Pub Date : 2024-10-31Epub Date: 2024-10-29DOI: 10.21037/tcr-24-1974
Tingyu Cao, Jinchun Wang
Background: Gliomas are highly aggressive brain tumors with complex metabolic and molecular alterations. The role of glycolysis in glioma progression and its regulation by hypoxia remain poorly understood. This study investigated the function of glycogen phosphorylase L (PYGL) in glioma and its interaction with glycolytic pathways under hypoxic conditions.
Methods: Differential expression analysis was conducted using The Cancer Genome Atlas (TCGA) glioma and GSE67089 datasets, revealing significant changes in the expression of genes. A prognostic risk model incorporating PYGL was built by univariate and multivariate Cox regression analyses. The impacts of PYGL on glioma cell proliferation, glycolysis, apoptosis, and metabolic activities were evaluated by in vitro assays. Additionally, the influences of hypoxia and hypoxia-inducible factor 1-alpha (HIF1α) on PYGL expression were evaluated.
Results: Our prognostic prediction model showed a C-index of 0.76 [95% confidence interval (CI): 0.70-0.82], indicating a good predictive accuracy of the model. In addition, genetic predictors included in the nomogram included PYGL, HIF1α, and other genes associated with the glycolytic pathway. Differential expression analysis identified PYGL as a key gene associated with glioma survival. PYGL expression was significantly upregulated in glioma cells. PYGL knockdown inhibited cell invasion, proliferation, migration, and colony formation and enhanced apoptosis via modulation of Bcl-2, caspase-3, and Bax. Glycolysis was impaired in PYGL-knockdown cells, as indicated by increased glycogen levels and a reduced extracellular acidification rate (ECAR), adenosine triphosphate (ATP) levels, lactate levels, and PKM2 and LDHA expression. PYGL overexpression promoted glycolysis and cell viability, which was counteracted by 2-deoxy-D-glucose (2-DG). Hypoxia-induced PYGL expression was regulated by HIF1α, underscoring the interplay between the hypoxia and glycolysis pathways.
Conclusions: PYGL is a crucial regulator of glycolysis in gliomas and contributes to tumor progression under hypoxic conditions. Targeting PYGL and its associated metabolic pathways may offer new therapeutic strategies for glioma treatment.
背景:胶质瘤是一种侵袭性很强的脑肿瘤,具有复杂的代谢和分子改变。人们对糖酵解在胶质瘤进展中的作用及其受缺氧的调控仍知之甚少。本研究探讨了糖原磷酸化酶 L(PYGL)在胶质瘤中的功能及其在缺氧条件下与糖酵解途径的相互作用:方法:利用癌症基因组图谱(TCGA)胶质瘤数据集和GSE67089数据集进行差异表达分析,结果显示基因表达发生了显著变化。通过单变量和多变量Cox回归分析,建立了包含PYGL的预后风险模型。体外实验评估了PYGL对胶质瘤细胞增殖、糖酵解、凋亡和代谢活动的影响。此外,还评估了缺氧和缺氧诱导因子1-α(HIF1α)对PYGL表达的影响:我们的预后预测模型的 C 指数为 0.76 [95% 置信区间 (CI):0.70-0.82],表明该模型具有良好的预测准确性。此外,提名图中的遗传预测因子还包括PYGL、HIF1α和其他与糖酵解途径相关的基因。差异表达分析发现PYGL是与胶质瘤存活率相关的关键基因。PYGL在胶质瘤细胞中的表达明显上调。PYGL基因敲除抑制了细胞的侵袭、增殖、迁移和集落形成,并通过调节Bcl-2、caspase-3和Bax增强了细胞凋亡。PYGL敲除细胞的糖酵解功能受损,表现为糖原水平升高,细胞外酸化率(ECAR)、三磷酸腺苷(ATP)水平、乳酸水平以及PKM2和LDHA表达降低。PYGL过表达可促进糖酵解和细胞活力,而2-脱氧-D-葡萄糖(2-DG)可抵消这种作用。缺氧诱导的PYGL表达受HIF1α调控,强调了缺氧和糖酵解途径之间的相互作用:结论:PYGL是胶质瘤中糖酵解的关键调节因子,有助于缺氧条件下的肿瘤进展。以PYGL及其相关代谢途径为靶点可为胶质瘤治疗提供新的治疗策略。
{"title":"<i>PYGL</i> regulation of glycolysis and apoptosis in glioma cells under hypoxic conditions via HIF1α-dependent mechanisms.","authors":"Tingyu Cao, Jinchun Wang","doi":"10.21037/tcr-24-1974","DOIUrl":"https://doi.org/10.21037/tcr-24-1974","url":null,"abstract":"<p><strong>Background: </strong>Gliomas are highly aggressive brain tumors with complex metabolic and molecular alterations. The role of glycolysis in glioma progression and its regulation by hypoxia remain poorly understood. This study investigated the function of glycogen phosphorylase L (<i>PYGL</i>) in glioma and its interaction with glycolytic pathways under hypoxic conditions.</p><p><strong>Methods: </strong>Differential expression analysis was conducted using The Cancer Genome Atlas (TCGA) glioma and GSE67089 datasets, revealing significant changes in the expression of genes. A prognostic risk model incorporating <i>PYGL</i> was built by univariate and multivariate Cox regression analyses. The impacts of <i>PYGL</i> on glioma cell proliferation, glycolysis, apoptosis, and metabolic activities were evaluated by <i>in vitro</i> assays. Additionally, the influences of hypoxia and hypoxia-inducible factor 1-alpha (<i>HIF1α</i>) on <i>PYGL</i> expression were evaluated.</p><p><strong>Results: </strong>Our prognostic prediction model showed a C-index of 0.76 [95% confidence interval (CI): 0.70-0.82], indicating a good predictive accuracy of the model. In addition, genetic predictors included in the nomogram included PYGL, HIF1α, and other genes associated with the glycolytic pathway. Differential expression analysis identified <i>PYGL</i> as a key gene associated with glioma survival. <i>PYGL</i> expression was significantly upregulated in glioma cells. <i>PYGL</i> knockdown inhibited cell invasion, proliferation, migration, and colony formation and enhanced apoptosis via modulation of Bcl-2, caspase-3, and Bax. Glycolysis was impaired in <i>PYGL</i>-knockdown cells, as indicated by increased glycogen levels and a reduced extracellular acidification rate (ECAR), adenosine triphosphate (ATP) levels, lactate levels, and PKM2 and LDHA expression. <i>PYGL</i> overexpression promoted glycolysis and cell viability, which was counteracted by 2-deoxy-D-glucose (2-DG). Hypoxia-induced <i>PYGL</i> expression was regulated by <i>HIF1α</i>, underscoring the interplay between the hypoxia and glycolysis pathways.</p><p><strong>Conclusions: </strong><i>PYGL</i> is a crucial regulator of glycolysis in gliomas and contributes to tumor progression under hypoxic conditions. Targeting <i>PYGL</i> and its associated metabolic pathways may offer new therapeutic strategies for glioma treatment.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 10","pages":"5627-5648"},"PeriodicalIF":1.5,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11543057/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142628838","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 : 2024-10-31Epub Date: 2024-10-29DOI: 10.21037/tcr-24-1947
Yan Gao, Meng Wu, Syed A A Rizvi, Qiang Wei
<p><strong>Background: </strong>Lung cancer often metastasizes to the bone, which significantly complicates treatment and worsens patient prognosis. Thus, new therapeutic strategies need to be established. Using network pharmacology and bioinformatics analysis, this study sought to determine the molecular targets and associated mechanisms of the traditional Chinese medicine (TCM) <i>Sophorae Flavescentis radix</i> in the treatment of lung cancer bone metastasis.</p><p><strong>Methods: </strong>The active components of <i>Sophorae Flavescentis radix</i> were screened using the TCM Systems Pharmacology (TCMSP) platform based on drug-likeness and oral bioavailability. The target genes of these active compounds were obtained from the DrugBank database. Differentially expressed genes (DEGs) between primary and bone metastatic lung cancer samples were screened in the GSE175601 dataset from the Gene Expression Omnibus (GEO) database using GEO2R. The intersecting DEGs from both groups were used to construct a Venn diagram to identify the candidate target genes. The expression and prognostic relevance of these genes were validated in The Cancer Genome Atlas (TCGA) database. The GeneMania and Search Tool for Recurring Instances of Neighbouring Genes (STRING) databases were used to generate the protein-protein interaction networks. Molecular docking was performed using the PubChem, Protein Data Bank (PDB), and CB-DOCK2 databases. A Gene Set Enrichment Analysis (GSEA) was conducted to explore the possible mechanisms of action.</p><p><strong>Results: </strong>In the TCMSP database, 28 active compounds and 227 target genes of the <i>Sophorae Flavescentis radix</i> were identified. In total, 952 DEGs related to lung cancer bone metastasis were found in the GSE175601 dataset from the GEO database. Five common DEGs were identified via Venn diagram construction (i.e., <i>F10, JUN, AKR1B1, MMP1,</i> and <i>CCND1</i>). <i>MMP1</i> was selected as the candidate gene. <i>MMP1</i> was upregulated in lung cancer tissues, and patients with low <i>MMP1</i> expression had better survival rates than those with high <i>MMP1</i> expression (P<0.05). <i>MMP1</i> has an affinity of -8.9 with luteolin. The GSEA results suggested that <i>MMP1</i> might influence biological processes in lung cancer by participating in pathways such as chemokine signaling, apoptosis, Wingless/Integrated (Wnt) signaling, tumor protein p53-regulated cell cycle arrest, Hedgehog signaling, and mitogen-activated protein kinase signaling.</p><p><strong>Conclusions: </strong>Patients with lower <i>MMP1</i> levels had prolonged overall survival and may serve as a novel predictive biomarker for lung cancer. <i>Sophorae Flavescentis radix</i> appears to exert therapeutic effects on lung cancer bone metastasis by inhibiting <i>MMP1</i> expression and modulating the abnormal activation of the Wnt pathway. Our findings further extend the understanding of the pathogenic mechanisms and potential therapeutic inter
{"title":"Exploring the key pathogenic mechanisms and potential intervention targets for <i>Sophorae Flavescentis radix</i> in managing bone metastasis of lung cancer based on network pharmacology and molecular docking techniques.","authors":"Yan Gao, Meng Wu, Syed A A Rizvi, Qiang Wei","doi":"10.21037/tcr-24-1947","DOIUrl":"https://doi.org/10.21037/tcr-24-1947","url":null,"abstract":"<p><strong>Background: </strong>Lung cancer often metastasizes to the bone, which significantly complicates treatment and worsens patient prognosis. Thus, new therapeutic strategies need to be established. Using network pharmacology and bioinformatics analysis, this study sought to determine the molecular targets and associated mechanisms of the traditional Chinese medicine (TCM) <i>Sophorae Flavescentis radix</i> in the treatment of lung cancer bone metastasis.</p><p><strong>Methods: </strong>The active components of <i>Sophorae Flavescentis radix</i> were screened using the TCM Systems Pharmacology (TCMSP) platform based on drug-likeness and oral bioavailability. The target genes of these active compounds were obtained from the DrugBank database. Differentially expressed genes (DEGs) between primary and bone metastatic lung cancer samples were screened in the GSE175601 dataset from the Gene Expression Omnibus (GEO) database using GEO2R. The intersecting DEGs from both groups were used to construct a Venn diagram to identify the candidate target genes. The expression and prognostic relevance of these genes were validated in The Cancer Genome Atlas (TCGA) database. The GeneMania and Search Tool for Recurring Instances of Neighbouring Genes (STRING) databases were used to generate the protein-protein interaction networks. Molecular docking was performed using the PubChem, Protein Data Bank (PDB), and CB-DOCK2 databases. A Gene Set Enrichment Analysis (GSEA) was conducted to explore the possible mechanisms of action.</p><p><strong>Results: </strong>In the TCMSP database, 28 active compounds and 227 target genes of the <i>Sophorae Flavescentis radix</i> were identified. In total, 952 DEGs related to lung cancer bone metastasis were found in the GSE175601 dataset from the GEO database. Five common DEGs were identified via Venn diagram construction (i.e., <i>F10, JUN, AKR1B1, MMP1,</i> and <i>CCND1</i>). <i>MMP1</i> was selected as the candidate gene. <i>MMP1</i> was upregulated in lung cancer tissues, and patients with low <i>MMP1</i> expression had better survival rates than those with high <i>MMP1</i> expression (P<0.05). <i>MMP1</i> has an affinity of -8.9 with luteolin. The GSEA results suggested that <i>MMP1</i> might influence biological processes in lung cancer by participating in pathways such as chemokine signaling, apoptosis, Wingless/Integrated (Wnt) signaling, tumor protein p53-regulated cell cycle arrest, Hedgehog signaling, and mitogen-activated protein kinase signaling.</p><p><strong>Conclusions: </strong>Patients with lower <i>MMP1</i> levels had prolonged overall survival and may serve as a novel predictive biomarker for lung cancer. <i>Sophorae Flavescentis radix</i> appears to exert therapeutic effects on lung cancer bone metastasis by inhibiting <i>MMP1</i> expression and modulating the abnormal activation of the Wnt pathway. Our findings further extend the understanding of the pathogenic mechanisms and potential therapeutic inter","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 10","pages":"5616-5626"},"PeriodicalIF":1.5,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11543056/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142628033","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 : 2024-10-31Epub Date: 2024-10-28DOI: 10.21037/tcr-24-692
Tong-Dan Lyu, Ming-Peng Luo, Hao-Wei Hu
Background: With the advancement of various auxiliary examination techniques, the detection rate of stage I gastric cancer has gradually increased, and its clinical first-choice treatment is surgery. Although patients with stage I gastric cancer generally have a good postoperative survival rate, there is still a certain probability of recurrence. Given the large number of gastric cancer cases, there is a vast population of patients with stage I disease. We are aiming to identify the risk factors for postoperative recurrence of stage I gastric cancer and to establish a reliable predictive model to assess the risk of recurrence in the population for clinical practice.
Methods: In this retrospective cohort study, we utilized the Surveillance, Epidemiology, and End Results (SEER) database to investigate predictive factors for recurrence among stage I gastric cancer patients who underwent curative gastrectomy between 2000 and 2018. The cohort was divided into training and validation sets for the development and validation of a nomogram. Prognostic factors were evaluated through univariate and multivariate Cox regression analyses. Significant variables identified by the concordance index (C-index) and calibration plots were used to construct nomograms predicting the probability of 5- and 10-year recurrence.
Results: Risk factors for recurrence included sex, age, race, histology, tumor size, American Joint Committee on Cancer Tumor (AJCC T) and primary site, which were used to construct the nomogram. The C-index for both the training and validation cohorts indicated that the nomogram possessed good calibration and discrimination abilities in predicting the probability of 5- and 10-year recurrence after curative surgery for stage I gastric cancer.
Conclusions: This study established a reliable predictive model for recurrence following curative gastrectomy in stage I gastric cancer based on a population cohort. The findings of this study have the potential to significantly impact clinical practice by providing clinicians with tools for personalized risk assessment and for making informed treatment decisions.
背景:随着各种辅助检查技术的进步,I 期胃癌的检出率逐渐提高,临床上首选的治疗方法是手术治疗。虽然 I 期胃癌患者的术后生存率普遍较高,但仍有一定的复发概率。由于胃癌病例数量庞大,I 期胃癌患者群体也十分庞大。我们的目的是找出 I 期胃癌术后复发的风险因素,并建立一个可靠的预测模型来评估人群的复发风险,以利于临床实践:在这项回顾性队列研究中,我们利用监测、流行病学和最终结果(SEER)数据库调查了2000年至2018年期间接受治愈性胃切除术的I期胃癌患者的复发预测因素。队列被分为训练集和验证集,以开发和验证提名图。通过单变量和多变量 Cox 回归分析评估了预后因素。通过一致性指数(C-index)和校准图确定的重要变量被用于构建预测5年和10年复发概率的提名图:复发风险因素包括性别、年龄、种族、组织学、肿瘤大小、美国癌症联合委员会肿瘤(AJCC T)和原发部位,这些因素被用于构建提名图。训练队列和验证队列的 C 指数表明,提名图在预测 I 期胃癌根治术后 5 年和 10 年复发概率方面具有良好的校准和区分能力:本研究基于人群队列建立了一个可靠的 I 期胃癌根治性胃切除术后复发预测模型。这项研究的结果为临床医生提供了个性化风险评估和做出明智治疗决策的工具,从而有可能对临床实践产生重大影响。
{"title":"Nomogram for predicting 10-year postoperative recurrence of stage I gastric cancer.","authors":"Tong-Dan Lyu, Ming-Peng Luo, Hao-Wei Hu","doi":"10.21037/tcr-24-692","DOIUrl":"https://doi.org/10.21037/tcr-24-692","url":null,"abstract":"<p><strong>Background: </strong>With the advancement of various auxiliary examination techniques, the detection rate of stage I gastric cancer has gradually increased, and its clinical first-choice treatment is surgery. Although patients with stage I gastric cancer generally have a good postoperative survival rate, there is still a certain probability of recurrence. Given the large number of gastric cancer cases, there is a vast population of patients with stage I disease. We are aiming to identify the risk factors for postoperative recurrence of stage I gastric cancer and to establish a reliable predictive model to assess the risk of recurrence in the population for clinical practice.</p><p><strong>Methods: </strong>In this retrospective cohort study, we utilized the Surveillance, Epidemiology, and End Results (SEER) database to investigate predictive factors for recurrence among stage I gastric cancer patients who underwent curative gastrectomy between 2000 and 2018. The cohort was divided into training and validation sets for the development and validation of a nomogram. Prognostic factors were evaluated through univariate and multivariate Cox regression analyses. Significant variables identified by the concordance index (C-index) and calibration plots were used to construct nomograms predicting the probability of 5- and 10-year recurrence.</p><p><strong>Results: </strong>Risk factors for recurrence included sex, age, race, histology, tumor size, American Joint Committee on Cancer Tumor (AJCC T) and primary site, which were used to construct the nomogram. The C-index for both the training and validation cohorts indicated that the nomogram possessed good calibration and discrimination abilities in predicting the probability of 5- and 10-year recurrence after curative surgery for stage I gastric cancer.</p><p><strong>Conclusions: </strong>This study established a reliable predictive model for recurrence following curative gastrectomy in stage I gastric cancer based on a population cohort. The findings of this study have the potential to significantly impact clinical practice by providing clinicians with tools for personalized risk assessment and for making informed treatment decisions.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 10","pages":"5497-5508"},"PeriodicalIF":1.5,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11543093/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142628539","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 : 2024-10-31Epub Date: 2024-10-18DOI: 10.21037/tcr-24-378
Tao Chen, Qiusheng Jiang, Zhenlin Wang, Fang Wang, Zan Fu
<p><strong>Background: </strong>Colorectal cancer (CRC) is one of the most common malignant tumors in the digestive system with a high incidence, a poor prognosis and an unsatisfactory therapeutic effect. Long non-coding RNAs (lncRNAs) play crucial roles in various biological processes related to tumor progression. Immune-related lncRNA gene <i>AF117829.1</i> has been reported to participate in the construction of clinical predictive signature in CRC patients, suggesting that it may be involved in regulating the immune landscape and progression of CRC. However, the clinical and immunological significance and biological function of <i>AF117829.1</i> in CRC remain unclear. In this study, we aim to explore the roles of <i>AF117829.1</i> in CRC progression by bioinformatics analysis and experimental studies, thereby providing new targets for CRC treatment.</p><p><strong>Methods: </strong>This study collected data from The Cancer Genome Atlas (TCGA) database and explored the role of <i>AF117829.1</i> in CRC by bioinformatics analysis. Cell-type Identification By Estimating Relative Subsets Of known RNA Transcripts (CIBERSORT) and Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE) methods estimated the immune infiltration. Gene Set Enrichment Analysis (GSEA) was performed to evaluate the enrichment of functional pathways and gene signatures. The biological functions and mechanism of <i>AF117829.1</i> in CRC progression were validated using CRC tissues and <i>in vitro</i> experiments.</p><p><strong>Results: </strong>In our study, high expression of <i>AF117829.1</i> was found in pan-cancer including CRC and was positively associated with tumor (T) stage and tumor-node-metastasis (TNM) stage in CRC. The survival analysis results showed that CRC patients with high-<i>AF117829.1</i> expression had significantly shorter overall survival (OS) time than those with low-<i>AF117829.1</i> expression. Moreover, <i>AF117829.1</i> expression was negatively associated with microsatellite instability (MSI) in colon adenocarcinoma (COAD). Subsequently, <i>AF117829.1</i> expression was confirmed to be significantly associated with StromalScore, immune cell infiltration (ICI) levels and immune checkpoints (ICP) genes expression in CRC. The immunophenoscore (IPS) results indicated that immunotherapy could be more effective in CRC patients with low-<i>AF117829.1</i> expression. Then we confirmed that <i>AF117829.1</i> was highly expressed in CRC cell lines and tissues. Furthermore, our GSEA results showed that olfactory transduction-related signaling pathways were significantly enriched in the high-<i>AF117829.1</i> expression group. Finally, <i>in vitro</i> experiments confirmed that <i>AF117829.1</i> overexpression promoted the proliferation, migration and invasion of CRC cells by targeting olfactory receptor family 7 subfamily C member 1 (<i>OR7C1</i>).</p><p><strong>Conclusions: </strong>LncRNA <i>AF117829.1</i> is close
{"title":"LncRNA <i>AF117829.1</i> is correlated with prognosis and immune infiltration and facilitates tumor progression by targeting <i>OR7C1</i> in colorectal cancer.","authors":"Tao Chen, Qiusheng Jiang, Zhenlin Wang, Fang Wang, Zan Fu","doi":"10.21037/tcr-24-378","DOIUrl":"https://doi.org/10.21037/tcr-24-378","url":null,"abstract":"<p><strong>Background: </strong>Colorectal cancer (CRC) is one of the most common malignant tumors in the digestive system with a high incidence, a poor prognosis and an unsatisfactory therapeutic effect. Long non-coding RNAs (lncRNAs) play crucial roles in various biological processes related to tumor progression. Immune-related lncRNA gene <i>AF117829.1</i> has been reported to participate in the construction of clinical predictive signature in CRC patients, suggesting that it may be involved in regulating the immune landscape and progression of CRC. However, the clinical and immunological significance and biological function of <i>AF117829.1</i> in CRC remain unclear. In this study, we aim to explore the roles of <i>AF117829.1</i> in CRC progression by bioinformatics analysis and experimental studies, thereby providing new targets for CRC treatment.</p><p><strong>Methods: </strong>This study collected data from The Cancer Genome Atlas (TCGA) database and explored the role of <i>AF117829.1</i> in CRC by bioinformatics analysis. Cell-type Identification By Estimating Relative Subsets Of known RNA Transcripts (CIBERSORT) and Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE) methods estimated the immune infiltration. Gene Set Enrichment Analysis (GSEA) was performed to evaluate the enrichment of functional pathways and gene signatures. The biological functions and mechanism of <i>AF117829.1</i> in CRC progression were validated using CRC tissues and <i>in vitro</i> experiments.</p><p><strong>Results: </strong>In our study, high expression of <i>AF117829.1</i> was found in pan-cancer including CRC and was positively associated with tumor (T) stage and tumor-node-metastasis (TNM) stage in CRC. The survival analysis results showed that CRC patients with high-<i>AF117829.1</i> expression had significantly shorter overall survival (OS) time than those with low-<i>AF117829.1</i> expression. Moreover, <i>AF117829.1</i> expression was negatively associated with microsatellite instability (MSI) in colon adenocarcinoma (COAD). Subsequently, <i>AF117829.1</i> expression was confirmed to be significantly associated with StromalScore, immune cell infiltration (ICI) levels and immune checkpoints (ICP) genes expression in CRC. The immunophenoscore (IPS) results indicated that immunotherapy could be more effective in CRC patients with low-<i>AF117829.1</i> expression. Then we confirmed that <i>AF117829.1</i> was highly expressed in CRC cell lines and tissues. Furthermore, our GSEA results showed that olfactory transduction-related signaling pathways were significantly enriched in the high-<i>AF117829.1</i> expression group. Finally, <i>in vitro</i> experiments confirmed that <i>AF117829.1</i> overexpression promoted the proliferation, migration and invasion of CRC cells by targeting olfactory receptor family 7 subfamily C member 1 (<i>OR7C1</i>).</p><p><strong>Conclusions: </strong>LncRNA <i>AF117829.1</i> is close","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 10","pages":"5347-5364"},"PeriodicalIF":1.5,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11543043/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142628534","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 : 2024-10-31Epub Date: 2024-10-18DOI: 10.21037/tcr-24-543
Zhao Guo, Jing-Jie Tian, Yao Wang, Lei Jiang, Yang Chen, He-Jun Dai, Lei Wang, Yi Zhang
Background: Glioma characterized by the high degree of drug resistance and the poor prognosis is the most common primary malignant tumors of the brain. And miRNA is involved in a variety of biological behaviors of tumors, enhancing or inhibiting the occurrence and development of tumors. Therefore, the present study aims to explore whether miR-155-5p can regulate autophagy and apoptosis of glioma through RICTOR.
Methods: The significantly differential gene miR-155-5p was identified from the Gene Expression Omnibus (GEO; http://www.ncbi.nlm.nih.gov/geo) databases GSE165937 and GSE138764 using bioinformatics analysis, and its expression was validated by quantitative real-time polymerase chain reaction (qRT-PCR). The putative target genes of miR-155-5p were predicted through interrogation of relevant databases, followed by identification of key target genes. Subsequently, core target genes were selected for functional enrichment analysis. The U87MG cell line was utilized as the experimental model and divided into Negative Control1 (NC1) group, Mimic group, Negative Control2 (NC2) group, Inhibitor group, and NC + 3-methyladenine (3-MA) group. The expression levels of miR-155-5p, RICTOR, P62, LC-3, Bax, Bcl-2, and Caspase-3 were assessed using qRT-PCR, cellular fluorescence imaging, and Western blotting; while apoptosis in the U87MG cell line was evaluated via flow cytometry.
Results: The results showed that miR-155-5P was highly expressed in glioma cells, which could inhibit the expression of Bax, Caspase-3, LCII/LCI and Beclin-1, and increase the expression of Bcl2 and P62. Flow cytometry and cell fluorescence were used to verify the above results. Moreover, when U87MG cells treated with miR-155-5p inhibitor were inhibited by 3-MA, the results showed that miR-155-5p enhanced the anti-apoptotic ability of U87MG cells by regulating autophagy. In addition, the bioinformatics results show that miR-155-5p survival prognosis in glioma into a strong negative correlation, while the survival prognosis of RICTOR in glioma showed a strong positive correlation. The core target genes Kyoto Encyclopedia of Genes and Genomes (KEGG) mainly occurred in PI3K-AKT signaling pathway; in addition, qRT-PCR and Western blot confirmed the regulatory effect of miR-155-5P on RICTOR.
Conclusions: MiR-155-5p regulates autophagy and apoptosis-related proteins in glioma cells through RICTOR, affecting the occurrence and development of glioma.
{"title":"MiR-155-5p regulates autophagy and apoptosis of glioma cells through RICTOR.","authors":"Zhao Guo, Jing-Jie Tian, Yao Wang, Lei Jiang, Yang Chen, He-Jun Dai, Lei Wang, Yi Zhang","doi":"10.21037/tcr-24-543","DOIUrl":"https://doi.org/10.21037/tcr-24-543","url":null,"abstract":"<p><strong>Background: </strong>Glioma characterized by the high degree of drug resistance and the poor prognosis is the most common primary malignant tumors of the brain. And miRNA is involved in a variety of biological behaviors of tumors, enhancing or inhibiting the occurrence and development of tumors. Therefore, the present study aims to explore whether miR-155-5p can regulate autophagy and apoptosis of glioma through RICTOR.</p><p><strong>Methods: </strong>The significantly differential gene miR-155-5p was identified from the Gene Expression Omnibus (GEO; http://www.ncbi.nlm.nih.gov/geo) databases GSE165937 and GSE138764 using bioinformatics analysis, and its expression was validated by quantitative real-time polymerase chain reaction (qRT-PCR). The putative target genes of miR-155-5p were predicted through interrogation of relevant databases, followed by identification of key target genes. Subsequently, core target genes were selected for functional enrichment analysis. The U87MG cell line was utilized as the experimental model and divided into Negative Control1 (NC1) group, Mimic group, Negative Control2 (NC2) group, Inhibitor group, and NC + 3-methyladenine (3-MA) group. The expression levels of miR-155-5p, RICTOR, P62, LC-3, Bax, Bcl-2, and Caspase-3 were assessed using qRT-PCR, cellular fluorescence imaging, and Western blotting; while apoptosis in the U87MG cell line was evaluated via flow cytometry.</p><p><strong>Results: </strong>The results showed that miR-155-5P was highly expressed in glioma cells, which could inhibit the expression of Bax, Caspase-3, LCII/LCI and Beclin-1, and increase the expression of Bcl2 and P62. Flow cytometry and cell fluorescence were used to verify the above results. Moreover, when U87MG cells treated with miR-155-5p inhibitor were inhibited by 3-MA, the results showed that miR-155-5p enhanced the anti-apoptotic ability of U87MG cells by regulating autophagy. In addition, the bioinformatics results show that miR-155-5p survival prognosis in glioma into a strong negative correlation, while the survival prognosis of RICTOR in glioma showed a strong positive correlation. The core target genes Kyoto Encyclopedia of Genes and Genomes (KEGG) mainly occurred in PI3K-AKT signaling pathway; in addition, qRT-PCR and Western blot confirmed the regulatory effect of miR-155-5P on RICTOR.</p><p><strong>Conclusions: </strong>MiR-155-5p regulates autophagy and apoptosis-related proteins in glioma cells through RICTOR, affecting the occurrence and development of glioma.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 10","pages":"5509-5521"},"PeriodicalIF":1.5,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11543026/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142628535","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 : 2024-10-31Epub Date: 2024-10-18DOI: 10.21037/tcr-24-679
Run Qu, Yanhong Zhao, Yuzhe Zhang
Background and objective: The occurrence and development of tumors in human tissues widely depend on their surrounding environment, known as the tumor microenvironment (TME), which comprises various cells, molecules, and blood vessels. Through modifications, organization, and integration, these elements serve as potential therapeutic targets in anti-cancer therapy, supporting and promoting the proliferation, invasion, and metabolism of tumor cells. Cytokines within TME are responsible for immune cell activation, proliferation, and differentiation, thereby influencing the tumor's behavior. This article reviews the use of cytokines in tumor immunotherapy and combs the network signals that cytokines mediate in the development of malignancies.
Methods: A literature search of international sources was carried out on the PubMed and Web of Science databases, using main keywords such as "tumor immunotherapy", "cytokines", "chemokines", "tumor microenvironment", "recombinant cytokine engineering", and "tumor necrosis factor superfamily".
Key content and findings: The review provides a thorough summary of the functions of tumor necrosis factor superfamilies, chemokines, and interleukins within the TME as well as their therapeutic uses. Their potential as novel targets for tumor treatment is also evaluated. Furthermore, this paper focuses on various feasible strategies for recombinant cytokines reported in recent years, especially the cytokine engineering methods for targeting tumors. Ultimately, this paper contributes to an enhanced understanding among researchers of the mechanisms underlying the impact of the TME on disease development, thereby laying a solid foundation for the future development of new tumor therapies based on cytokines within the TME.
Conclusions: Cytokine immunotherapy holds promise on antitumor therapy. It is anticipated that the effectiveness of tumor treatment and the quality of life for tumor patients will continue to improve with ongoing research and development in this field.
背景和目的:人体组织中肿瘤的发生和发展广泛依赖于其周围环境,即肿瘤微环境(TME),它由各种细胞、分子和血管组成。通过修饰、组织和整合,这些元素成为抗癌疗法的潜在治疗靶点,支持和促进肿瘤细胞的增殖、侵袭和新陈代谢。TME中的细胞因子负责免疫细胞的活化、增殖和分化,从而影响肿瘤的行为。本文回顾了细胞因子在肿瘤免疫疗法中的应用,并梳理了细胞因子在恶性肿瘤发展过程中介导的网络信号:方法:使用 "肿瘤免疫疗法"、"细胞因子"、"趋化因子"、"肿瘤微环境"、"重组细胞因子工程 "和 "肿瘤坏死因子超家族 "等主要关键词,在 PubMed 和 Web of Science 数据库中进行了国际文献检索:综述全面总结了肿瘤坏死因子超家族、趋化因子和白细胞介素在肿瘤微环境中的功能及其治疗用途。本文还评估了它们作为肿瘤治疗新靶点的潜力。此外,本文还重点介绍了近年来报道的重组细胞因子的各种可行策略,尤其是针对肿瘤的细胞因子工程方法。最终,本文有助于加深研究人员对肿瘤组织器官对疾病发展的影响机制的理解,从而为未来开发基于肿瘤组织器官内细胞因子的新型肿瘤疗法奠定坚实的基础:细胞因子免疫疗法为抗肿瘤治疗带来了希望。预计随着这一领域研究和开发的不断深入,肿瘤治疗的有效性和肿瘤患者的生活质量将不断提高。
{"title":"The mechanism of cytokine regulation of cancer occurrence and development in the tumor microenvironment and its application in cancer treatment: a narrative review.","authors":"Run Qu, Yanhong Zhao, Yuzhe Zhang","doi":"10.21037/tcr-24-679","DOIUrl":"https://doi.org/10.21037/tcr-24-679","url":null,"abstract":"<p><strong>Background and objective: </strong>The occurrence and development of tumors in human tissues widely depend on their surrounding environment, known as the tumor microenvironment (TME), which comprises various cells, molecules, and blood vessels. Through modifications, organization, and integration, these elements serve as potential therapeutic targets in anti-cancer therapy, supporting and promoting the proliferation, invasion, and metabolism of tumor cells. Cytokines within TME are responsible for immune cell activation, proliferation, and differentiation, thereby influencing the tumor's behavior. This article reviews the use of cytokines in tumor immunotherapy and combs the network signals that cytokines mediate in the development of malignancies.</p><p><strong>Methods: </strong>A literature search of international sources was carried out on the PubMed and Web of Science databases, using main keywords such as \"tumor immunotherapy\", \"cytokines\", \"chemokines\", \"tumor microenvironment\", \"recombinant cytokine engineering\", and \"tumor necrosis factor superfamily\".</p><p><strong>Key content and findings: </strong>The review provides a thorough summary of the functions of tumor necrosis factor superfamilies, chemokines, and interleukins within the TME as well as their therapeutic uses. Their potential as novel targets for tumor treatment is also evaluated. Furthermore, this paper focuses on various feasible strategies for recombinant cytokines reported in recent years, especially the cytokine engineering methods for targeting tumors. Ultimately, this paper contributes to an enhanced understanding among researchers of the mechanisms underlying the impact of the TME on disease development, thereby laying a solid foundation for the future development of new tumor therapies based on cytokines within the TME.</p><p><strong>Conclusions: </strong>Cytokine immunotherapy holds promise on antitumor therapy. It is anticipated that the effectiveness of tumor treatment and the quality of life for tumor patients will continue to improve with ongoing research and development in this field.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 10","pages":"5649-5663"},"PeriodicalIF":1.5,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11543031/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142628764","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 and objective: The bone marrow microenvironment is closely related to normal hematopoiesis and hematologic tumors. Adipocytes are an important part of the bone marrow microenvironment, in which they can release free fatty acids (FFAs) through lipolysis and secrete adipocytokines, etc., and participate in normal hematopoiesis, which is closely related to the occurrence and treatment of hematological tumors. In this review, we aim to discuss how bone marrow adipocytes (BMAs) can influence the proliferation, apoptosis, and chemotherapy resistance of cancer cells by reprogramming lipid metabolism and the secretion of various adipocytokines.
Methods: Studies from 2000 to July 2024 were reviewed from PubMed, Springer Link, and the Web of Science using the keywords bone marrow microenvironment, adipocytes, lipid metabolism, adipocytokines, hematological tumor, cancer, and their combinations. Unreliable articles such as those that are old and have a low impact factor are excluded, and there is no restriction on language.
Key content and findings: Adipocytes can regulate the proliferation and differentiation of hematopoietic stem cells (HSCs) by secreting inflammatory factors and adipocytokines to maintain hematopoietic homeostasis. Adipocytes can also stimulate and accelerate the occurrence and progression of hematological tumors by secreting adipocytokines and mediating the reprogramming of lipid metabolism. Moreover, abundant adipocytes in bone marrow can protect tumor cells by physically blocking and/or secreting cytokines, leading to chemotherapy resistance.
Conclusions: Therefore, the targeted inhibition of related lipid metabolism pathways and adipocytokines might be a potential therapeutic target for hematological tumors, which would be helpful to inhibit tumor growth and correct chemotherapy resistance.
背景和目的:骨髓微环境与正常造血和血液肿瘤密切相关。脂肪细胞是骨髓微环境的重要组成部分,可通过脂肪分解释放游离脂肪酸(FFAs)、分泌脂肪细胞因子等,参与正常造血,与血液肿瘤的发生和治疗密切相关。本综述旨在探讨骨髓脂肪细胞(BMAs)如何通过重编程脂质代谢和分泌各种脂肪细胞因子来影响癌细胞的增殖、凋亡和化疗耐药性:方法:以骨髓微环境、脂肪细胞、脂质代谢、脂肪细胞因子、血液肿瘤、癌症及其组合为关键词,从 PubMed、Springer Link 和 Web of Science 上查阅了 2000 年至 2024 年 7 月期间的研究。不可靠的文章,如年代久远、影响因子低的文章将被排除在外,语言不限:脂肪细胞可以通过分泌炎症因子和脂肪细胞因子来调节造血干细胞(HSCs)的增殖和分化,从而维持造血干细胞的平衡。脂肪细胞还可通过分泌脂肪细胞因子和介导脂质代谢重编程,刺激和加速血液肿瘤的发生和发展。此外,骨髓中丰富的脂肪细胞可通过物理阻断和/或分泌细胞因子来保护肿瘤细胞,从而导致化疗耐药:因此,靶向抑制相关脂质代谢通路和脂肪细胞因子可能是血液肿瘤的潜在治疗靶点,有助于抑制肿瘤生长和纠正化疗耐药。
{"title":"Relationship between adipocytes and hematological tumors in the bone marrow microenvironment: a literature review.","authors":"Yuchun Li, Linlin Wang, Jingyu Wang, Yaping Xin, Xiaodong Lyu","doi":"10.21037/tcr-24-52","DOIUrl":"https://doi.org/10.21037/tcr-24-52","url":null,"abstract":"<p><strong>Background and objective: </strong>The bone marrow microenvironment is closely related to normal hematopoiesis and hematologic tumors. Adipocytes are an important part of the bone marrow microenvironment, in which they can release free fatty acids (FFAs) through lipolysis and secrete adipocytokines, etc., and participate in normal hematopoiesis, which is closely related to the occurrence and treatment of hematological tumors. In this review, we aim to discuss how bone marrow adipocytes (BMAs) can influence the proliferation, apoptosis, and chemotherapy resistance of cancer cells by reprogramming lipid metabolism and the secretion of various adipocytokines.</p><p><strong>Methods: </strong>Studies from 2000 to July 2024 were reviewed from PubMed, Springer Link, and the Web of Science using the keywords bone marrow microenvironment, adipocytes, lipid metabolism, adipocytokines, hematological tumor, cancer, and their combinations. Unreliable articles such as those that are old and have a low impact factor are excluded, and there is no restriction on language.</p><p><strong>Key content and findings: </strong>Adipocytes can regulate the proliferation and differentiation of hematopoietic stem cells (HSCs) by secreting inflammatory factors and adipocytokines to maintain hematopoietic homeostasis. Adipocytes can also stimulate and accelerate the occurrence and progression of hematological tumors by secreting adipocytokines and mediating the reprogramming of lipid metabolism. Moreover, abundant adipocytes in bone marrow can protect tumor cells by physically blocking and/or secreting cytokines, leading to chemotherapy resistance.</p><p><strong>Conclusions: </strong>Therefore, the targeted inhibition of related lipid metabolism pathways and adipocytokines might be a potential therapeutic target for hematological tumors, which would be helpful to inhibit tumor growth and correct chemotherapy resistance.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 10","pages":"5691-5701"},"PeriodicalIF":1.5,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11543051/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142628711","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 : 2024-10-31Epub Date: 2024-09-11DOI: 10.21037/tcr-24-258
Haodi Wang, Yue Wang, Pengyu Tan, Yichi Liu, Sa Zhou, Wenjian Ma
Background: Transmembrane p24 trafficking protein 9 (TMED9) belongs to the TMED family, and its overexpression frequently induces cancer. Studies have demonstrated the association between the overexpression of TMED9 and cancer development and proliferative migration in cancers such as ovarian cancer, hepatocellular carcinoma, and breast cancer. However, there has been no study investigating the clinical value, biological function, and anti-tumor immune effects of TMED9 from a pan-cancer perspective. The aim of this study is to evaluate the prognostic value and anti-tumor immunity role of TMED9 across pan-cancers.
Methods: We utilized R language along with The Cancer Genome Atlas (TCGA), UCSC Xena (University of California, Santa Cruz Xena Browser), Human Protein Atlas (HPA), and other datasets to investigate TMED9 expression in various tumors. The association between high TMED9 expression and clinical prognosis and patient survival was examined using the Kaplan-Meier method, log-rank test, as well as univariate and multivariate Cox regression analyses. Tumor Immune Estimation Resource 2.0 (TIMER2.0) and various algorithms were employed to explore the relationship between TMED9 and the tumor microenvironment (TME). Additionally, the biological function of TMED9 in cancer was investigated through Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and gene set enrichment analysis (GSEA) analyses.
Results: TMED9 was over-expressed in the majority of cancers. Patients exhibiting elevated TMED9 expression typically experienced diminished survival rates and unfavorable clinical outcomes. TMED9 played a role as a mediator in the aggressive phenotype of numerous tumors, actively engaging in various biological and signaling pathways linked to cancer development. TMED9 demonstrated the capacity to modulate the anti-tumor immune response in pan-cancer patients, exerting its influence on the infiltration levels of immune cells and cancer-associated fibroblasts (CAFs).
Conclusions: TMED9 serves as a novel "cancer indicator" and "clinical prognostic marker", capable of reshaping the TME, impacting the immunotherapeutic response, and guiding precise treatments for cancers to a certain extent.
{"title":"Prognostic value and anti-tumor immunity role of <i>TMED9</i> in pan-cancer: a bioinformatics study.","authors":"Haodi Wang, Yue Wang, Pengyu Tan, Yichi Liu, Sa Zhou, Wenjian Ma","doi":"10.21037/tcr-24-258","DOIUrl":"https://doi.org/10.21037/tcr-24-258","url":null,"abstract":"<p><strong>Background: </strong>Transmembrane p24 trafficking protein 9 (<i>TMED9</i>) belongs to the TMED family, and its overexpression frequently induces cancer. Studies have demonstrated the association between the overexpression of <i>TMED9</i> and cancer development and proliferative migration in cancers such as ovarian cancer, hepatocellular carcinoma, and breast cancer. However, there has been no study investigating the clinical value, biological function, and anti-tumor immune effects of <i>TMED9</i> from a pan-cancer perspective. The aim of this study is to evaluate the prognostic value and anti-tumor immunity role of <i>TMED9</i> across pan-cancers.</p><p><strong>Methods: </strong>We utilized R language along with The Cancer Genome Atlas (TCGA), UCSC Xena (University of California, Santa Cruz Xena Browser), Human Protein Atlas (HPA), and other datasets to investigate <i>TMED9</i> expression in various tumors. The association between high <i>TMED9</i> expression and clinical prognosis and patient survival was examined using the Kaplan-Meier method, log-rank test, as well as univariate and multivariate Cox regression analyses. Tumor Immune Estimation Resource 2.0 (TIMER2.0) and various algorithms were employed to explore the relationship between <i>TMED9</i> and the tumor microenvironment (TME). Additionally, the biological function of <i>TMED9</i> in cancer was investigated through Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and gene set enrichment analysis (GSEA) analyses.</p><p><strong>Results: </strong><i>TMED9</i> was over-expressed in the majority of cancers. Patients exhibiting elevated <i>TMED9</i> expression typically experienced diminished survival rates and unfavorable clinical outcomes. <i>TMED9</i> played a role as a mediator in the aggressive phenotype of numerous tumors, actively engaging in various biological and signaling pathways linked to cancer development. <i>TMED9</i> demonstrated the capacity to modulate the anti-tumor immune response in pan-cancer patients, exerting its influence on the infiltration levels of immune cells and cancer-associated fibroblasts (CAFs).</p><p><strong>Conclusions: </strong><i>TMED9</i> serves as a novel \"cancer indicator\" and \"clinical prognostic marker\", capable of reshaping the TME, impacting the immunotherapeutic response, and guiding precise treatments for cancers to a certain extent.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 10","pages":"5429-5445"},"PeriodicalIF":1.5,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11543038/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142628709","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: Oral squamous cell carcinoma (OSCC) is one of the most malignant tumors in the oral and maxillofacial region, with a poor prognosis. Previous studies have shown that long non-coding RNAs (lncRNAs) play a crucial role in tumor development by regulating the biological behavior of various cancer cells. The aim of this study is to explore the role and potential mechanisms of lncRNA PRKCA-AS1 in OSCC.
Methods: Real-time fluorescent quantitative polymerase chain reaction (RT-qPCR) was used to detect the expression levels of lncRNA PRKCA-AS1 in OSCC tissues and cell lines. Cell proliferation, migration and invasion were conducted to assess the biological functions of OSCC cell lines.
Results: The expression of lncRNA PRKCA-AS1 in OSCC tissues was higher compared to that of adjacent non-cancerous tissues, and its expression level was associated with the depth of tumor infiltration, lymph node metastasis, and tumor node metastasis (TNM) staging. Compared to the control group of normal human oral keratinocytes (HOK), the expression of lncRNA PRKCA-AS1 was also elevated in OSCC cell lines. Knockdown of lncRNA PRKCA-AS1 significantly affected the proliferation, migration, and invasion ability of OSCC cells. However, when lncRNA PRKCA-AS1 was further overexpressed, changes in cell proliferation and migration ability did not show statistical differences.
Conclusions: LncRNA PRKCA-AS1 is highly expressed in OSCC, and its expression level is positively correlated with the depth of tumor infiltration, lymph node metastasis, and TNM staging. LncRNA PRKCA-AS1 is involved in regulating the proliferation, migration, and invasion of OSCC cells.
{"title":"Study on the expression of lncRNA <i>PRKCA-AS1</i> in oral squamous cell carcinoma.","authors":"Dongyong Wang, Rui Li, Qian Liu, Chao Deng, Qiong Zhou, Wenjie Wen, Lin Chai","doi":"10.21037/tcr-24-467","DOIUrl":"https://doi.org/10.21037/tcr-24-467","url":null,"abstract":"<p><strong>Background: </strong>Oral squamous cell carcinoma (OSCC) is one of the most malignant tumors in the oral and maxillofacial region, with a poor prognosis. Previous studies have shown that long non-coding RNAs (lncRNAs) play a crucial role in tumor development by regulating the biological behavior of various cancer cells. The aim of this study is to explore the role and potential mechanisms of lncRNA <i>PRKCA-AS1</i> in OSCC.</p><p><strong>Methods: </strong>Real-time fluorescent quantitative polymerase chain reaction (RT-qPCR) was used to detect the expression levels of lncRNA <i>PRKCA-AS1</i> in OSCC tissues and cell lines. Cell proliferation, migration and invasion were conducted to assess the biological functions of OSCC cell lines.</p><p><strong>Results: </strong>The expression of lncRNA <i>PRKCA-AS1</i> in OSCC tissues was higher compared to that of adjacent non-cancerous tissues, and its expression level was associated with the depth of tumor infiltration, lymph node metastasis, and tumor node metastasis (TNM) staging. Compared to the control group of normal human oral keratinocytes (HOK), the expression of lncRNA <i>PRKCA-AS1</i> was also elevated in OSCC cell lines. Knockdown of lncRNA <i>PRKCA-AS1</i> significantly affected the proliferation, migration, and invasion ability of OSCC cells. However, when lncRNA <i>PRKCA-AS1</i> was further overexpressed, changes in cell proliferation and migration ability did not show statistical differences.</p><p><strong>Conclusions: </strong>LncRNA <i>PRKCA-AS1</i> is highly expressed in OSCC, and its expression level is positively correlated with the depth of tumor infiltration, lymph node metastasis, and TNM staging. LncRNA <i>PRKCA-AS1</i> is involved in regulating the proliferation, migration, and invasion of OSCC cells.</p>","PeriodicalId":23216,"journal":{"name":"Translational cancer research","volume":"13 10","pages":"5202-5213"},"PeriodicalIF":1.5,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11543052/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142628761","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}