Pub Date : 2024-12-31Epub Date: 2024-12-27DOI: 10.21037/tcr-24-802
Tongtong Bai, Chengyu Wu
Background: The causal relationship between percentage of fat in milk consumption and cancer risk lacks sufficient investigation. The purpose of this study was to explore whether the percentage of fat in milk consumption is a factor that affects the risk variation of several common types of cancer.
Methods: Mendelian randomization (MR) was performed to estimate the unconfounded causal relationship between the percentage of fat in milk consumption and the risk of six cancers related to milk intake, as well as to assess the associations between body fat percentage and these cancers. Data corresponding to the percentage of fat in milk consumption (n=411,503), body fat percentage (n=401,772), breast cancer (n=139,274), ovarian cancer (n=66,450), endometrial cancer (n=121,885), colorectal cancer (n=32,072), prostate cancer (n=140,254), and bladder cancer (n=373,295) were obtained from the Integrative Epidemiology Unit (IEU) or the genome-wide association study (GWAS) Catalog databases. The primary analytical strategy employed the inverse-variance weighted (IVW) method. Sensitivity analysis, including assessments of heterogeneity and pleiotropy, was conducted to assess the robustness of the findings.
Results: The percentage of fat in milk consumption only exhibited a causal relationship with breast cancer (β=2.993, P=0.01). The study identified significant causal effects of body fat percentage on the risk of several cancers, including ovarian cancer (β=0.225, P=0.002), endometrial cancer (β=0.669, P<0.001), and colorectal cancer (β=0.344, P<0.001), as well as a protective effect on prostate cancer (β=-0.104, P=0.046). Sensitivity analysis demonstrated that the findings were robust.
Conclusions: Our study findings indicated that a higher percentage of fat in milk consumption was associated with an increased risk of breast cancer, providing valuable insights for cancer prevention strategies among the European population.
{"title":"Percentage of fat in milk consumption and risk of six cancers: a Mendelian randomization study.","authors":"Tongtong Bai, Chengyu Wu","doi":"10.21037/tcr-24-802","DOIUrl":"https://doi.org/10.21037/tcr-24-802","url":null,"abstract":"<p><strong>Background: </strong>The causal relationship between percentage of fat in milk consumption and cancer risk lacks sufficient investigation. The purpose of this study was to explore whether the percentage of fat in milk consumption is a factor that affects the risk variation of several common types of cancer.</p><p><strong>Methods: </strong>Mendelian randomization (MR) was performed to estimate the unconfounded causal relationship between the percentage of fat in milk consumption and the risk of six cancers related to milk intake, as well as to assess the associations between body fat percentage and these cancers. Data corresponding to the percentage of fat in milk consumption (n=411,503), body fat percentage (n=401,772), breast cancer (n=139,274), ovarian cancer (n=66,450), endometrial cancer (n=121,885), colorectal cancer (n=32,072), prostate cancer (n=140,254), and bladder cancer (n=373,295) were obtained from the Integrative Epidemiology Unit (IEU) or the genome-wide association study (GWAS) Catalog databases. The primary analytical strategy employed the inverse-variance weighted (IVW) method. Sensitivity analysis, including assessments of heterogeneity and pleiotropy, was conducted to assess the robustness of the findings.</p><p><strong>Results: </strong>The percentage of fat in milk consumption only exhibited a causal relationship with breast cancer (β=2.993, P=0.01). The study identified significant causal effects of body fat percentage on the risk of several cancers, including ovarian cancer (β=0.225, P=0.002), endometrial cancer (β=0.669, P<0.001), and colorectal cancer (β=0.344, P<0.001), as well as a protective effect on prostate cancer (β=-0.104, P=0.046). Sensitivity analysis demonstrated that the findings were robust.</p><p><strong>Conclusions: </strong>Our study findings indicated that a higher percentage of fat in milk consumption was associated with an increased risk of breast cancer, providing valuable insights for cancer prevention strategies among the European population.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 12","pages":"6613-6622"},"PeriodicalIF":1.5,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11730444/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012317","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}
Pub Date : 2024-12-31Epub Date: 2024-12-27DOI: 10.21037/tcr-24-1258
Jiakai He, Jiayin Guo, Pengfei Sun
<p><strong>Background: </strong>Within the realm of primary brain tumors, specifically glioblastoma (GBM), presents a notable obstacle due to their unfavorable prognosis and differing median survival rates contingent upon tumor grade and subtype. Despite a plethora of research connecting cardiotrophin-1 (CTF1) modifications to a range of illnesses, its correlation with glioma remains uncertain. This study investigated the clinical value of CTF1 in glioma and its potential as a biomarker of the disease.</p><p><strong>Methods: </strong>Glioma project in The Cancer Genome Atlas (TCGA) database served as the training cohort, and CGGA 325 series in the Chinese Glioma Genome Atlas (CGGA) database served as the external independent validation cohort. First, the difference in the expression level of CTF1 between glioma tissue and normal tissue was analyzed, and the results were verified with the CGGA database. The relationship between CTF1 expression and the prognosis of glioma patients was evaluated using Univariate and Multivariate Cox analysis and the Kaplan-Meier (KM) curve. We used CIBERSOFT to explore the association between CTF1 and immune cell infiltration in GBM, as well as performing gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA) analyses. Furthermore, we analyzed the relationship between CTF1 and gene mutations and drug sensitivity. Using Weighted gene co-expression network analysis (WGCNA) analysis, we pinpointed the gene set most correlated with CTF1 and conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) gene enrichment analyses to anticipate the pathways that could be influenced by CTF1. Finally, we constructed a nomogram using a multifactorial regression model to further predict patient prognosis.</p><p><strong>Results: </strong>CTF1 expression was significantly elevated in glioma tissues compared to normal tissues in the TCGA dataset (P<0.001) and was associated with poorer survival in both TCGA and CGGA datasets (P<0.001). Receiver operating characteristic (ROC) analysis demonstrated the diagnostic potential of CTF1, with an area under the curve (AUC) of 0.889 [95% confidence interval (CI): 0.803-0.974] in TCGA and 0.664 (95% CI: 0.599-0.729) in CGGA. High CTF1 levels were correlated with advanced glioma grades, and Cox regression analysis identified CTF1 as an independent risk factor. A nomogram incorporating CTF1 levels, isocitrate dehydrogenase 1 (<i>IDH1</i>) mutation status, O6-methylguanine-DNA methyltransferase (<i>MGMT</i>) methylation status, age, and gender were developed and validated to predict 1- and 2-year survival probabilities. In GBM, drug sensitivity analysis revealed significant associations between CTF1 expression and responsiveness to gemcitabine, dasatinib, and other agents. CTF1 expression was also linked to immune infiltration (monocytes, neutrophils, M0 macrophages) and pathways involved in tumor progression, including IL2_STAT5, P53, and IL6_JAK_STAT3 sign
{"title":"Prognostic value of CTF1 in glioma and its role in the tumor microenvironment.","authors":"Jiakai He, Jiayin Guo, Pengfei Sun","doi":"10.21037/tcr-24-1258","DOIUrl":"https://doi.org/10.21037/tcr-24-1258","url":null,"abstract":"<p><strong>Background: </strong>Within the realm of primary brain tumors, specifically glioblastoma (GBM), presents a notable obstacle due to their unfavorable prognosis and differing median survival rates contingent upon tumor grade and subtype. Despite a plethora of research connecting cardiotrophin-1 (CTF1) modifications to a range of illnesses, its correlation with glioma remains uncertain. This study investigated the clinical value of CTF1 in glioma and its potential as a biomarker of the disease.</p><p><strong>Methods: </strong>Glioma project in The Cancer Genome Atlas (TCGA) database served as the training cohort, and CGGA 325 series in the Chinese Glioma Genome Atlas (CGGA) database served as the external independent validation cohort. First, the difference in the expression level of CTF1 between glioma tissue and normal tissue was analyzed, and the results were verified with the CGGA database. The relationship between CTF1 expression and the prognosis of glioma patients was evaluated using Univariate and Multivariate Cox analysis and the Kaplan-Meier (KM) curve. We used CIBERSOFT to explore the association between CTF1 and immune cell infiltration in GBM, as well as performing gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA) analyses. Furthermore, we analyzed the relationship between CTF1 and gene mutations and drug sensitivity. Using Weighted gene co-expression network analysis (WGCNA) analysis, we pinpointed the gene set most correlated with CTF1 and conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) gene enrichment analyses to anticipate the pathways that could be influenced by CTF1. Finally, we constructed a nomogram using a multifactorial regression model to further predict patient prognosis.</p><p><strong>Results: </strong>CTF1 expression was significantly elevated in glioma tissues compared to normal tissues in the TCGA dataset (P<0.001) and was associated with poorer survival in both TCGA and CGGA datasets (P<0.001). Receiver operating characteristic (ROC) analysis demonstrated the diagnostic potential of CTF1, with an area under the curve (AUC) of 0.889 [95% confidence interval (CI): 0.803-0.974] in TCGA and 0.664 (95% CI: 0.599-0.729) in CGGA. High CTF1 levels were correlated with advanced glioma grades, and Cox regression analysis identified CTF1 as an independent risk factor. A nomogram incorporating CTF1 levels, isocitrate dehydrogenase 1 (<i>IDH1</i>) mutation status, O6-methylguanine-DNA methyltransferase (<i>MGMT</i>) methylation status, age, and gender were developed and validated to predict 1- and 2-year survival probabilities. In GBM, drug sensitivity analysis revealed significant associations between CTF1 expression and responsiveness to gemcitabine, dasatinib, and other agents. CTF1 expression was also linked to immune infiltration (monocytes, neutrophils, M0 macrophages) and pathways involved in tumor progression, including IL2_STAT5, P53, and IL6_JAK_STAT3 sign","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 12","pages":"6862-6879"},"PeriodicalIF":1.5,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11730202/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012319","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}
Pub Date : 2024-12-31Epub Date: 2024-12-27DOI: 10.21037/tcr-24-1113
Abrar I Aljohani, Ieman A Aljahdali, Ohud A Alsalmi, Meshari A Alsuwat, Abdulaziz A Alsharif, Khalid J Alzahrani, Batool S Alsaleh, Ameen Nadheef, Turki S Alqurashi
Background: Invasive breast cancer (BC) is a highly life-threatening disease affecting women world-wide. While its early identification may benefit the provision of more effective therapies, several BC-associated factors may influence BC patients' therapeutic outcomes. Therefore, identifying novel prognostic and therapeutic targets for invasive BC can help with accurate prognosis and therapy-related decisions. The BRI3 binding protein (BRI3BP) gene was found to be a principal gene in invasive BC cohorts using artificial neural network (ANN) techniques. Thus, this study aimed to evaluate the clinicopathological significance of BRI3BP at the transcriptomic and proteomic levels in invasive BC.
Methods: Two transcriptomic BC cohorts, the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC; n=1,980) and The Cancer Genome Atlas (TCGA; n=854), were used to evaluate BRI3BP expression at the mRNA level. Formalin-fixed paraffin-embedded (FFPE) tissues from an invasive BC cohort (n=100) were also used to evaluate BRI3BP expression at the protein level via immunohistochemistry. The association between BRI3BP expression, clinicopathological characteristics, and patient outcomes was evaluated.
Results: In both METABRIC and TCGA cohorts, high expression of BRI3BP was significantly associated with aggressive tumor features such as high histological grade, large tumor size, and lymph vascular invasion (LVI) positivity. At the protein level, high BRI3BP expression was associated with high histological grade, hormone receptor negativity, high expression of Ki67, and poor outcome.
Conclusions: This study revealed the prognostic significance of BRI3BP in invasive BC patients. Further functional assessment is needed to confirm the biological role of BRI3BP in BC.
{"title":"The clinicopathological significance of BRI3BP in women with invasive breast cancer.","authors":"Abrar I Aljohani, Ieman A Aljahdali, Ohud A Alsalmi, Meshari A Alsuwat, Abdulaziz A Alsharif, Khalid J Alzahrani, Batool S Alsaleh, Ameen Nadheef, Turki S Alqurashi","doi":"10.21037/tcr-24-1113","DOIUrl":"10.21037/tcr-24-1113","url":null,"abstract":"<p><strong>Background: </strong>Invasive breast cancer (BC) is a highly life-threatening disease affecting women world-wide. While its early identification may benefit the provision of more effective therapies, several BC-associated factors may influence BC patients' therapeutic outcomes. Therefore, identifying novel prognostic and therapeutic targets for invasive BC can help with accurate prognosis and therapy-related decisions. The BRI3 binding protein (<i>BRI3BP</i>) gene was found to be a principal gene in invasive BC cohorts using artificial neural network (ANN) techniques. Thus, this study aimed to evaluate the clinicopathological significance of BRI3BP at the transcriptomic and proteomic levels in invasive BC.</p><p><strong>Methods: </strong>Two transcriptomic BC cohorts, the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC; n=1,980) and The Cancer Genome Atlas (TCGA; n=854), were used to evaluate BRI3BP expression at the mRNA level. Formalin-fixed paraffin-embedded (FFPE) tissues from an invasive BC cohort (n=100) were also used to evaluate BRI3BP expression at the protein level via immunohistochemistry. The association between BRI3BP expression, clinicopathological characteristics, and patient outcomes was evaluated.</p><p><strong>Results: </strong>In both METABRIC and TCGA cohorts, high expression of <i>BRI3BP</i> was significantly associated with aggressive tumor features such as high histological grade, large tumor size, and lymph vascular invasion (LVI) positivity. At the protein level, high BRI3BP expression was associated with high histological grade, hormone receptor negativity, high expression of Ki67, and poor outcome.</p><p><strong>Conclusions: </strong>This study revealed the prognostic significance of BRI3BP in invasive BC patients. Further functional assessment is needed to confirm the biological role of BRI3BP in BC.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 12","pages":"6837-6849"},"PeriodicalIF":1.5,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11730447/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143011905","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}
Background: Many cancer cells exhibit aberrant metabolic reprogramming through abnormal mitochondrial respiration. Protein tyrosine phosphatase mitochondrial 1 (PTPMT1) is a protein tyrosine phosphatase localized to the mitochondria and linked to mitochondrial respiration. However, the expression and role of PTPMT1 in regulating the biological characteristics of small cell lung cancer (SCLC) has not yet been explored. The aim of this study was to evaluate the role of PTPMT1 on SCLC cell survival and mitochondrial function.
Methods: SCLC and adjacent normal tissues were obtained from surgery. The expression level of PTPMT1 in the SCLC tissues and cell lines was determined by immunohistochemical staining, western blot, and quantitative real-time polymerase chain reaction (qRT-PCR). PTPMT1 knockdown was induced by lentivirus-mediated short-hairpin RNA (shRNA) transduction and PTPMT1 inhibition (alexidine dihydrochloride). The biological characteristics of the cells were measured by cell counting kit 8 (CCK-8), colony formation assay, and cell migration assay. The mitochondrial function of the cells was measured by 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolylcarbocyanine iodide (JC-1) staining. The H69 cells were treated with alexidine dihydrochloride, after which transcriptome sequencing and an untargeted metabolomic analysis were performed. The transcriptome differentially expressed genes were measured by qRT-PCR.
Results: PTPMT1 was upregulated in the SCLC tissues compared to the adjacent normal tissues. PTPMT1 inhibition by lentiviral shRNA transduction or specific inhibition resulted in significant growth arrest and apoptosis. The transcriptome sequencing analysis revealed that pathways related to the respiration chain and mitochondrial member protein were disrupted. Several mitochondrial metabolism-related genes, such as FGF21, GDF-15, APLN, and MT-DN6, were dysregulated. Further, PTPMT1 inhibition was found to downregulate Glut expression and disturb mitochondrial function.
Conclusions: PTPMT1 was shown to play a critical role in the survival and growth of SCLC cells, and may become a potential therapeutic target.
{"title":"PTPMT1 inhibition induces apoptosis and growth arrest of human SCLC cells by disrupting mitochondrial metabolism.","authors":"Xiang Liu, Yang Sun, Chuancheng Gao, Huiyan Sun, Fang Tian, Fengjun Xiao, Qinqin Xu","doi":"10.21037/tcr-2024-2379","DOIUrl":"https://doi.org/10.21037/tcr-2024-2379","url":null,"abstract":"<p><strong>Background: </strong>Many cancer cells exhibit aberrant metabolic reprogramming through abnormal mitochondrial respiration. Protein tyrosine phosphatase mitochondrial 1 (PTPMT1) is a protein tyrosine phosphatase localized to the mitochondria and linked to mitochondrial respiration. However, the expression and role of PTPMT1 in regulating the biological characteristics of small cell lung cancer (SCLC) has not yet been explored. The aim of this study was to evaluate the role of PTPMT1 on SCLC cell survival and mitochondrial function.</p><p><strong>Methods: </strong>SCLC and adjacent normal tissues were obtained from surgery. The expression level of PTPMT1 in the SCLC tissues and cell lines was determined by immunohistochemical staining, western blot, and quantitative real-time polymerase chain reaction (qRT-PCR). PTPMT1 knockdown was induced by lentivirus-mediated short-hairpin RNA (shRNA) transduction and PTPMT1 inhibition (alexidine dihydrochloride). The biological characteristics of the cells were measured by cell counting kit 8 (CCK-8), colony formation assay, and cell migration assay. The mitochondrial function of the cells was measured by 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolylcarbocyanine iodide (JC-1) staining. The H69 cells were treated with alexidine dihydrochloride, after which transcriptome sequencing and an untargeted metabolomic analysis were performed. The transcriptome differentially expressed genes were measured by qRT-PCR.</p><p><strong>Results: </strong>PTPMT1 was upregulated in the SCLC tissues compared to the adjacent normal tissues. PTPMT1 inhibition by lentiviral shRNA transduction or specific inhibition resulted in significant growth arrest and apoptosis. The transcriptome sequencing analysis revealed that pathways related to the respiration chain and mitochondrial member protein were disrupted. Several mitochondrial metabolism-related genes, such as <i>FGF21</i>, <i>GDF-15</i>, <i>APLN</i>, and <i>MT-DN6</i>, were dysregulated. Further, PTPMT1 inhibition was found to downregulate Glut expression and disturb mitochondrial function.</p><p><strong>Conclusions: </strong>PTPMT1 was shown to play a critical role in the survival and growth of SCLC cells, and may become a potential therapeutic target.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 12","pages":"6956-6969"},"PeriodicalIF":1.5,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11730198/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012320","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}
Pub Date : 2024-12-31Epub Date: 2024-12-17DOI: 10.21037/tcr-24-1116
Jing Dong, Ye Jin, Zhi Zhang, Zhaohuan Yang, Xuemei Zhang
Background: Esophageal cancer (EC) is one of the most common malignant tumors worldwide, which has severely threatened human health. This study aims to evaluate the prognostic factors and predictors of survival in patients diagnosed with advanced lower third esophageal carcinoma (aLEC). Based on the Surveillance, Epidemiology, and End Results (SEER) database, we developed a model (nomogram) to provide accurate and individualized survival prediction for the patients who have lost the opportunity to undergo radical surgery.
Methods: Using SEER database, the basic information and Medicare data of patients diagnosed with aLEC from 2010 to 2015 were collected. The patients were randomly divided into the training and validation set according to an 8:2 ratio. Univariate and multivariate Cox analyses were used to investigate variables significantly correlated with disease-specific survival (DSS). A nomogram was constructed to predict the prognosis of EC patients. We used the area under the curve (AUC) of the receiver operating characteristic (ROC) curve for the evaluation of performance. Furthermore, calibration curves were used to evaluate the accuracy of the model. The clinical utility was also assessed via decision curve analysis (DCA). Differences in clinicopathological characteristics between T1NanyM1 and T2-4NanyM1 stages were evaluated using the Chi-squared test. Cox regression analysis was performed and Kaplan-Meier curves were plotted to evaluate the impact of T-stage, chemotherapy, and radiotherapy on the survival time of EC patients.
Results: Results of multivariate regression analysis demonstrated that histology type, T stage, and chemotherapy were independent prognostic factors for predicting survival time in patients with aLEC. Notably, the constructed nomogram suggested that patients with stage T2 or T3 had a higher survival rate at 6 months, 1 year, and 2 years compared with those with stage T1. DCAs showed that the predictive nomogram was clinically useful. There were fewer patients with stage T1NanyM1 receiving chemotherapy (P=0.004) or radiotherapy (P<0.001) than patients with stage T2-4NanyM1. Moreover, patients with stage T1NanyM1 who underwent chemotherapy had a better prognosis than those who did not [hazard ratio (HR) 3.15, 95% confidence interval (CI): 2.58-3.83; P<0.001]. For patients with stage T1NanyM1, radiotherapy did not improve outcomes (HR 0.98, 95% CI: 0.82-1.17; P=0.80).
Conclusions: A prognostic nomogram integrating three clinicopathological factors was constructed to predict survival in aLEC patients. Chemotherapy improves outcomes of patients with stage T1NanyM1 aLEC.
{"title":"A predictive model for advanced esophageal cancer involving the lower third of the esophagus.","authors":"Jing Dong, Ye Jin, Zhi Zhang, Zhaohuan Yang, Xuemei Zhang","doi":"10.21037/tcr-24-1116","DOIUrl":"https://doi.org/10.21037/tcr-24-1116","url":null,"abstract":"<p><strong>Background: </strong>Esophageal cancer (EC) is one of the most common malignant tumors worldwide, which has severely threatened human health. This study aims to evaluate the prognostic factors and predictors of survival in patients diagnosed with advanced lower third esophageal carcinoma (aLEC). Based on the Surveillance, Epidemiology, and End Results (SEER) database, we developed a model (nomogram) to provide accurate and individualized survival prediction for the patients who have lost the opportunity to undergo radical surgery.</p><p><strong>Methods: </strong>Using SEER database, the basic information and Medicare data of patients diagnosed with aLEC from 2010 to 2015 were collected. The patients were randomly divided into the training and validation set according to an 8:2 ratio. Univariate and multivariate Cox analyses were used to investigate variables significantly correlated with disease-specific survival (DSS). A nomogram was constructed to predict the prognosis of EC patients. We used the area under the curve (AUC) of the receiver operating characteristic (ROC) curve for the evaluation of performance. Furthermore, calibration curves were used to evaluate the accuracy of the model. The clinical utility was also assessed via decision curve analysis (DCA). Differences in clinicopathological characteristics between T1NanyM1 and T2-4NanyM1 stages were evaluated using the Chi-squared test. Cox regression analysis was performed and Kaplan-Meier curves were plotted to evaluate the impact of T-stage, chemotherapy, and radiotherapy on the survival time of EC patients.</p><p><strong>Results: </strong>Results of multivariate regression analysis demonstrated that histology type, T stage, and chemotherapy were independent prognostic factors for predicting survival time in patients with aLEC. Notably, the constructed nomogram suggested that patients with stage T2 or T3 had a higher survival rate at 6 months, 1 year, and 2 years compared with those with stage T1. DCAs showed that the predictive nomogram was clinically useful. There were fewer patients with stage T1NanyM1 receiving chemotherapy (P=0.004) or radiotherapy (P<0.001) than patients with stage T2-4NanyM1. Moreover, patients with stage T1NanyM1 who underwent chemotherapy had a better prognosis than those who did not [hazard ratio (HR) 3.15, 95% confidence interval (CI): 2.58-3.83; P<0.001]. For patients with stage T1NanyM1, radiotherapy did not improve outcomes (HR 0.98, 95% CI: 0.82-1.17; P=0.80).</p><p><strong>Conclusions: </strong>A prognostic nomogram integrating three clinicopathological factors was constructed to predict survival in aLEC patients. Chemotherapy improves outcomes of patients with stage T1NanyM1 aLEC.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 12","pages":"6661-6674"},"PeriodicalIF":1.5,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11730189/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012053","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}
Pub Date : 2024-12-31Epub Date: 2024-12-18DOI: 10.21037/tcr-24-2049
Xin He, Yating Wang, Yanning Zhang, Arvind Sahu, Khaldoun Almhanna, Yan Liu
Background: Chromosome segregation 1 like (CSE1L) overexpression can promote proliferation and migration in cancer. In previous study, we found that CSE1L expression was higher in gastric cancer (GC) tissues compared to normal tissues. However, the biological function and molecular mechanism of CSE1L in GC remains unclear. In this study, we investigate the function of CSE1L in GC biology and its related molecular mechanisms and therapeutic potentials.
Methods: Transcriptome data from public databases were used to assess CSE1L messenger RNA (mRNA) expression levels in GC. A total of 83 pairs of GC surgical samples were evaluated to determine CSE1L protein expression levels. CSE1L was knocked out in MGC-803 and overexpressed in BGC-823 to evaluate its biological function in GC cells. RNA sequencing (RNA-seq) was used to identify the signaling pathways regulated by CSE1L and the underlying molecular mechanisms, and the transcriptome data were validated. Western blotting and immunofluorescence were used to clarify the regulatory effect of CSE1L on phosphorylated focal adhesion kinase (p-FAK).
Results: CSE1L mRNA was increased in patients with GC, and the high expression of CSE1L was associated with poor prognosis in these patients. Protein levels of CSE1L were also increased in GC surgical samples. CSE1L promoted cell proliferation, cell migration, cell invasion, clone formation, and adhesion ability in GC cells. RNA-seq results suggested that CSE1L upregulated the focal adhesion pathway, which was verified at the mRNA level and protein level. Moreover, CSE1L upregulated p-FAK tyrosine 397 [p-FAK (Y397)] and enhanced the efficacy of defactinib. After the knockout of CSE1L, the killing effect of defactinib on GC cells was intensified.
Conclusions: CSE1L is a potential biomarker for evaluating the prognosis of patients with GC. Knockdown of CSE1L can enhance the efficacy of defactinib by inhibiting p-FAK (Y397), which may be a synergistic target of defactinib.
{"title":"<i>CSE1L</i> in enhancing the effect of defactinib on gastric cancer cells via the inhibition of FAK phosphorylation.","authors":"Xin He, Yating Wang, Yanning Zhang, Arvind Sahu, Khaldoun Almhanna, Yan Liu","doi":"10.21037/tcr-24-2049","DOIUrl":"https://doi.org/10.21037/tcr-24-2049","url":null,"abstract":"<p><strong>Background: </strong>Chromosome segregation 1 like (<i>CSE1L</i>) overexpression can promote proliferation and migration in cancer. In previous study, we found that CSE1L expression was higher in gastric cancer (GC) tissues compared to normal tissues. However, the biological function and molecular mechanism of CSE1L in GC remains unclear. In this study, we investigate the function of CSE1L in GC biology and its related molecular mechanisms and therapeutic potentials.</p><p><strong>Methods: </strong>Transcriptome data from public databases were used to assess <i>CSE1L</i> messenger RNA (mRNA) expression levels in GC. A total of 83 pairs of GC surgical samples were evaluated to determine CSE1L protein expression levels. <i>CSE1L</i> was knocked out in MGC-803 and overexpressed in BGC-823 to evaluate its biological function in GC cells. RNA sequencing (RNA-seq) was used to identify the signaling pathways regulated by CSE1L and the underlying molecular mechanisms, and the transcriptome data were validated. Western blotting and immunofluorescence were used to clarify the regulatory effect of CSE1L on phosphorylated focal adhesion kinase (p-FAK).</p><p><strong>Results: </strong><i>CSE1L</i> mRNA was increased in patients with GC, and the high expression of CSE1L was associated with poor prognosis in these patients. Protein levels of CSE1L were also increased in GC surgical samples. CSE1L promoted cell proliferation, cell migration, cell invasion, clone formation, and adhesion ability in GC cells. RNA-seq results suggested that CSE1L upregulated the focal adhesion pathway, which was verified at the mRNA level and protein level. Moreover, CSE1L upregulated p-FAK tyrosine 397 [p-FAK (Y397)] and enhanced the efficacy of defactinib. After the knockout of CSE1L, the killing effect of defactinib on GC cells was intensified.</p><p><strong>Conclusions: </strong>CSE1L is a potential biomarker for evaluating the prognosis of patients with GC. Knockdown of CSE1L can enhance the efficacy of defactinib by inhibiting p-FAK (Y397), which may be a synergistic target of defactinib.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 12","pages":"6905-6918"},"PeriodicalIF":1.5,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11730453/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012081","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}
Pub Date : 2024-12-31Epub Date: 2024-12-24DOI: 10.21037/tcr-24-1053
Changgang Guo, Xiling Jiang, Yinglang Zhang, Guochang Bao
<p><strong>Background: </strong>Bladder urothelial carcinoma (BLCA) is globally recognized as a prevalent malignancy. Its treatment remains challenging due to the extensive morbidity, high mortality rates, and compromised quality of life from postoperative complications and the lack of specific molecular targets. Our aim was to establish a prognostic model to evaluate the prognostic significance, assess immunotherapy responses, and determine drug susceptibility in patients with BLCA.</p><p><strong>Methods: </strong>From The Cancer Genome Atlas (TCGA) datasets, we obtained BLCA clinical details and expression data of immune-related genes. These data were analyzed using R and related packages. Differential expression analysis, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses, weighted gene co-expression network analysis (WGCNA), univariate and multivariate Cox regression analysis, prognostic analysis, model establishment and evaluation, gene set variation analysis (GSVA), immune function and checkpoint analysis, immunotherapy response prediction, and prediction of drug sensitivity were conducted.</p><p><strong>Results: </strong>A total of 11 differentially expressed immune genes (DEIGs) were selected to establish the bladder carcinoma immune-related gene signature for BLCA prognosis prediction. In both the training and testing groups, the high-risk cohort showed a lower overall survival (OS) than the low-risk cohort. The area under the receiver operating characteristic curve (AUC) was 0.712 in the training group and 0.631 in the testing group, highlighting its predictive capacity. In the external validation datasets GSE39281 and IMvigor210, the OS of the high-risk group was significantly lower than that of the low-risk group, with AUC values of 0.609 and 0.563, respectively. Patients in the training group were categorized into low- and high-risk groups based on the bladder carcinoma immune gene signature (BCIGS) median risk score. GSVA showed 21 KEGG pathways positively correlated with model risk scores. The high-risk group presented with elevated stromal score, immune score, ESTIMATE score, and T cell exclusion score level. Conversely, the low-risk group displayed heightened cytotoxic T-lymphocyte antigen 4 (CTLA4) expression, indicative of a better response to immune checkpoint inhibitors (ICIs). Notably, significant disparities were found in immune subtypes, immune-related function, and immune-related survival between the two risk groups. The AUC values of our model are 0.765 and 0.660, respectively, surpassing those of other models, such as the tumor inflammation signature (TIS), tumor immune dysfunction and exclusion (TIDE), and various clinical factors. We also presented a nomogram, with the AUCs for predicting 1-, 2-, and 3-year OS at 0.727, 0.772, and 0.765 respectively, suggesting the signature's robust predictive power. Finally, 20 small molecular compounds were identified, with the TW.37 drug's half maximum
{"title":"Comprehensive analysis of tumor immune-related gene signature for predicting prognosis, immunotherapy, and drug sensitivity in bladder urothelial carcinoma.","authors":"Changgang Guo, Xiling Jiang, Yinglang Zhang, Guochang Bao","doi":"10.21037/tcr-24-1053","DOIUrl":"https://doi.org/10.21037/tcr-24-1053","url":null,"abstract":"<p><strong>Background: </strong>Bladder urothelial carcinoma (BLCA) is globally recognized as a prevalent malignancy. Its treatment remains challenging due to the extensive morbidity, high mortality rates, and compromised quality of life from postoperative complications and the lack of specific molecular targets. Our aim was to establish a prognostic model to evaluate the prognostic significance, assess immunotherapy responses, and determine drug susceptibility in patients with BLCA.</p><p><strong>Methods: </strong>From The Cancer Genome Atlas (TCGA) datasets, we obtained BLCA clinical details and expression data of immune-related genes. These data were analyzed using R and related packages. Differential expression analysis, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses, weighted gene co-expression network analysis (WGCNA), univariate and multivariate Cox regression analysis, prognostic analysis, model establishment and evaluation, gene set variation analysis (GSVA), immune function and checkpoint analysis, immunotherapy response prediction, and prediction of drug sensitivity were conducted.</p><p><strong>Results: </strong>A total of 11 differentially expressed immune genes (DEIGs) were selected to establish the bladder carcinoma immune-related gene signature for BLCA prognosis prediction. In both the training and testing groups, the high-risk cohort showed a lower overall survival (OS) than the low-risk cohort. The area under the receiver operating characteristic curve (AUC) was 0.712 in the training group and 0.631 in the testing group, highlighting its predictive capacity. In the external validation datasets GSE39281 and IMvigor210, the OS of the high-risk group was significantly lower than that of the low-risk group, with AUC values of 0.609 and 0.563, respectively. Patients in the training group were categorized into low- and high-risk groups based on the bladder carcinoma immune gene signature (BCIGS) median risk score. GSVA showed 21 KEGG pathways positively correlated with model risk scores. The high-risk group presented with elevated stromal score, immune score, ESTIMATE score, and T cell exclusion score level. Conversely, the low-risk group displayed heightened cytotoxic T-lymphocyte antigen 4 (CTLA4) expression, indicative of a better response to immune checkpoint inhibitors (ICIs). Notably, significant disparities were found in immune subtypes, immune-related function, and immune-related survival between the two risk groups. The AUC values of our model are 0.765 and 0.660, respectively, surpassing those of other models, such as the tumor inflammation signature (TIS), tumor immune dysfunction and exclusion (TIDE), and various clinical factors. We also presented a nomogram, with the AUCs for predicting 1-, 2-, and 3-year OS at 0.727, 0.772, and 0.765 respectively, suggesting the signature's robust predictive power. Finally, 20 small molecular compounds were identified, with the TW.37 drug's half maximum ","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 12","pages":"6732-6752"},"PeriodicalIF":1.5,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11730456/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012114","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}
Pub Date : 2024-12-31Epub Date: 2024-12-27DOI: 10.21037/tcr-2024-2436
Lei Wang, Guangyu Tian
Dipeptidase 1 (DPEP1), initially identified as a renal membrane enzyme in mature human kidneys, plays a pivotal role in various cellular processes. It facilitates the exchange of materials and signal transduction across cell membranes, contributing significantly to dipeptide hydrolysis, glucose and lipid metabolism, immune inflammation, and ferroptosis, among other cellular functions. Extensive research has delineated the complex role of DPEP1 in oncogenesis and tumor progression, with its influence being context dependent. DPEP1 has been observed to promote oncogenic activities in hepatocellular carcinoma, non-small cell lung cancer, colorectal cancer, and lymphoblastic malignancies and is hypothesized to participate in multiple biological processes, including tumor cell invasion, metastatic spread, cellular signaling pathways, cell-matrix interactions, and evasion of immune surveillance. Conversely, DPEP1 has been identified as a tumor suppressor in pancreatic adenocarcinoma, lobular breast carcinoma, and Wilms tumor. Moreover, the role of DPEP1 in colorectal cancer has been increasingly recognized in recent research. Emerging evidence suggests that DPEP1 substantially augments the metastatic and invasive potential of colorectal cancer cells, facilitates immune evasion, and confers resistance to chemotherapeutic agents. Despite these findings, the precise molecular mechanisms remain to be fully characterized. This systematic review endeavors to elucidate the structural and functional attributes of the DPEP1 protein, with the aim to clarify its regulatory mechanisms and assess its clinical relevance in oncology. Gaining a thorough understanding of the physiological role and molecular underpinnings of DPEP1 is critical to informing the diagnostic, therapeutic, and prognostic paradigms of related pathologies. It is anticipated that these insights will facilitate the discovery of novel therapeutic targets and generate new investigative trajectories, particularly in the clinical management of colorectal cancer.
{"title":"Insight into dipeptidase 1: structure, function, and mechanism in gastrointestinal cancer diseases.","authors":"Lei Wang, Guangyu Tian","doi":"10.21037/tcr-2024-2436","DOIUrl":"https://doi.org/10.21037/tcr-2024-2436","url":null,"abstract":"<p><p>Dipeptidase 1 (DPEP1), initially identified as a renal membrane enzyme in mature human kidneys, plays a pivotal role in various cellular processes. It facilitates the exchange of materials and signal transduction across cell membranes, contributing significantly to dipeptide hydrolysis, glucose and lipid metabolism, immune inflammation, and ferroptosis, among other cellular functions. Extensive research has delineated the complex role of DPEP1 in oncogenesis and tumor progression, with its influence being context dependent. DPEP1 has been observed to promote oncogenic activities in hepatocellular carcinoma, non-small cell lung cancer, colorectal cancer, and lymphoblastic malignancies and is hypothesized to participate in multiple biological processes, including tumor cell invasion, metastatic spread, cellular signaling pathways, cell-matrix interactions, and evasion of immune surveillance. Conversely, DPEP1 has been identified as a tumor suppressor in pancreatic adenocarcinoma, lobular breast carcinoma, and Wilms tumor. Moreover, the role of DPEP1 in colorectal cancer has been increasingly recognized in recent research. Emerging evidence suggests that DPEP1 substantially augments the metastatic and invasive potential of colorectal cancer cells, facilitates immune evasion, and confers resistance to chemotherapeutic agents. Despite these findings, the precise molecular mechanisms remain to be fully characterized. This systematic review endeavors to elucidate the structural and functional attributes of the DPEP1 protein, with the aim to clarify its regulatory mechanisms and assess its clinical relevance in oncology. Gaining a thorough understanding of the physiological role and molecular underpinnings of DPEP1 is critical to informing the diagnostic, therapeutic, and prognostic paradigms of related pathologies. It is anticipated that these insights will facilitate the discovery of novel therapeutic targets and generate new investigative trajectories, particularly in the clinical management of colorectal cancer.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 12","pages":"7015-7025"},"PeriodicalIF":1.5,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11730190/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012306","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}
Background: In the clinic, the primary conventional treatments of advanced non-small cell lung cancer (NSCLC) are surgery, radiation therapy, and chemotherapy. In recent years, immune checkpoint inhibitors (ICIs) have shown promise in optimizing therapeutic benefits when combined with other immunotherapies or standard therapies. However, effective biomarkers for distant metastasis or recurrence have yet to be identified, making it difficult to determine the best therapeutic approaches. The effect of tumor immunotherapy, as well as metastasis and recurrence, are thought to be significantly affected by the tumor immunosuppressive microenvironment. Transcription factor interferon regulatory factor 5 (IRF5) is a critical regulator of the immune response. It has been found to play an important role in malignant tumor transformation, immune regulation, clinical prognosis, and the treatment response. Nevertheless, its precise role in the advancement of NSCLC, including lung adenocarcinoma (LUAD) remains poorly understood. This study sought to investigate the expression of IRF5 in LUAD and its effect on patient prognosis, and examine the biological function of IRF5. Additionally, the study aimed to examine the association between IRF5 expression and immune cell infiltration, as well as its correlation with key immune checkpoint genes relevant to NSCLC.
Methods: LUAD RNA-sequencing data and clinical information were downloaded from The Cancer Genome Atlas (TCGA) and analyzed. A tissue microarray (TMA) analysis was conducted to detect IRF5 expression, and immunofluorescence staining was performed to determine immune infiltration. Bioinformatics and TMA analyses, including a differential expression analysis, prognosis prediction analysis, correlation analysis, immune infiltration analysis, and gene set enrichment analysis (GSEA), were conducted using the TCGA dataset.
Results: The results showed that the expression levels of IRF5 were lower in the LUAD tissues than the normal lung tissues. Patients with high IRF5 expression had longer survival times than those with low IRF5 expression. IRF5 was also found to be correlated with lymph node metastasis. Nine distinct types of immune cells were identified between the groups with high and low IRF5 expression levels. Eight major immune checkpoint genes were found to be upregulated in LUAD patients with high IRF5 expression levels. The enrichment analyses showed that various immune pathways were enriched in the LUAD samples with IRF5, including T cell activation, lymphocyte activation, and T cell receptor activation.
Conclusions: IRF5 expression is closely related to tumor immunity and immunotherapy in LUAD patients. IRF5 may be indicative of prognosis in LUAD patients.
背景:在临床上,晚期非小细胞肺癌(NSCLC)的主要常规治疗方法是手术、放疗和化疗。近年来,免疫检查点抑制剂(ICIs)在与其他免疫疗法或标准疗法联合使用时显示出优化治疗效果的希望。然而,远端转移或复发的有效生物标志物尚未确定,因此很难确定最佳治疗方法。肿瘤免疫治疗的效果,以及肿瘤的转移和复发,被认为受到肿瘤免疫抑制微环境的显著影响。转录因子干扰素调节因子5 (IRF5)是免疫应答的关键调节因子。已发现它在恶性肿瘤转化、免疫调节、临床预后和治疗反应中起重要作用。然而,其在包括肺腺癌(LUAD)在内的非小细胞肺癌(NSCLC)进展中的确切作用仍知之甚少。本研究旨在探讨IRF5在LUAD中的表达及其对患者预后的影响,并探讨IRF5的生物学功能。此外,本研究旨在研究IRF5表达与免疫细胞浸润的关系,以及其与NSCLC相关的关键免疫检查点基因的相关性。方法:从The Cancer Genome Atlas (TCGA)下载LUAD rna测序数据和临床资料进行分析。组织微阵列(TMA)检测IRF5表达,免疫荧光染色检测免疫浸润。使用TCGA数据集进行生物信息学和TMA分析,包括差异表达分析、预后预测分析、相关性分析、免疫浸润分析和基因集富集分析(GSEA)。结果:结果显示,IRF5在LUAD组织中的表达水平低于正常肺组织。IRF5高表达的患者比低表达的患者生存时间更长。IRF5也被发现与淋巴结转移相关。在IRF5高表达组和低表达组之间鉴定出9种不同类型的免疫细胞。8个主要免疫检查点基因在IRF5高表达的LUAD患者中被发现上调。富集分析表明,IRF5在LUAD样品中富集了多种免疫途径,包括T细胞活化、淋巴细胞活化和T细胞受体活化。结论:IRF5表达与LUAD患者的肿瘤免疫及免疫治疗密切相关。IRF5可能指示LUAD患者的预后。
{"title":"IRF5 as a potential immunological biomarker in lung adenocarcinoma.","authors":"Simei Li, Fangling Zhou, Huahong Lan, Xiaofei Cao, Alice Mogenet, Roberto Díaz-Peña, Meiling Zhao","doi":"10.21037/tcr-2024-2354","DOIUrl":"https://doi.org/10.21037/tcr-2024-2354","url":null,"abstract":"<p><strong>Background: </strong>In the clinic, the primary conventional treatments of advanced non-small cell lung cancer (NSCLC) are surgery, radiation therapy, and chemotherapy. In recent years, immune checkpoint inhibitors (ICIs) have shown promise in optimizing therapeutic benefits when combined with other immunotherapies or standard therapies. However, effective biomarkers for distant metastasis or recurrence have yet to be identified, making it difficult to determine the best therapeutic approaches. The effect of tumor immunotherapy, as well as metastasis and recurrence, are thought to be significantly affected by the tumor immunosuppressive microenvironment. Transcription factor interferon regulatory factor 5 (IRF5) is a critical regulator of the immune response. It has been found to play an important role in malignant tumor transformation, immune regulation, clinical prognosis, and the treatment response. Nevertheless, its precise role in the advancement of NSCLC, including lung adenocarcinoma (LUAD) remains poorly understood. This study sought to investigate the expression of IRF5 in LUAD and its effect on patient prognosis, and examine the biological function of IRF5. Additionally, the study aimed to examine the association between IRF5 expression and immune cell infiltration, as well as its correlation with key immune checkpoint genes relevant to NSCLC.</p><p><strong>Methods: </strong>LUAD RNA-sequencing data and clinical information were downloaded from The Cancer Genome Atlas (TCGA) and analyzed. A tissue microarray (TMA) analysis was conducted to detect IRF5 expression, and immunofluorescence staining was performed to determine immune infiltration. Bioinformatics and TMA analyses, including a differential expression analysis, prognosis prediction analysis, correlation analysis, immune infiltration analysis, and gene set enrichment analysis (GSEA), were conducted using the TCGA dataset.</p><p><strong>Results: </strong>The results showed that the expression levels of IRF5 were lower in the LUAD tissues than the normal lung tissues. Patients with high IRF5 expression had longer survival times than those with low IRF5 expression. IRF5 was also found to be correlated with lymph node metastasis. Nine distinct types of immune cells were identified between the groups with high and low IRF5 expression levels. Eight major immune checkpoint genes were found to be upregulated in LUAD patients with high IRF5 expression levels. The enrichment analyses showed that various immune pathways were enriched in the LUAD samples with IRF5, including T cell activation, lymphocyte activation, and T cell receptor activation.</p><p><strong>Conclusions: </strong>IRF5 expression is closely related to tumor immunity and immunotherapy in LUAD patients. IRF5 may be indicative of prognosis in LUAD patients.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 12","pages":"6936-6946"},"PeriodicalIF":1.5,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11729752/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012309","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}
Pub Date : 2024-12-31Epub Date: 2024-12-27DOI: 10.21037/tcr-24-1077
Lei Shi, Jun-Feng Jiang, Jing Zhai
Background: N6-methyladenosine (m6A)-mediated epitranscriptomic pathway has been shown to contribute to chemoresistance and radioresistance. Our previous work confirmed the defense of lycorine against tamoxifen resistance of breast cancer (BC) through targeting HOXD antisense growth-associated long non-coding RNA (HAGLR). Whereas, the precise regulation among them remains to be elucidated. The aim of this study was to investigate the role of IGF2BP2-mediated m6A methylation in the regulation of HAGLR and its impact on lycorine's effect on tamoxifen resistance in BC.
Methods: m6A status was detected via methylated RNA immunoprecipitation-quantitative polymerase chain reaction (MeRIP-qPCR). Relative expression of HAGLR and IGF2BP2 were tested by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blot analysis, respectively. Cell viability, proliferation and apoptosis were estimated via Cell Counting Kit-8 (CCK-8), colony formation and flow cytometer analysis. Interplay among IGF2BP2 and HAGLR was tested by RNA immunoprecipitation (RIP) assay. IC50 value of BC cells to tamoxifen was determined by 2,5-diphenyl-2H-tetrazolium bromide (MTT) assay.
Results: Total m6A level in tamoxifen-resistant BC cells (TAMR/MCF-7 and TAMR/T47D) was elevated relative to corresponding parental cells and normal mammary epithelial cell line, MCF10A, either with the presence of m6A modifications within HAGLR sequence. Moreover, IGF2BP2-mediated m6A methylation drove the upregulation and stability of HAGLR in TAMR BC cells. IGF2BP2 served as a key downstream target mediating the anti-tumors of lycorine on TAMR BC. Knockdown of IGF2BP2 or HAGLR could reduce the IC50 value of TAMR/MCF-7 and TAMR/T47D cells to tamoxifen.
Conclusions: Our results demonstrated that lycorine inhibits tamoxifen-resistant BC by repressing IGF2BP2-mediated m6A methylation of HAGLR.
{"title":"Lycorine affects tamoxifen resistance of breast cancer via m<sup>6</sup>A-based HAGLR.","authors":"Lei Shi, Jun-Feng Jiang, Jing Zhai","doi":"10.21037/tcr-24-1077","DOIUrl":"https://doi.org/10.21037/tcr-24-1077","url":null,"abstract":"<p><strong>Background: </strong>N6-methyladenosine (m<sup>6</sup>A)-mediated epitranscriptomic pathway has been shown to contribute to chemoresistance and radioresistance. Our previous work confirmed the defense of lycorine against tamoxifen resistance of breast cancer (BC) through targeting HOXD antisense growth-associated long non-coding RNA (HAGLR). Whereas, the precise regulation among them remains to be elucidated. The aim of this study was to investigate the role of IGF2BP2-mediated m<sup>6</sup>A methylation in the regulation of HAGLR and its impact on lycorine's effect on tamoxifen resistance in BC.</p><p><strong>Methods: </strong>m<sup>6</sup>A status was detected via methylated RNA immunoprecipitation-quantitative polymerase chain reaction (MeRIP-qPCR). Relative expression of HAGLR and IGF2BP2 were tested by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blot analysis, respectively. Cell viability, proliferation and apoptosis were estimated via Cell Counting Kit-8 (CCK-8), colony formation and flow cytometer analysis. Interplay among IGF2BP2 and HAGLR was tested by RNA immunoprecipitation (RIP) assay. IC<sub>50</sub> value of BC cells to tamoxifen was determined by 2,5-diphenyl-2H-tetrazolium bromide (MTT) assay.</p><p><strong>Results: </strong>Total m<sup>6</sup>A level in tamoxifen-resistant BC cells (TAMR/MCF-7 and TAMR/T47D) was elevated relative to corresponding parental cells and normal mammary epithelial cell line, MCF10A, either with the presence of m<sup>6</sup>A modifications within HAGLR sequence. Moreover, IGF2BP2-mediated m<sup>6</sup>A methylation drove the upregulation and stability of HAGLR in TAMR BC cells. IGF2BP2 served as a key downstream target mediating the anti-tumors of lycorine on TAMR BC. Knockdown of IGF2BP2 or HAGLR could reduce the IC<sub>50</sub> value of TAMR/MCF-7 and TAMR/T47D cells to tamoxifen.</p><p><strong>Conclusions: </strong>Our results demonstrated that lycorine inhibits tamoxifen-resistant BC by repressing IGF2BP2-mediated m<sup>6</sup>A methylation of HAGLR.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 12","pages":"6675-6687"},"PeriodicalIF":1.5,"publicationDate":"2024-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11730692/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143012312","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}