used to assess the prognostic utility of the model. The potential biological function of the hub gene was examined using gene set enrichment analysis (GSEA).

Results: Four hub genes, FAM174B, CREB3L1, SPDEF, and RAP1GAP, were identified between MAC and AC by differential genes expression analysis, WGCNA, and LASSO regression analysis. The prognostic signature model was constructed based on these four hub genes, which could divide MAC into low- and high-risk groups. The overall survival (OS) was notably lower in the high-risk group compared to the low-risk group (P=0.007). The area under the curves (AUCs) for 1-, 3-, and 5-year OS were 0.61 [95% confidence interval (CI): 0.73-0.49], 0.69 (95% CI: 0.76-0.63), and 0.77 (95% CI: 0.83-0.71), respectively. We also found that FAM174B expression was closely related to the OS of MAC (P=0.02). Further, the expression of FAM174B was positively correlated with MAC's Mucin type O-glycan biosynthesis. Finally, it was indicated that FAM174B was positively correlated with the critical molecules of mucus formation, MUC5AC (P=0.004, r=0.33), MUC5B (P<0.001, r=0.43), and MUC2 (P<0.001, r=0.39).

Conclusions: We have developed and validated a four-gene prognostic model to predict the survival of MAC. Additionally, we found that FAM174B might correlate with mucin production in MAC.

背景:黏液腺癌(MAC)是结直肠癌(CRC)的一种特殊组织学亚型,具有独特的医学、疾病相关和遗传学特征。与非特异性腺癌(AC)相比,MAC 的预后一般较差,但 MAC 的预后指标却很少见。因此,本研究旨在确定潜在的生物标志物并构建预后模型,以更好地预测 MAC 患者的预后:方法:我们利用癌症基因组图谱(TCGA)中的RNA测序(RNA-seq)数据进行了差异基因表达调查、加权基因共表达网络分析(WGCNA)和最小绝对收缩与选择算子(LASSO)-Cox回归模型,以确定枢纽基因。然后,利用这些中心基因构建了MAC的预后模型。卡普兰-梅耶生存率、接收器操作特征(ROC)和考克斯回归分析被用来评估该模型的预后效用。利用基因组富集分析(GSEA)检验了枢纽基因的潜在生物学功能:结果:通过差异基因表达分析、WGCNA和LASSO回归分析,确定了MAC和AC之间的四个枢纽基因,即FAM174B、CREB3L1、SPDEF和RAP1GAP。根据这四个枢纽基因构建的预后特征模型可将 MAC 分成低危和高危两组。与低风险组相比,高风险组的总生存期(OS)明显较低(P=0.007)。1年、3年和5年OS的曲线下面积(AUC)分别为0.61[95%置信区间(CI):0.73-0.49]、0.69(95% CI:0.76-0.63)和0.77(95% CI:0.83-0.71)。我们还发现,FAM174B的表达与MAC的OS密切相关(P=0.02)。此外,FAM174B的表达与MAC的粘蛋白型O-糖生物合成呈正相关。最后,研究表明 FAM174B 与粘液形成的关键分子 MUC5AC(P=0.004,r=0.33)、MUC5B(PMUC2)(PConclusions)呈正相关:我们开发并验证了预测 MAC 存活率的四基因预后模型。此外,我们还发现 FAM174B 可能与 MAC 中粘蛋白的产生有关。
{"title":"Construction and validation of prognostic model for colorectal mucinous adenocarcinoma patients and identification of a new prognosis related gene <i>FAM174B</i>.","authors":"Xiangwen Tan, Qing Fang, Yunhua Xu, Shuxiang Li, Jinyi Yuan, Kunming Xu, Xiguang Chen, Guang Fu, Yarui Liu, Qiulin Huang, Xiuda Peng, Shuai Xiao","doi":"10.21037/tcr-24-347","DOIUrl":"https://doi.org/10.21037/tcr-24-347","url":null,"abstract":"<p><strong>Background: </strong>Mucinous adenocarcinoma (MAC) is a peculiar histological subtype of colorectal cancer (CRC) with distinct medical, disease-related, and genetic characteristics. The prognosis of MAC is generally poorer less favorable compared to non-specific adenocarcinoma (AC), but the prognostic indicator of MAC is rare. Therefore, this study aims to identify potential biomarkers and construct a prognostic model to better predict patient outcomes in MAC.</p><p><strong>Methods: </strong>We conducted differential genes expression investigation, weighted gene co-expression network analysis (WGCNA), and least absolute shrinkage and selection operator (LASSO)-Cox regression model using RNA sequencing (RNA-seq) data from The Cancer Genome Atlas (TCGA) to pinpoint hub genes. Then, the hub genes were used to construct a prognostic model for MAC. Kaplan-Meier survival, receiver operating characteristic (ROC), and Cox regression analysis were used to assess the prognostic utility of the model. The potential biological function of the hub gene was examined using gene set enrichment analysis (GSEA).</p><p><strong>Results: </strong>Four hub genes, <i>FAM174B</i>, <i>CREB3L1</i>, <i>SPDEF</i>, and <i>RAP1GAP</i>, were identified between MAC and AC by differential genes expression analysis, WGCNA, and LASSO regression analysis. The prognostic signature model was constructed based on these four hub genes, which could divide MAC into low- and high-risk groups. The overall survival (OS) was notably lower in the high-risk group compared to the low-risk group (P=0.007). The area under the curves (AUCs) for 1-, 3-, and 5-year OS were 0.61 [95% confidence interval (CI): 0.73-0.49], 0.69 (95% CI: 0.76-0.63), and 0.77 (95% CI: 0.83-0.71), respectively. We also found that <i>FAM174B</i> expression was closely related to the OS of MAC (P=0.02). Further, the expression of <i>FAM174B</i> was positively correlated with MAC's Mucin type O-glycan biosynthesis. Finally, it was indicated that <i>FAM174B</i> was positively correlated with the critical molecules of mucus formation, <i>MUC5AC</i> (P=0.004, r=0.33), <i>MUC5B</i> (P<0.001, r=0.43), and <i>MUC2</i> (P<0.001, r=0.39).</p><p><strong>Conclusions: </strong>We have developed and validated a four-gene prognostic model to predict the survival of MAC. Additionally, we found that <i>FAM174B</i> might correlate with mucin production in MAC.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 10","pages":"5233-5246"},"PeriodicalIF":1.5,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11543054/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142626539","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
Exosomal AHSG in ovarian cancer ascites inhibits malignant progression of ovarian cancer by p53/FAK/Src signaling. 卵巢癌腹水外泌体AHSG通过p53/FAK/Src信号传导抑制卵巢癌的恶性进展
IF 1.5 4区 医学 Q4 ONCOLOGY Pub Date : 2024-10-31 Epub Date: 2024-10-29 DOI: 10.21037/tcr-24-789
Guangyan Xie, Yongli Zhang, Jiachen Ma, Xiaoli Guo, Jiahao Xu, Linna Chen, Jingbo Zhang, Yanyu Li, Bei Zhang, Xueyan Zhou

Background: The primary cause of mortality in patients with ovarian cancer (OC) is tumor metastasis. A comprehensive understanding of the mechanisms underlying metastasis in OC is essential for accurate prognosis prediction and the development of targeted therapeutic agents. Our findings indicate that alpha-2 Heremans Schmid glycoprotein (AHSG) is downregulated in OC exosomes. Consequently, the objective of this study was to identify novel prognostic markers and potential therapeutic targets for OC.

Methods: Exosomes derived from OC cells and patient ascites were purified and applied to OC cells to assess their migratory ability using wound-healing and transwell assays. AHSG expression was enhanced by overexpressing lentivirus, and the resulting exosomes were isolated and co-cultured with OC cells to verify their effect on the migration ability of OC.

Results: Exosomes in ovarian malignant ascites have been demonstrated to promote OC metastasis. However, our findings indicate that AHSG is down-regulated in OC tissues and ascites exosomes. Furthermore, overexpression of AHSG in OC cells has been shown to markedly decrease their migratory ability, as well as reduce the migratory ability of cancer cells after co-culture of its exosomes with cancer cells.

Conclusions: The low expression of AHSG in exosomes derived from OC tissues and ascites is associated with metastatic progression in OC patients. Additionally, cancer-derived AHSG can be transported to OC cells via exosomes, where it inhibits OC migration in vitro and in vivo by regulating the p53/FAK/Src signaling pathway. The present study demonstrated that AHSG, derived from cancer cells, exerts a negative regulatory effect on OC cell motility, migration, and metastasis. These findings suggest that AHSG is a potential candidate for OC treatment.

背景:卵巢癌(OC)患者死亡的主要原因是肿瘤转移。全面了解卵巢癌转移的内在机制对于准确预测预后和开发靶向治疗药物至关重要。我们的研究结果表明,α-2 Heremans Schmid glycoprotein(AHSG)在OC外泌体中下调。因此,本研究的目的是确定OC的新型预后标志物和潜在治疗靶点:方法:纯化从OC细胞和患者腹水中提取的外泌体,并将其应用于OC细胞,使用伤口愈合和透孔试验评估其迁移能力。通过过表达慢病毒增强AHSG的表达,分离得到的外泌体并与OC细胞共培养,验证其对OC迁移能力的影响:结果:卵巢恶性腹水中的外泌体已被证实能促进OC转移。然而,我们的研究结果表明,AHSG在OC组织和腹水外泌体中被下调。此外,在OC细胞中过表达AHSG可明显降低其迁移能力,其外泌体与癌细胞共培养后也可降低癌细胞的迁移能力:结论:从OC组织和腹水中提取的外泌体中AHSG的低表达与OC患者的转移进展有关。此外,癌症衍生的AHSG可通过外泌体转运至OC细胞,并通过调节p53/FAK/Src信号通路抑制体外和体内的OC迁移。本研究表明,从癌细胞中提取的AHSG对OC细胞的运动、迁移和转移具有负向调节作用。这些发现表明,AHSG 是治疗 OC 的潜在候选药物。
{"title":"Exosomal AHSG in ovarian cancer ascites inhibits malignant progression of ovarian cancer by p53/FAK/Src signaling.","authors":"Guangyan Xie, Yongli Zhang, Jiachen Ma, Xiaoli Guo, Jiahao Xu, Linna Chen, Jingbo Zhang, Yanyu Li, Bei Zhang, Xueyan Zhou","doi":"10.21037/tcr-24-789","DOIUrl":"https://doi.org/10.21037/tcr-24-789","url":null,"abstract":"<p><strong>Background: </strong>The primary cause of mortality in patients with ovarian cancer (OC) is tumor metastasis. A comprehensive understanding of the mechanisms underlying metastasis in OC is essential for accurate prognosis prediction and the development of targeted therapeutic agents. Our findings indicate that alpha-2 Heremans Schmid glycoprotein (AHSG) is downregulated in OC exosomes. Consequently, the objective of this study was to identify novel prognostic markers and potential therapeutic targets for OC.</p><p><strong>Methods: </strong>Exosomes derived from OC cells and patient ascites were purified and applied to OC cells to assess their migratory ability using wound-healing and transwell assays. AHSG expression was enhanced by overexpressing lentivirus, and the resulting exosomes were isolated and co-cultured with OC cells to verify their effect on the migration ability of OC.</p><p><strong>Results: </strong>Exosomes in ovarian malignant ascites have been demonstrated to promote OC metastasis. However, our findings indicate that AHSG is down-regulated in OC tissues and ascites exosomes. Furthermore, overexpression of AHSG in OC cells has been shown to markedly decrease their migratory ability, as well as reduce the migratory ability of cancer cells after co-culture of its exosomes with cancer cells.</p><p><strong>Conclusions: </strong>The low expression of AHSG in exosomes derived from OC tissues and ascites is associated with metastatic progression in OC patients. Additionally, cancer-derived AHSG can be transported to OC cells via exosomes, where it inhibits OC migration <i>in vitro</i> and <i>in vivo</i> by regulating the p53/FAK/Src signaling pathway. The present study demonstrated that AHSG, derived from cancer cells, exerts a negative regulatory effect on OC cell motility, migration, and metastasis. These findings suggest that AHSG is a potential candidate for OC treatment.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 10","pages":"5365-5380"},"PeriodicalIF":1.5,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11543047/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142627325","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
Establishment and validation of a prediction model for gastric cancer with perineural invasion based on preoperative inflammatory markers. 基于术前炎症标志物的胃癌会厌浸润预测模型的建立与验证
IF 1.5 4区 医学 Q4 ONCOLOGY Pub Date : 2024-10-31 Epub Date: 2024-10-12 DOI: 10.21037/tcr-24-481
Pan Jiang, Lijun Zheng, Yining Yang, Dongping Mo

Background: Gastric cancer (GC) is a prevalent malignant tumor of the digestive system, characterized by a poor prognosis and high recurrence rate. Perineural invasion (PNI), the neoplastic infiltration of nerves, is a significant predictor of survival outcome in GC. Accurate preoperative identification of PNI could facilitate patient stratification and optimal preoperative treatment. We therefore established and validated a preoperative risk assessment model for GC patients with PNI.

Methods: We collected data from 1,195 patients who underwent surgical resection at our hospital between October 2020 and December 2023, with PNI confirmed by pathological examination. We gathered laboratory data, including blood cell count, blood type, coagulation index, biochemical indexes, and tumor markers. Eligible patients were randomly divided into a training set and a testing set at a ratio of 7:3. The important risk factors of PNI were evaluated by random forest package in RStudio. Receiver operating characteristic-area under the curve (ROC-AUC) analysis was used to evaluate the discriminatory ability of the factors for PNI. Univariate and multivariate logistic regression analyses were utilized to verity independent risk factors for patients with PNI, and the logistic regression model and nomogram were constructed based on the results. Calibration curve and decision curve analysis (DCA) were conducted to assess the predictive model. Finally, we verified the prediction equation model using the testing set.

Results: In the training set, 416 GC patients were pathologically diagnosed with PNI. The top 5 important risk factors for PNI were identified as carcinoembryonic antigen (CEA), fibrinogen-to-lymphocyte ratio (FLR), D-dimer, platelet-to-lymphocyte ratio (PLR), and carbohydrate antigen 19-9 (CA19-9), with optimal cut-off values of 3.89 ng/mL, 2.08, 0.24 mg/L, 122.37, and 14.85 U/mL, respectively. Multivariate logistic regression analysis confirmed that CEA, FLR, D-dimer, PLR, CA19-9, and CA72-4 as independent risk factors for PNI (P<0.05). We formulated the following predictive equation: Logit(P) = -1.211 + 0.695 × CEA + 0.546 × FLR + 0.686 × D-dimer + 0.653 × PLR + 0.515 × CA19-9 + 0.518 × CA72-4 (χ2=105.675, P<0.001). The model demonstrated an ROC-AUC value of 0.719 [95% confidence interval (CI): 0.681-0.757] in the training set, with a sensitivity of 68.51% and a specificity of 67.60%. The ROC-AUC value was 0.791 (95% CI: 0.750-0.831) in the testing set (sensitivity: 69.57%, specificity: 56.41%). Calibration curve and DCA confirmed that the model has good discrimination and accuracy.

Conclusions: We successfully established and validated a prediction model for GC patients with PNI based on hematological indicators, hoping that this model can provide an adjunctive tool for predicting PNI in clinical work.

背景:胃癌(GC)是一种常见的消化系统恶性肿瘤,其特点是预后差、复发率高。神经周围侵犯(PNI)是指肿瘤对神经的浸润,是预测胃癌患者生存结果的一个重要指标。术前准确识别 PNI 有助于对患者进行分层和优化术前治疗。因此,我们建立并验证了针对有 PNI 的 GC 患者的术前风险评估模型:我们收集了2020年10月至2023年12月期间在我院接受手术切除并经病理检查证实有PNI的1195名患者的数据。我们收集了实验室数据,包括血细胞计数、血型、凝血指数、生化指标和肿瘤标志物。符合条件的患者按 7:3 的比例随机分为训练集和测试集。使用 RStudio 中的随机森林软件包评估 PNI 的重要风险因素。受试者操作特征曲线下面积(ROC-AUC)分析用于评估这些因素对 PNI 的判别能力。利用单变量和多变量逻辑回归分析来验证 PNI 患者的独立风险因素,并根据结果构建逻辑回归模型和提名图。校准曲线和决策曲线分析(DCA)用于评估预测模型。最后,我们利用测试集验证了预测方程模型:在训练集中,416 名 GC 患者被病理诊断为 PNI。结果:在训练集中,416 名 GC 患者被病理诊断为 PNI,其中前 5 个重要的 PNI 风险因素分别为癌胚抗原(CEA)、纤维蛋白原与淋巴细胞比值(FLR)、D-二聚体、血小板与淋巴细胞比值(PLR)和碳水化合物抗原 19-9(CA19-9),最佳临界值分别为 3.89 ng/mL、2.08、0.24 mg/L、122.37 和 14.85 U/mL。多变量逻辑回归分析证实,CEA、FLR、D-二聚体、PLR、CA19-9 和 CA72-4 是 PNI 的独立危险因素(P2=105.675,PConclusions:我们成功建立并验证了基于血液学指标的GC患者PNI预测模型,希望该模型能为临床工作中预测PNI提供辅助工具。
{"title":"Establishment and validation of a prediction model for gastric cancer with perineural invasion based on preoperative inflammatory markers.","authors":"Pan Jiang, Lijun Zheng, Yining Yang, Dongping Mo","doi":"10.21037/tcr-24-481","DOIUrl":"https://doi.org/10.21037/tcr-24-481","url":null,"abstract":"<p><strong>Background: </strong>Gastric cancer (GC) is a prevalent malignant tumor of the digestive system, characterized by a poor prognosis and high recurrence rate. Perineural invasion (PNI), the neoplastic infiltration of nerves, is a significant predictor of survival outcome in GC. Accurate preoperative identification of PNI could facilitate patient stratification and optimal preoperative treatment. We therefore established and validated a preoperative risk assessment model for GC patients with PNI.</p><p><strong>Methods: </strong>We collected data from 1,195 patients who underwent surgical resection at our hospital between October 2020 and December 2023, with PNI confirmed by pathological examination. We gathered laboratory data, including blood cell count, blood type, coagulation index, biochemical indexes, and tumor markers. Eligible patients were randomly divided into a training set and a testing set at a ratio of 7:3. The important risk factors of PNI were evaluated by random forest package in RStudio. Receiver operating characteristic-area under the curve (ROC-AUC) analysis was used to evaluate the discriminatory ability of the factors for PNI. Univariate and multivariate logistic regression analyses were utilized to verity independent risk factors for patients with PNI, and the logistic regression model and nomogram were constructed based on the results. Calibration curve and decision curve analysis (DCA) were conducted to assess the predictive model. Finally, we verified the prediction equation model using the testing set.</p><p><strong>Results: </strong>In the training set, 416 GC patients were pathologically diagnosed with PNI. The top 5 important risk factors for PNI were identified as carcinoembryonic antigen (CEA), fibrinogen-to-lymphocyte ratio (FLR), D-dimer, platelet-to-lymphocyte ratio (PLR), and carbohydrate antigen 19-9 (CA19-9), with optimal cut-off values of 3.89 ng/mL, 2.08, 0.24 mg/L, 122.37, and 14.85 U/mL, respectively. Multivariate logistic regression analysis confirmed that CEA, FLR, D-dimer, PLR, CA19-9, and CA72-4 as independent risk factors for PNI (P<0.05). We formulated the following predictive equation: Logit(P) = -1.211 + 0.695 × CEA + 0.546 × FLR + 0.686 × D-dimer + 0.653 × PLR + 0.515 × CA19-9 + 0.518 × CA72-4 (χ<sup>2</sup>=105.675, P<0.001). The model demonstrated an ROC-AUC value of 0.719 [95% confidence interval (CI): 0.681-0.757] in the training set, with a sensitivity of 68.51% and a specificity of 67.60%. The ROC-AUC value was 0.791 (95% CI: 0.750-0.831) in the testing set (sensitivity: 69.57%, specificity: 56.41%). Calibration curve and DCA confirmed that the model has good discrimination and accuracy.</p><p><strong>Conclusions: </strong>We successfully established and validated a prediction model for GC patients with PNI based on hematological indicators, hoping that this model can provide an adjunctive tool for predicting PNI in clinical work.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 10","pages":"5381-5394"},"PeriodicalIF":1.5,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11543023/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142626825","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 and validation of a novel innate lymphoid cell-based signature to predict prognosis and immune response in liver cancer by integrated single-cell RNA analysis and bulk RNA sequencing. 通过整合单细胞 RNA 分析和大量 RNA 测序,鉴定和验证基于先天淋巴细胞的新型特征,以预测肝癌的预后和免疫反应。
IF 1.5 4区 医学 Q4 ONCOLOGY Pub Date : 2024-10-31 Epub Date: 2024-10-28 DOI: 10.21037/tcr-24-725
Meng Pan, Xiaolong Yuan, Junlu Peng, Ruiqi Wu, Xiaopeng Chen
<p><strong>Background: </strong>Innate lymphoid cells (ILCs) exert tumor suppressive and tumor promoting effects. However, the prognostic significance of ILC-associated genes remains unclear in hepatocellular carcinoma (HCC). Hence, the aim of this research was to develop an innovative predictive risk classification system using bioinformatics examination.</p><p><strong>Methods: </strong>We explored the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases to gather data pertaining to HCC and its clinical details. Significantly different ILC-associated genes were investigated by Seurat analysis. The number of signaling interactions of ILCs with other cells was discovered by CellPhoneDB analysis. ClusterProfiler and Metascape were utilized to perform Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses on ILC genes. In order to identify potential ILC predictors, we utilized univariate Cox regression and least absolute shrinkage and selection operator (LASSO) analyses, subsequently validating these predictors in TCGA and GEO groups. The multi-omics ILC signature model's clinical predictive capabilities, along with drug sensitivity and immune factor relations, were assessed using Cell-type Identification by Estimating Relative Subsets of RNA Transcripts (CIBERSORT) and pRRophetic. We investigated the possible molecular pathways in our predictive ILC signature through the utilization of gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA). Five key genes were screened out by constructing a competing endogenous RNA (ceRNA) network using Cytoscape and their values in clinical indexes were demonstrated. Immunohistochemistry (IHC) in HCC cases confirmed the expression of these genes.</p><p><strong>Results: </strong>ILC cell subsets were identified, and cell-cell communication analysis revealed that the signaling pathways involving ILC cell subsets were mostly abundant in the HCC microenvironment. Subsequently, 270 marker genes of the ILC clusters were subjected to GO and KEGG enrichment analysis. Furthermore, a total of 58 prognostically relevant genes were screened as features for prognostic prediction models. Next, the models were validated and clinically evaluated (P values of Kaplan-Meier survival curves below 0.05). Five key genes (<i>C2, STK4, CALM1, IL7R</i>, and <i>RORA</i>) were further screened by multi omics analysis of immune cell and factor and drug sensitivity and correlation analysis of tumor regulatory genes in liver cancer. Furthermore, the potential clinical value of the 5 key genes was confirmed in HCC patients. Finally, the IHC results confirmed the expression of <i>C2</i>, <i>STK4</i>, <i>CALM1</i>, <i>IL7R</i>, and <i>RORA</i> in HCC. Our experimental results provided preliminary evidence supporting the oncogenic roles of <i>STK4</i> and <i>CALM1</i>, as well as the tumor-suppressive roles of <i>C2</i>, <i>RORA</i>, and <i>IL7R</i> in HCC.</p><p><strong>
背景:先天性淋巴细胞(ILC)具有抑制肿瘤和促进肿瘤生长的作用。然而,ILC相关基因在肝细胞癌(HCC)中的预后意义仍不明确。因此,本研究的目的是利用生物信息学检查开发一种创新的预测风险分类系统:我们探索了基因表达总库(GEO)和癌症基因组图谱(TCGA)数据库,以收集有关 HCC 及其临床细节的数据。通过Seurat分析研究了与ILC相关的显著不同基因。通过CellPhoneDB分析发现了ILC与其他细胞之间的信号交互数量。利用ClusterProfiler和Metascape对ILC基因进行了基因本体(GO)和京都基因组百科全书(KEGG)富集分析。为了确定潜在的ILC预测因子,我们使用了单变量考克斯回归和最小绝对收缩与选择算子(LASSO)分析,随后在TCGA和GEO组中验证了这些预测因子。我们利用RNA转录本相对子集估算(CIBERSORT)和pRRophetic评估了多组学ILC特征模型的临床预测能力以及药物敏感性和免疫因子关系。我们利用基因组富集分析(GSEA)和基因组变异分析(GSVA)研究了预测性 ILC 特征中可能的分子通路。通过使用Cytoscape构建竞争性内源性RNA(ceRNA)网络,筛选出了五个关键基因,并展示了它们在临床指标中的价值。HCC病例中的免疫组织化学(IHC)证实了这些基因的表达:结果:确定了 ILC 细胞亚群,细胞-细胞通讯分析表明,涉及 ILC 细胞亚群的信号通路在 HCC 微环境中最为丰富。随后,对 ILC 群的 270 个标记基因进行了 GO 和 KEGG 富集分析。此外,共筛选出 58 个与预后相关的基因作为预后预测模型的特征。接下来,对模型进行了验证和临床评估(Kaplan-Meier 生存曲线的 P 值低于 0.05)。通过对肝癌免疫细胞和因子、药物敏感性和肿瘤调控基因的相关性分析,进一步筛选了五个关键基因(C2、STK4、CALM1、IL7R和RORA)。此外,还证实了这 5 个关键基因在 HCC 患者中的潜在临床价值。最后,IHC 结果证实了 C2、STK4、CALM1、IL7R 和 RORA 在 HCC 中的表达。我们的实验结果为 STK4 和 CALM1 的致癌作用以及 C2、RORA 和 IL7R 在 HCC 中的抑瘤作用提供了初步证据:结论:发现了一种可能与 HCC 有关的 ILC 新预后特征。结论:发现了一种可能参与 HCC 的 ILC 新预后特征,它在预测患者总生存期(OS)方面显示出很高的价值,在免疫和药物敏感性方面也有很好的差异。因此,针对这些 ILC 特征可能是治疗 HCC 的一种潜在有效方法。
{"title":"Identification and validation of a novel innate lymphoid cell-based signature to predict prognosis and immune response in liver cancer by integrated single-cell RNA analysis and bulk RNA sequencing.","authors":"Meng Pan, Xiaolong Yuan, Junlu Peng, Ruiqi Wu, Xiaopeng Chen","doi":"10.21037/tcr-24-725","DOIUrl":"https://doi.org/10.21037/tcr-24-725","url":null,"abstract":"&lt;p&gt;&lt;strong&gt;Background: &lt;/strong&gt;Innate lymphoid cells (ILCs) exert tumor suppressive and tumor promoting effects. However, the prognostic significance of ILC-associated genes remains unclear in hepatocellular carcinoma (HCC). Hence, the aim of this research was to develop an innovative predictive risk classification system using bioinformatics examination.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Methods: &lt;/strong&gt;We explored the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases to gather data pertaining to HCC and its clinical details. Significantly different ILC-associated genes were investigated by Seurat analysis. The number of signaling interactions of ILCs with other cells was discovered by CellPhoneDB analysis. ClusterProfiler and Metascape were utilized to perform Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses on ILC genes. In order to identify potential ILC predictors, we utilized univariate Cox regression and least absolute shrinkage and selection operator (LASSO) analyses, subsequently validating these predictors in TCGA and GEO groups. The multi-omics ILC signature model's clinical predictive capabilities, along with drug sensitivity and immune factor relations, were assessed using Cell-type Identification by Estimating Relative Subsets of RNA Transcripts (CIBERSORT) and pRRophetic. We investigated the possible molecular pathways in our predictive ILC signature through the utilization of gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA). Five key genes were screened out by constructing a competing endogenous RNA (ceRNA) network using Cytoscape and their values in clinical indexes were demonstrated. Immunohistochemistry (IHC) in HCC cases confirmed the expression of these genes.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;Results: &lt;/strong&gt;ILC cell subsets were identified, and cell-cell communication analysis revealed that the signaling pathways involving ILC cell subsets were mostly abundant in the HCC microenvironment. Subsequently, 270 marker genes of the ILC clusters were subjected to GO and KEGG enrichment analysis. Furthermore, a total of 58 prognostically relevant genes were screened as features for prognostic prediction models. Next, the models were validated and clinically evaluated (P values of Kaplan-Meier survival curves below 0.05). Five key genes (&lt;i&gt;C2, STK4, CALM1, IL7R&lt;/i&gt;, and &lt;i&gt;RORA&lt;/i&gt;) were further screened by multi omics analysis of immune cell and factor and drug sensitivity and correlation analysis of tumor regulatory genes in liver cancer. Furthermore, the potential clinical value of the 5 key genes was confirmed in HCC patients. Finally, the IHC results confirmed the expression of &lt;i&gt;C2&lt;/i&gt;, &lt;i&gt;STK4&lt;/i&gt;, &lt;i&gt;CALM1&lt;/i&gt;, &lt;i&gt;IL7R&lt;/i&gt;, and &lt;i&gt;RORA&lt;/i&gt; in HCC. Our experimental results provided preliminary evidence supporting the oncogenic roles of &lt;i&gt;STK4&lt;/i&gt; and &lt;i&gt;CALM1&lt;/i&gt;, as well as the tumor-suppressive roles of &lt;i&gt;C2&lt;/i&gt;, &lt;i&gt;RORA&lt;/i&gt;, and &lt;i&gt;IL7R&lt;/i&gt; in HCC.&lt;/p&gt;&lt;p&gt;&lt;strong&gt;","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 10","pages":"5395-5416"},"PeriodicalIF":1.5,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11543044/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142627942","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 pan-cancer landscape of crosstalk between leukocyte transendothelial migration-related genes and tumor microenvironment relevant to prognosis and immunotherapy response. 与预后和免疫疗法反应相关的白细胞跨内皮细胞迁移相关基因与肿瘤微环境之间的串扰泛癌症景观。
IF 1.5 4区 医学 Q4 ONCOLOGY Pub Date : 2024-10-31 Epub Date: 2024-10-25 DOI: 10.21037/tcr-24-556
Hao Li, Xiaochen Niu, Rui Cheng

Background: Leukocyte transendothelial migration-related genes (LTEMGs) play a crucial role in the immune response and have been extensively studied in various pathological conditions, including inflammation, infection, and cancer. In recent years, increasing attention has been given to understanding the biological mechanisms of LTEMGs in the context of tumor progression and metastasis. The potential function of LTEMGs in cancer progression remains unclear. The aim of this study is to systematically delineate the relationship between LTEMGs and tumor prognosis and immune microenvironment at the pan-cancer level, providing new biomarkers for personalized immunotherapy.

Methods: The gene alteration, messenger RNA (mRNA) expression, and prognostic value of LTEMGs in pan-cancer were evaluated using Bulk and single-cell RNA (scRNA) sequence data. The LTEMGs score was calculated by R package "GSVA". The association of LTEMGs score with tumor microenvironment and immunotherapy response were deeply explored.

Results: We assessed the mRNA expression of 114 LTEMGs across various cancers, finding significant upregulation in acute myeloid leukemia (LAML) and pancreatic adenocarcinoma (PAAD). Prognostic analysis indicated most LTEMGs were risk factors in low-grade glioma (LGG), PAAD, uveal melanoma (UVM), and LAML. The LTEMGs score, highest in kidney renal clear cell carcinoma (KIRC) and lowest in UVM, was higher in tumor tissues compared to normal tissues in several cancers. The score was a risk factor for overall survival (OS) in LGG, UVM, and others, but protective in KIRC and some others. LTEMGs score correlated positively with Kirsten rat sarcoma viral oncogene homolog (KRAS) signaling, apoptosis, and immune responses. It also correlated with immune and stromal scores, and immune-related pathways. Higher LTEMGs score was linked to greater immune cell infiltration and poorer immunotherapy outcomes. Single-cell analysis revealed higher LTEMGs score in endothelial and monocyte cells, consistent with reduced immunotherapy responsiveness.

Conclusions: Our results reveal that LTEMGs are closely associated with tumor microenvironment. Patients with high LTEMGs score might be resistant to immunotherapy.

背景:白细胞跨内皮细胞迁移相关基因(LTEMGs)在免疫反应中起着至关重要的作用,在炎症、感染和癌症等各种病理情况下都有广泛的研究。近年来,人们越来越关注了解 LTEMGs 在肿瘤进展和转移过程中的生物学机制。LTEMGs在癌症进展中的潜在功能仍不清楚。本研究的目的是在泛癌症水平上系统地阐明LTEMGs与肿瘤预后和免疫微环境之间的关系,为个性化免疫疗法提供新的生物标志物:方法:利用大样本和单细胞RNA(scRNA)序列数据评估LTEMGs在泛癌症中的基因改变、信使RNA(mRNA)表达和预后价值。LTEMGs 评分由 R 软件包 "GSVA "计算。结果:我们评估了114种LTEMGs在不同癌症中的mRNA表达,发现它们在急性髓性白血病(LAML)和胰腺癌(PAAD)中显著上调。预后分析表明,大多数LTEMGs是低级别胶质瘤(LGG)、PAAD、葡萄膜黑色素瘤(UVM)和LAML的风险因素。在几种癌症中,与正常组织相比,肿瘤组织中的LTEMGs得分最高的是肾透明细胞癌(KIRC),最低的是葡萄膜黑色素瘤。在LGG、UVM和其他癌症中,LTEMGs评分是总生存率(OS)的风险因素,但在KIRC和其他一些癌症中,LTEMGs评分则具有保护作用。LTEMGs 评分与 Kirsten 大鼠肉瘤病毒癌基因同源物(KRAS)信号转导、细胞凋亡和免疫反应呈正相关。它还与免疫和基质评分以及免疫相关途径相关。LTEMGs得分越高,免疫细胞浸润越多,免疫治疗效果越差。单细胞分析显示,内皮细胞和单核细胞的LTEMGs得分较高,这与免疫治疗反应性降低一致:我们的研究结果表明,LTEMGs 与肿瘤微环境密切相关。结论:我们的研究结果表明,LTEMGs与肿瘤微环境密切相关,LTEMGs得分高的患者可能会对免疫疗法产生耐药性。
{"title":"The pan-cancer landscape of crosstalk between leukocyte transendothelial migration-related genes and tumor microenvironment relevant to prognosis and immunotherapy response.","authors":"Hao Li, Xiaochen Niu, Rui Cheng","doi":"10.21037/tcr-24-556","DOIUrl":"https://doi.org/10.21037/tcr-24-556","url":null,"abstract":"<p><strong>Background: </strong>Leukocyte transendothelial migration-related genes (LTEMGs) play a crucial role in the immune response and have been extensively studied in various pathological conditions, including inflammation, infection, and cancer. In recent years, increasing attention has been given to understanding the biological mechanisms of LTEMGs in the context of tumor progression and metastasis. The potential function of LTEMGs in cancer progression remains unclear. The aim of this study is to systematically delineate the relationship between LTEMGs and tumor prognosis and immune microenvironment at the pan-cancer level, providing new biomarkers for personalized immunotherapy.</p><p><strong>Methods: </strong>The gene alteration, messenger RNA (mRNA) expression, and prognostic value of LTEMGs in pan-cancer were evaluated using Bulk and single-cell RNA (scRNA) sequence data. The LTEMGs score was calculated by R package \"GSVA\". The association of LTEMGs score with tumor microenvironment and immunotherapy response were deeply explored.</p><p><strong>Results: </strong>We assessed the mRNA expression of 114 LTEMGs across various cancers, finding significant upregulation in acute myeloid leukemia (LAML) and pancreatic adenocarcinoma (PAAD). Prognostic analysis indicated most LTEMGs were risk factors in low-grade glioma (LGG), PAAD, uveal melanoma (UVM), and LAML. The LTEMGs score, highest in kidney renal clear cell carcinoma (KIRC) and lowest in UVM, was higher in tumor tissues compared to normal tissues in several cancers. The score was a risk factor for overall survival (OS) in LGG, UVM, and others, but protective in KIRC and some others. LTEMGs score correlated positively with Kirsten rat sarcoma viral oncogene homolog (KRAS) signaling, apoptosis, and immune responses. It also correlated with immune and stromal scores, and immune-related pathways. Higher LTEMGs score was linked to greater immune cell infiltration and poorer immunotherapy outcomes. Single-cell analysis revealed higher LTEMGs score in endothelial and monocyte cells, consistent with reduced immunotherapy responsiveness.</p><p><strong>Conclusions: </strong>Our results reveal that LTEMGs are closely associated with tumor microenvironment. Patients with high LTEMGs score might be resistant to immunotherapy.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 10","pages":"5247-5264"},"PeriodicalIF":1.5,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11543035/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142628772","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 potential role of Ral-interacting protein 76 and vascular endothelial growth factor on angiogenesis in the tumor and ovarian corpus luteum microenvironment. Ral-interacting protein 76 和血管内皮生长因子对肿瘤和卵巢黄体微环境中血管生成的潜在作用。
IF 1.5 4区 医学 Q4 ONCOLOGY Pub Date : 2024-10-31 Epub Date: 2024-10-17 DOI: 10.21037/tcr-24-770
Seunghyung Lee

Tumors and the ovarian corpus luteum have complex mechanisms in the growth microenvironment. Angiogenesis is especially important for demonstrating the molecular mechanism of dynamic cellular function in tumors and corpus luteum. Angiogenesis in tumors and corpus luteum seems to have a similar function, and Ral-interacting protein 76 (RLIP76) and vascular endothelial growth factor (VEGF) are expressed in the tissues of tumors and ovarian corpus luteum. RLIP76 is a potential factor with VEGF in the tumor and corpus luteum angiogenesis. RLIP76 regulates a small GTPase (R-Ras) in cell survival, spreading, and migration. VEGF activates angiogenic functions in tumor and endothelial cells. Hypoxia-inducible factor-1 (HIF-1) is important in tumor growth, tumor angiogenesis, and corpus luteum. VEGF and HIF-1 regulate the angiogenic function of RLIP76, and RLIP76 controls vascular growth in endothelial and tumor cells. RLIP76, R-Ras, VEGF, and HIF-1 may be useful in the research of corpus luteum and cancer therapy and the study of mechanisms of tumor and corpus luteum angiogenesis. This review will help to elucidate the roles of RLIP76 and VEGF in tumor and corpus luteum angiogenesis, tumorigenesis, and the specific regulation of RLIP76 and VEGF. Thus, we reviewed the potential role of RLIP76 and VEGF in the angiogenesis of the tumor and corpus luteum in the tumor and ovarian microenvironment.

肿瘤和卵巢黄体的生长微环境机制复杂。血管生成对于展示肿瘤和黄体动态细胞功能的分子机制尤为重要。肿瘤和黄体的血管生成似乎具有相似的功能,Ral-互作蛋白76(RLIP76)和血管内皮生长因子(VEGF)在肿瘤和卵巢黄体组织中均有表达。RLIP76 与血管内皮生长因子是肿瘤和黄体血管生成的潜在因子。RLIP76 在细胞存活、扩散和迁移过程中调节一种小 GTPase(R-Ras)。血管内皮生长因子可激活肿瘤细胞和内皮细胞的血管生成功能。低氧诱导因子-1(HIF-1)在肿瘤生长、肿瘤血管生成和黄体中具有重要作用。血管内皮生长因子和 HIF-1 调节 RLIP76 的血管生成功能,RLIP76 控制内皮细胞和肿瘤细胞的血管生长。RLIP76、R-Ras、VEGF和HIF-1可能有助于黄体和癌症治疗的研究以及肿瘤和黄体血管生成机制的研究。本综述将有助于阐明 RLIP76 和 VEGF 在肿瘤和黄体血管生成、肿瘤发生中的作用,以及 RLIP76 和 VEGF 的特异性调控。因此,我们综述了 RLIP76 和 VEGF 在肿瘤和卵巢微环境中对肿瘤和黄体血管生成的潜在作用。
{"title":"The potential role of Ral-interacting protein 76 and vascular endothelial growth factor on angiogenesis in the tumor and ovarian corpus luteum microenvironment.","authors":"Seunghyung Lee","doi":"10.21037/tcr-24-770","DOIUrl":"https://doi.org/10.21037/tcr-24-770","url":null,"abstract":"<p><p>Tumors and the ovarian corpus luteum have complex mechanisms in the growth microenvironment. Angiogenesis is especially important for demonstrating the molecular mechanism of dynamic cellular function in tumors and corpus luteum. Angiogenesis in tumors and corpus luteum seems to have a similar function, and Ral-interacting protein 76 (RLIP76) and vascular endothelial growth factor (VEGF) are expressed in the tissues of tumors and ovarian corpus luteum. RLIP76 is a potential factor with VEGF in the tumor and corpus luteum angiogenesis. RLIP76 regulates a small GTPase (R-Ras) in cell survival, spreading, and migration. VEGF activates angiogenic functions in tumor and endothelial cells. Hypoxia-inducible factor-1 (HIF-1) is important in tumor growth, tumor angiogenesis, and corpus luteum. VEGF and HIF-1 regulate the angiogenic function of RLIP76, and RLIP76 controls vascular growth in endothelial and tumor cells. RLIP76, R-Ras, VEGF, and HIF-1 may be useful in the research of corpus luteum and cancer therapy and the study of mechanisms of tumor and corpus luteum angiogenesis. This review will help to elucidate the roles of RLIP76 and VEGF in tumor and corpus luteum angiogenesis, tumorigenesis, and the specific regulation of RLIP76 and VEGF. Thus, we reviewed the potential role of RLIP76 and VEGF in the angiogenesis of the tumor and corpus luteum in the tumor and ovarian microenvironment.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 10","pages":"5702-5710"},"PeriodicalIF":1.5,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11543058/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142628776","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
Risk signature of NETosis-related subtype predicts prognosis and evaluates immunotherapy effectiveness in gastric cancer. NETosis相关亚型的风险特征可预测胃癌的预后并评估免疫疗法的效果。
IF 1.5 4区 医学 Q4 ONCOLOGY Pub Date : 2024-10-31 Epub Date: 2024-10-29 DOI: 10.21037/tcr-24-377
Ruyue Chen, Zengwu Yao, Lixin Jiang

Background: Gastric cancer (GC) has a high incidence and mortality rate with a poor prognosis, so it is crucial to search for new biomarkers. The role of NETosis, a newly identified type of programmed cell death, in GC and its underlying mechanisms have yet to be explored and still require thorough investigation. Our research seeks to enhance our comprehension of NETosis and may offer novel approaches for treating GC.

Methods: Utilizing The Cancer Genome Atlas-stomach adenocarcinoma (TCGA-STAD) dataset for training and the GSE84433 dataset for validation, our study delved into the associations between NETosis-related genes and the clinical risk of GC. Through comprehensive clustering, enrichment, and immune infiltration analyses, we evaluated the prognostic relevance of these NETosis genes in vivo. Furthermore, we devised a NETosis-related risk signature (NRRS) to assess its implications in risk stratification, survival prognosis, immune infiltration, and drug sensitivity. The NRRS's accuracy was validated by immunohistochemical staining.

Results: By applying consensus clustering to data from 62 NETosis-related genes, we categorized patients into two distinct subgroups, C1 and C2. These subgroups demonstrated significant differences. Following this, we developed the NRRS using the least absolute shrinkage and selection operator (LASSO) regression analysis. This process involved the selection of the following genes: CXCR4, NRP1, PDCD1, CTLA4, AKR1B1, SERPINE1, RGS2, SLCO2A1, TNFAIP2, RNASE1, DOC2B, APOD, ENTPD2, and CCL24. High-risk and low-risk groups can be accurately distinguished. We further verify in the verification set. These results suggest that NETosis is related to the microenvironment of GC. Our designed NRRS can predict the survival of patients with GC.

Conclusions: We emphasized the relationship between NETosis and GC. We built and validated the value of NRRS. This contributes to deepening our view of NETosis and potentially provides new strategies for GC treatment.

背景:胃癌(GC)发病率高、死亡率高、预后差,因此寻找新的生物标志物至关重要。NETosis是一种新发现的程序性细胞死亡类型,它在胃癌中的作用及其内在机制尚待探索,仍需深入研究。我们的研究旨在加深我们对 NETosis 的理解,并为治疗 GC 提供新的方法:我们的研究利用癌症基因组图谱-胃腺癌(TCGA-STAD)数据集进行训练,并利用GSE84433数据集进行验证,深入研究NETosis相关基因与GC临床风险之间的关联。通过综合聚类、富集和免疫浸润分析,我们评估了这些 NETosis 基因在体内的预后相关性。此外,我们还设计了NETosis相关风险特征(NRRS),以评估其在风险分层、生存预后、免疫浸润和药物敏感性方面的意义。免疫组化染色验证了NRRS的准确性:通过对 62 个 NETosis 相关基因的数据进行共识聚类,我们将患者分为两个不同的亚组:C1 和 C2。这些亚组显示出显著差异。随后,我们利用最小绝对收缩和选择算子(LASSO)回归分析法开发了 NRRS。在此过程中,我们选择了以下基因:CXCR4、NRP1、PDCD1、CTLA4、AKR1B1、SERPINE1、RGS2、SLCO2A1、TNFAIP2、RNASE1、DOC2B、APOD、ENTPD2 和 CCL24。高风险组和低风险组可以准确区分。我们在验证集中进行了进一步验证。这些结果表明,NETosis 与 GC 的微环境有关。我们设计的 NRRS 可以预测 GC 患者的生存期:我们强调了NETosis与GC之间的关系。我们建立并验证了 NRRS 的价值。这有助于加深我们对 NETosis 的认识,并有可能为 GC 治疗提供新策略。
{"title":"Risk signature of NETosis-related subtype predicts prognosis and evaluates immunotherapy effectiveness in gastric cancer.","authors":"Ruyue Chen, Zengwu Yao, Lixin Jiang","doi":"10.21037/tcr-24-377","DOIUrl":"https://doi.org/10.21037/tcr-24-377","url":null,"abstract":"<p><strong>Background: </strong>Gastric cancer (GC) has a high incidence and mortality rate with a poor prognosis, so it is crucial to search for new biomarkers. The role of NETosis, a newly identified type of programmed cell death, in GC and its underlying mechanisms have yet to be explored and still require thorough investigation. Our research seeks to enhance our comprehension of NETosis and may offer novel approaches for treating GC.</p><p><strong>Methods: </strong>Utilizing The Cancer Genome Atlas-stomach adenocarcinoma (TCGA-STAD) dataset for training and the GSE84433 dataset for validation, our study delved into the associations between NETosis-related genes and the clinical risk of GC. Through comprehensive clustering, enrichment, and immune infiltration analyses, we evaluated the prognostic relevance of these NETosis genes <i>in vivo</i>. Furthermore, we devised a NETosis-related risk signature (NRRS) to assess its implications in risk stratification, survival prognosis, immune infiltration, and drug sensitivity. The NRRS's accuracy was validated by immunohistochemical staining.</p><p><strong>Results: </strong>By applying consensus clustering to data from 62 NETosis-related genes, we categorized patients into two distinct subgroups, C1 and C2. These subgroups demonstrated significant differences. Following this, we developed the NRRS using the least absolute shrinkage and selection operator (LASSO) regression analysis. This process involved the selection of the following genes: <i>CXCR4, NRP1, PDCD1, CTLA4, AKR1B1, SERPINE1, RGS2, SLCO2A1, TNFAIP2, RNASE1, DOC2B, APOD, ENTPD2</i>, and <i>CCL24</i>. High-risk and low-risk groups can be accurately distinguished. We further verify in the verification set. These results suggest that NETosis is related to the microenvironment of GC. Our designed NRRS can predict the survival of patients with GC.</p><p><strong>Conclusions: </strong>We emphasized the relationship between NETosis and GC. We built and validated the value of NRRS. This contributes to deepening our view of NETosis and potentially provides new strategies for GC treatment.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 10","pages":"5165-5177"},"PeriodicalIF":1.5,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11543040/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142628736","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
Capicua transcriptional repressor (CIC)-rearranged sarcoma harboring CIC-LEUTX fusion with renal involvement: a rare case report. 卡皮库亚转录抑制因子(CIC)重排肉瘤携带CIC-LEUTX融合并累及肾脏:一例罕见病例报告。
IF 1.5 4区 医学 Q4 ONCOLOGY Pub Date : 2024-10-31 Epub Date: 2024-10-24 DOI: 10.21037/tcr-24-524
Jing Zhu, Chao Chen, Yan Li

Background: Capicua transcriptional repressor (CIC)-rearranged sarcoma (CRS) is a rare and highly aggressive undifferentiated small round cell sarcoma (USRCS), which genetically displays a characteristic gene fusion between CIC gene with other genes such as DUX4.

Case description: We report a rare case with CIC-LEUTX fusion. The 45-year-old male patient presented to our department with frequent dry cough and lumbar pain. Computed tomography (CT) scan indicated multiple pulmonary nodules on both sides, hilar lymph node enlargement, left lower lobe infection and presence of pleural effusion in left quadrant. Enlargement and uneven density were seen in the left kidney, together with perinephric exudate, renal calculi, thickening and filling deficiency in renal veins, and small retroperitoneal lymph nodes. Immunohistochemistry (IHC) showed negativity in CK7, CK20, CD10, vimentin, PAX-8, P63, MelanA and synapatophysin, but GATA3 and CK were positive. RNA-based next generation sequencing (NGS) detection revealed CRS of gene fusion in CIC (exon 20) and LEUTX (exon 3). After treatment with a variety of targeted and chemotherapy drugs, the patient showed a poor response with a survival time of merely 7 months.

Conclusions: This case of USRCS harboring CIC-LEUTX fusion with renal involvement could help to expand our understanding on the diagnosis and treatment of CRS harboring CIC-LEUTX fusion.

背景:卡皮夸转录抑制因子(CIC)重组肉瘤(CRS)是一种罕见的高侵袭性未分化小圆形细胞肉瘤(USRCS),在遗传学上表现出CIC基因与DUX4等其他基因融合的特征:我们报告了一例罕见的 CIC-LEUTX 融合病例。这名 45 岁的男性患者因频繁干咳和腰痛来我科就诊。计算机断层扫描(CT)显示患者两侧肺部多发结节,肺门淋巴结肿大,左下叶感染,左象限胸腔积液。左肾肿大,密度不均,肾周渗出物,肾结石,肾静脉增粗,充盈不足,腹膜后淋巴结肿大。免疫组化(IHC)显示,CK7、CK20、CD10、波形蛋白、PAX-8、P63、MelanA和突触素阴性,但GATA3和CK阳性。基于 RNA 的新一代测序(NGS)检测显示,CIC(20 号外显子)和 LEUTX(3 号外显子)的基因融合为 CRS。经过多种靶向药物和化疗药物治疗后,患者的反应不佳,存活时间仅为 7 个月:本例携带CIC-LEUTX基因融合并累及肾脏的USRCS病例有助于拓展我们对携带CIC-LEUTX基因融合的CRS诊断和治疗的认识。
{"title":"Capicua transcriptional repressor (CIC)-rearranged sarcoma harboring <i>CIC-LEUTX</i> fusion with renal involvement: a rare case report.","authors":"Jing Zhu, Chao Chen, Yan Li","doi":"10.21037/tcr-24-524","DOIUrl":"https://doi.org/10.21037/tcr-24-524","url":null,"abstract":"<p><strong>Background: </strong>Capicua transcriptional repressor (CIC)-rearranged sarcoma (CRS) is a rare and highly aggressive undifferentiated small round cell sarcoma (USRCS), which genetically displays a characteristic gene fusion between <i>CIC</i> gene with other genes such as <i>DUX4</i>.</p><p><strong>Case description: </strong>We report a rare case with <i>CIC-LEUTX</i> fusion. The 45-year-old male patient presented to our department with frequent dry cough and lumbar pain. Computed tomography (CT) scan indicated multiple pulmonary nodules on both sides, hilar lymph node enlargement, left lower lobe infection and presence of pleural effusion in left quadrant. Enlargement and uneven density were seen in the left kidney, together with perinephric exudate, renal calculi, thickening and filling deficiency in renal veins, and small retroperitoneal lymph nodes. Immunohistochemistry (IHC) showed negativity in CK7, CK20, CD10, vimentin, PAX-8, P63, MelanA and synapatophysin, but GATA3 and CK were positive. RNA-based next generation sequencing (NGS) detection revealed CRS of gene fusion in <i>CIC</i> (exon 20) and <i>LEUTX</i> (exon 3). After treatment with a variety of targeted and chemotherapy drugs, the patient showed a poor response with a survival time of merely 7 months.</p><p><strong>Conclusions: </strong>This case of USRCS harboring <i>CIC-LEUTX</i> fusion with renal involvement could help to expand our understanding on the diagnosis and treatment of CRS harboring <i>CIC-LEUTX</i> fusion.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 10","pages":"5711-5718"},"PeriodicalIF":1.5,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11543050/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142628859","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
Clinical prognostic value of TREM1 in patients with liver cancer lung metastasis. TREM1在肝癌肺转移患者中的临床预后价值
IF 1.5 4区 医学 Q4 ONCOLOGY Pub Date : 2024-10-31 Epub Date: 2024-10-29 DOI: 10.21037/tcr-24-492
Yi Luo, Jie Cai, Yanze Yin, Qiang Xia

Background: Patients diagnosed with hepatocellular carcinoma (HCC) generally have an unfavorable outlook, with lung metastasis being a prevalent factor contributing to mortality. The metastatic microenvironment is critical to the tumor metastatic process. The exact impact of Triggering Receptor Expressed on Myeloid Cells 1 (TREM1) on tumor metastasis and the microenvironment of metastasis is still not known. By analyzing online databases and a clinical cohort, we evaluated the predictive significance of TREM1 and its correlation with the tumor microenvironment (TME).

Methods: Using the Gene Expression Omnibus (GEO) dataset (GSE141016), genes differentially expressed in liver cancer and lung metastases were analyzed. Data from liver hepatocellular carcinoma (LIHC) of The Cancer Genome Atlas (TCGA) were acquired through RNA sequencing. The abundance of tumor-infiltrating immune cells was estimated using Estimation of Stromal and Immune cells in Malignant Tumor tissues using Expression data (ESTIMATE). The single sample gene set enrichment analysis (ssGSEA) algorithm was utilized to determine the association between TREM1 and immune cells. The level of TREM1 and immune cells were determined in formalin-fixed paraffin-embedding (FFPE) specimens.

Results: Increased expression of TREM1 in HCC was linked to a poorer clinical prognosis and elevated incidence of lung metastasis. Furthermore, TREM1 was found to be associated with multiple immune cells in the TME. We noticed that lung metastases in the same patient had higher levels of TREM1 protein compared to primary liver cancer. Additionally, lung metastases exhibited increased neutrophil numbers and neutrophil extracellular traps (NETs) formation compared to primary liver cancer. Moreover, there was a positive correlation between TREM1 and both neutrophils and NETs.

Conclusions: Increased expression of TREM1 in HCC is linked to a poorer clinical outlook and elevated incidence of lung metastasis, suggesting its potential as a prognostic biomarker for patients with liver cancer lung metastasis.

背景:确诊为肝细胞癌(HCC)的患者一般前景不佳,肺转移是导致死亡的主要因素。转移微环境对肿瘤转移过程至关重要。髓系细胞上表达的触发受体1(TREM1)对肿瘤转移和转移微环境的确切影响尚不清楚。通过分析在线数据库和临床队列,我们评估了TREM1的预测意义及其与肿瘤微环境(TME)的相关性:利用基因表达总库(GEO)数据集(GSE141016)分析了肝癌和肺转移瘤中差异表达的基因。癌症基因组图谱(TCGA)中肝肝细胞癌(LIHC)的数据是通过 RNA 测序获得的。利用表达数据估算恶性肿瘤组织中的基质和免疫细胞(ESTIMATE)估算了肿瘤浸润免疫细胞的丰度。利用单样本基因组富集分析(ssGSEA)算法确定了 TREM1 与免疫细胞之间的关联。在福尔马林固定石蜡包埋(FFPE)标本中测定了TREM1和免疫细胞的水平:结果:TREM1在HCC中的高表达与较差的临床预后和较高的肺转移发生率有关。此外,TREM1还与TME中的多种免疫细胞有关。我们注意到,与原发性肝癌相比,同一患者的肺转移灶的TREM1蛋白水平更高。此外,与原发性肝癌相比,肺转移灶的中性粒细胞数量和中性粒细胞胞外捕获物(NET)的形成均有所增加。此外,TREM1与中性粒细胞和NETs之间存在正相关:结论:TREM1在肝癌中的表达增加与较差的临床前景和较高的肺转移发生率有关,这表明它有可能成为肝癌肺转移患者的预后生物标志物。
{"title":"Clinical prognostic value of TREM1 in patients with liver cancer lung metastasis.","authors":"Yi Luo, Jie Cai, Yanze Yin, Qiang Xia","doi":"10.21037/tcr-24-492","DOIUrl":"https://doi.org/10.21037/tcr-24-492","url":null,"abstract":"<p><strong>Background: </strong>Patients diagnosed with hepatocellular carcinoma (HCC) generally have an unfavorable outlook, with lung metastasis being a prevalent factor contributing to mortality. The metastatic microenvironment is critical to the tumor metastatic process. The exact impact of Triggering Receptor Expressed on Myeloid Cells 1 (TREM1) on tumor metastasis and the microenvironment of metastasis is still not known. By analyzing online databases and a clinical cohort, we evaluated the predictive significance of TREM1 and its correlation with the tumor microenvironment (TME).</p><p><strong>Methods: </strong>Using the Gene Expression Omnibus (GEO) dataset (GSE141016), genes differentially expressed in liver cancer and lung metastases were analyzed. Data from liver hepatocellular carcinoma (LIHC) of The Cancer Genome Atlas (TCGA) were acquired through RNA sequencing. The abundance of tumor-infiltrating immune cells was estimated using Estimation of Stromal and Immune cells in Malignant Tumor tissues using Expression data (ESTIMATE). The single sample gene set enrichment analysis (ssGSEA) algorithm was utilized to determine the association between TREM1 and immune cells. The level of TREM1 and immune cells were determined in formalin-fixed paraffin-embedding (FFPE) specimens.</p><p><strong>Results: </strong>Increased expression of TREM1 in HCC was linked to a poorer clinical prognosis and elevated incidence of lung metastasis. Furthermore, TREM1 was found to be associated with multiple immune cells in the TME. We noticed that lung metastases in the same patient had higher levels of TREM1 protein compared to primary liver cancer. Additionally, lung metastases exhibited increased neutrophil numbers and neutrophil extracellular traps (NETs) formation compared to primary liver cancer. Moreover, there was a positive correlation between TREM1 and both neutrophils and NETs.</p><p><strong>Conclusions: </strong>Increased expression of TREM1 in HCC is linked to a poorer clinical outlook and elevated incidence of lung metastasis, suggesting its potential as a prognostic biomarker for patients with liver cancer lung metastasis.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 10","pages":"5446-5457"},"PeriodicalIF":1.5,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11543053/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142628867","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
S100A1 overexpression stimulates cell proliferation and is predictive of poor outcome in ovarian cancer. S100A1 过表达可刺激细胞增殖,并可预测卵巢癌的不良预后。
IF 1.5 4区 医学 Q4 ONCOLOGY Pub Date : 2024-10-31 Epub Date: 2024-10-21 DOI: 10.21037/tcr-24-430
Wen Jin, Hui Hui, Jie Jiang, Bin Li, Zhuo Deng, Xiaoqian Tuo

Background: Members of the S100 gene family are frequently dysregulated in various cancers, including ovarian cancer (OC). Despite this, the prognostic implications of individual S100 genes in OC remain poorly understood. This study aimed to explore the prognostic significance of S100A1 expression in OC and assess its potential as a therapeutic target.

Methods: To investigate the role of S100A1 in OC, we utilized the Gene Expression Profiling Interactive Analysis (GEPIA) database and the University of ALabama at Birmingham Cancer Data Analysis Portal (UALCAN) database. Protein levels of S100A1 in OC tissues were assessed using western blotting and immunohistochemistry. Bioinformatics analyses were performed to correlate S100A1 expression with clinical outcomes. Functional assays were conducted to evaluate the impact of S100A1 knockout on OC cell proliferation and migration. Additionally, we investigated the effect of S100A1 on ferroptosis and lipid reactive oxygen species (ROS) levels in tumor cells.

Results: Our analyses revealed that S100A1 protein levels were significantly elevated in OC tissues compared to normal tissues. Elevated S100A1 expression was associated with poor clinical outcomes in OC patients. Functional assays demonstrated that the knockout of S100A1 led to a decrease in both proliferation and migration of OC cells in vitro. Furthermore, S100A1 was found to inhibit ferroptosis in OC cells, resulting in lower levels of lipid ROS within tumor cells.

Conclusions: High levels of S100A1 are indicative of adverse clinical outcomes in OC. Our findings suggest that S100A1 could serve as a valuable prognostic marker and a potential therapeutic target for OC treatment.

背景:在包括卵巢癌(OC)在内的各种癌症中,S100 基因家族的成员经常发生调控失调。尽管如此,人们对单个S100基因在卵巢癌中的预后影响仍知之甚少。本研究旨在探讨S100A1表达在OC中的预后意义,并评估其作为治疗靶点的潜力:为了研究S100A1在OC中的作用,我们利用了基因表达谱交互分析(GEPIA)数据库和伯明翰阿拉巴马大学癌症数据分析门户(UALCAN)数据库。采用免疫印迹法和免疫组化法评估了OC组织中S100A1的蛋白水平。生物信息学分析将 S100A1 的表达与临床结果相关联。我们进行了功能测试,以评估 S100A1 基因敲除对 OC 细胞增殖和迁移的影响。此外,我们还研究了S100A1对肿瘤细胞中铁细胞凋亡和脂质活性氧(ROS)水平的影响:结果:我们的分析发现,与正常组织相比,OC 组织中 S100A1 蛋白水平明显升高。S100A1表达的升高与OC患者的不良临床预后有关。功能测试表明,敲除 S100A1 会导致体外 OC 细胞的增殖和迁移减少。此外,研究还发现S100A1能抑制OC细胞的铁突变,从而降低肿瘤细胞内的脂质ROS水平:结论:高水平的S100A1是OC不良临床结局的标志。我们的研究结果表明,S100A1可作为有价值的预后标志物和OC治疗的潜在治疗靶点。
{"title":"<i>S100A1</i> overexpression stimulates cell proliferation and is predictive of poor outcome in ovarian cancer.","authors":"Wen Jin, Hui Hui, Jie Jiang, Bin Li, Zhuo Deng, Xiaoqian Tuo","doi":"10.21037/tcr-24-430","DOIUrl":"https://doi.org/10.21037/tcr-24-430","url":null,"abstract":"<p><strong>Background: </strong>Members of the S100 gene family are frequently dysregulated in various cancers, including ovarian cancer (OC). Despite this, the prognostic implications of individual S100 genes in OC remain poorly understood. This study aimed to explore the prognostic significance of <i>S100A1</i> expression in OC and assess its potential as a therapeutic target.</p><p><strong>Methods: </strong>To investigate the role of <i>S100A1</i> in OC, we utilized the Gene Expression Profiling Interactive Analysis (GEPIA) database and the University of ALabama at Birmingham Cancer Data Analysis Portal (UALCAN) database. Protein levels of S100A1 in OC tissues were assessed using western blotting and immunohistochemistry. Bioinformatics analyses were performed to correlate <i>S100A1</i> expression with clinical outcomes. Functional assays were conducted to evaluate the impact of <i>S100A1</i> knockout on OC cell proliferation and migration. Additionally, we investigated the effect of <i>S100A1</i> on ferroptosis and lipid reactive oxygen species (ROS) levels in tumor cells.</p><p><strong>Results: </strong>Our analyses revealed that S100A1 protein levels were significantly elevated in OC tissues compared to normal tissues. Elevated <i>S100A1</i> expression was associated with poor clinical outcomes in OC patients. Functional assays demonstrated that the knockout of <i>S100A1</i> led to a decrease in both proliferation and migration of OC cells <i>in vitro</i>. Furthermore, <i>S100A1</i> was found to inhibit ferroptosis in OC cells, resulting in lower levels of lipid ROS within tumor cells.</p><p><strong>Conclusions: </strong>High levels of <i>S100A1</i> are indicative of adverse clinical outcomes in OC. Our findings suggest that <i>S100A1</i> could serve as a valuable prognostic marker and a potential therapeutic target for OC treatment.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 10","pages":"5265-5277"},"PeriodicalIF":1.5,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11543041/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142628849","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
Translational cancer research
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1