首页 > 最新文献

Oncology Research最新文献

英文 中文
Loss of Arhgap39 facilitates cell migration and invasion in murine hepatocellular cancer cells.
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2025-01-16 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.053791
Hung-Wei Lin, Pei Yu Lee, Yu-Shiuan Chang, Mau-Sun Chang

Background: Rho GTPases are essential regulators for cellular movement and intracellular membrane trafficking. Their enzymatic activities fluctuate between active GTP-bound and inactive GDP-bound states regulated by GTPase activating proteins (GAPs) and guanine nucleotide exchange factors (GEFs). Arhgap39/Vilse/Porf-2 is a newly identified GAP. The role of Arhgap39 in migration and invasion has not been addressed thoroughly.

Methods: The Arhgap39 gene was knocked out by Crispr-Cas9 gene editing in mouse Hepa1-6 and Hepa-1c1c7 cells to analyze the impact of Arhgap39 depletion on migration and invasion.

Results: Loss of Arhgap39 noticeably increased the migration and invasive potential. Purified Arhgap39 recombinant protein facilitated the hydrolysis of GTP in RhoA and Rac1 in vitro. RNA-seq analysis revealed that matrix metalloproteinase 13 (MMP13) and Laminin subunit beta 1 (LAMB1) were increased in Arhgap39-/- cells. We further crossed Arhgap39fl/fl with KrasLSL-G12D and p53fl/fl mice under the control of albumin-Cre recombinase to induce the spontaneous development of hepatocellular carcinomas. Intriguingly, the expression levels of MMP13 and the overall survival in Alb-Cre_KrasLSL-G12D_p53fl/fl_Arhgap39fl/fl (KPA) mice were comparable to control Alb-Cre_KrasLSL-G12D_p53fl/fl (KP) mice. The cell migration and invasion of KPA mice were also similar to those of control KP mice.

Conclusion: Arhgap39 loss could modulate the migration and invasion in some hepatocellular cancer cells, but not in those isolated from KPA mice.

{"title":"Loss of Arhgap39 facilitates cell migration and invasion in murine hepatocellular cancer cells.","authors":"Hung-Wei Lin, Pei Yu Lee, Yu-Shiuan Chang, Mau-Sun Chang","doi":"10.32604/or.2024.053791","DOIUrl":"10.32604/or.2024.053791","url":null,"abstract":"<p><strong>Background: </strong>Rho GTPases are essential regulators for cellular movement and intracellular membrane trafficking. Their enzymatic activities fluctuate between active GTP-bound and inactive GDP-bound states regulated by GTPase activating proteins (GAPs) and guanine nucleotide exchange factors (GEFs). Arhgap39/Vilse/Porf-2 is a newly identified GAP. The role of Arhgap39 in migration and invasion has not been addressed thoroughly.</p><p><strong>Methods: </strong>The Arhgap39 gene was knocked out by Crispr-Cas9 gene editing in mouse Hepa1-6 and Hepa-1c1c7 cells to analyze the impact of Arhgap39 depletion on migration and invasion.</p><p><strong>Results: </strong>Loss of Arhgap39 noticeably increased the migration and invasive potential. Purified Arhgap39 recombinant protein facilitated the hydrolysis of GTP in RhoA and Rac1 <i>in vitro</i>. RNA-seq analysis revealed that matrix metalloproteinase 13 (MMP13) and Laminin subunit beta 1 (LAMB1) were increased in Arhgap39<sup>-/-</sup> cells. We further crossed Arhgap39fl/fl with KrasLSL-G12D and p53fl/fl mice under the control of albumin-Cre recombinase to induce the spontaneous development of hepatocellular carcinomas. Intriguingly, the expression levels of MMP13 and the overall survival in Alb-Cre_KrasLSL-G12D_p53fl/fl_Arhgap39fl/fl (KPA) mice were comparable to control Alb-Cre_KrasLSL-G12D_p53fl/fl (KP) mice. The cell migration and invasion of KPA mice were also similar to those of control KP mice.</p><p><strong>Conclusion: </strong>Arhgap39 loss could modulate the migration and invasion in some hepatocellular cancer cells, but not in those isolated from KPA mice.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 2","pages":"493-503"},"PeriodicalIF":2.0,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11753993/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143047413","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Melanoma cell line-derived exosomal miR-424-5p: a key promoter of angiogenesis through LATS2 interaction. 黑色素瘤细胞系衍生的外泌体 miR-424-5p:通过 LATS2 相互作用促进血管生成的关键因素。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2025-01-16 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.050878
Junwei DU, Qiang Zhang, Jing Zhang, Maierdanjiang Maihemuti, Haiyang He, Renbing Jiang

Objectives: Melanoma is a highly aggressive and metastatic form of cancer, and the role of exosomal microRNAs (miRNAs) in its progression remains largely unexplored. This study aimed to investigate the effects of melanoma cell-derived exosomal miR-424-5p on angiogenesis and its underlying mechanisms.

Methods: Exosomes were isolated from melanoma cell lines A375 and A2058, and their effects on the proliferation, migration, and angiogenesis of human umbilical vein endothelial cells (HUVECs) were examined. The interaction between miR-424-5p and its target gene, large tumor suppressor kinase 2 (LATS2), was analyzed using luciferase reporter assays and functional experiments. In vivo, tumor growth and angiogenesis were studied in a xenograft model using nude mice.

Results: Melanoma cell-derived exosomes could be internalized by HUVECs, which promoted proliferation, migration, and angiogenesis. miR-424-5p was highly expressed in melanoma cells and their exosomes, and its inhibition in exosomes suppressed HUVEC proliferation, migration, and angiogenesis. LATS2 was identified as a direct target of miR-424-5p, and its silencing reversed the inhibitory effects of miR-424-5p inhibition on HUVEC functions. In vivo, exosomes derived from miR-424-5p-inhibited melanoma cells suppressed tumor growth and angiogenesis in xenograft models.

Conclusions: Melanoma cell-derived exosomal miR-424-5p promotes angiogenesis by targeting LATS2, contributing to melanoma progression. Targeting the exosomal miR-424-5p/LATS2 axis could be a potential therapeutic strategy for melanoma.

{"title":"Melanoma cell line-derived exosomal miR-424-5p: a key promoter of angiogenesis through LATS2 interaction.","authors":"Junwei DU, Qiang Zhang, Jing Zhang, Maierdanjiang Maihemuti, Haiyang He, Renbing Jiang","doi":"10.32604/or.2024.050878","DOIUrl":"10.32604/or.2024.050878","url":null,"abstract":"<p><strong>Objectives: </strong>Melanoma is a highly aggressive and metastatic form of cancer, and the role of exosomal microRNAs (miRNAs) in its progression remains largely unexplored. This study aimed to investigate the effects of melanoma cell-derived exosomal miR-424-5p on angiogenesis and its underlying mechanisms.</p><p><strong>Methods: </strong>Exosomes were isolated from melanoma cell lines A375 and A2058, and their effects on the proliferation, migration, and angiogenesis of human umbilical vein endothelial cells (HUVECs) were examined. The interaction between miR-424-5p and its target gene, large tumor suppressor kinase 2 (LATS2), was analyzed using luciferase reporter assays and functional experiments. <i>In vivo</i>, tumor growth and angiogenesis were studied in a xenograft model using nude mice.</p><p><strong>Results: </strong>Melanoma cell-derived exosomes could be internalized by HUVECs, which promoted proliferation, migration, and angiogenesis. miR-424-5p was highly expressed in melanoma cells and their exosomes, and its inhibition in exosomes suppressed HUVEC proliferation, migration, and angiogenesis. LATS2 was identified as a direct target of miR-424-5p, and its silencing reversed the inhibitory effects of miR-424-5p inhibition on HUVEC functions. <i>In vivo</i>, exosomes derived from miR-424-5p-inhibited melanoma cells suppressed tumor growth and angiogenesis in xenograft models.</p><p><strong>Conclusions: </strong>Melanoma cell-derived exosomal miR-424-5p promotes angiogenesis by targeting LATS2, contributing to melanoma progression. Targeting the exosomal miR-424-5p/LATS2 axis could be a potential therapeutic strategy for melanoma.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 2","pages":"357-367"},"PeriodicalIF":2.0,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11753990/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143047467","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Comprehensive analysis reveals PLK3 as a promising immune target and prognostic indicator in glioma.
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2025-01-16 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.050794
Tianyun Zhu, Cunyan Zhao, Rui Gong, A O Qian, Xiaoshu Wang, Fanghui Lu, Gang Huo, Liangjun Qiao, Song Chen

Background: PLK3, which played an important role in cell cycle progression and stress response, was identified as highly expressed in various carcinomas. However, the functions, molecular characteristics, and prognostic value of PLK3 in glioma remained unexplored.

Methods: We analyzed PLK3 expression in glioma samples from multiple databases. Both overexpression and knockdown of Plk3 were performed to investigate tumor cell growth in glioma, and the transplanted glioma mouse model demonstrated the role of Plk3 on tumor progression. Immunohistochemistry was conducted to detect PLK3 expression and immune cell infiltration. The trans-well assay for PLK3 on the immune cells recruitment was also determined. Additionally, we further evaluated the correlation between PLK3 and PD-1/PD-L1 as well as other immune checkpoints.

Results: We found that an increased level of PLK3 was associated with malignancy and poor prognosis of glioma, and further validated that PLK3 promoted glioma progression. PLK3 also played a crucial role in immune response and was involved in Tcell immune suppression. Specifically, we revealed that CD8+ and CD4+ Tcell infiltration was decreased in Plk3 overexpressed xenografts. Furthermore, it was predicted that PLK3 was synergistic with other checkpoint members in glioma. In general, high expression of PLK3 was associated with a malignant process and poor prognosis in glioma patients.

Conclusion: Our findings indicated that PLK3 expression level was highly correlated to the malignancy of gliomas, and we validated that PLK3 could promote the GBM progress in vitro and in vivo. Furthermore, PLK3 played important roles in Tcell and neutrophil immune response in glioma. Besides, the conspicuous association between PLK3 and other immune checkpoints was also observed. Crucially, high-level PLK3 expression was revealed to be related to poor clinical prognosis. These results demonstrated that PLK3 may serve as a prognostic biomarker and a potential target for glioma.

{"title":"Comprehensive analysis reveals PLK3 as a promising immune target and prognostic indicator in glioma.","authors":"Tianyun Zhu, Cunyan Zhao, Rui Gong, A O Qian, Xiaoshu Wang, Fanghui Lu, Gang Huo, Liangjun Qiao, Song Chen","doi":"10.32604/or.2024.050794","DOIUrl":"https://doi.org/10.32604/or.2024.050794","url":null,"abstract":"<p><strong>Background: </strong>PLK3, which played an important role in cell cycle progression and stress response, was identified as highly expressed in various carcinomas. However, the functions, molecular characteristics, and prognostic value of PLK3 in glioma remained unexplored.</p><p><strong>Methods: </strong>We analyzed PLK3 expression in glioma samples from multiple databases. Both overexpression and knockdown of Plk3 were performed to investigate tumor cell growth in glioma, and the transplanted glioma mouse model demonstrated the role of Plk3 on tumor progression. Immunohistochemistry was conducted to detect PLK3 expression and immune cell infiltration. The trans-well assay for PLK3 on the immune cells recruitment was also determined. Additionally, we further evaluated the correlation between PLK3 and PD-1/PD-L1 as well as other immune checkpoints.</p><p><strong>Results: </strong>We found that an increased level of PLK3 was associated with malignancy and poor prognosis of glioma, and further validated that PLK3 promoted glioma progression. PLK3 also played a crucial role in immune response and was involved in Tcell immune suppression. Specifically, we revealed that CD8<sup>+</sup> and CD4<sup>+</sup> Tcell infiltration was decreased in Plk3 overexpressed xenografts. Furthermore, it was predicted that PLK3 was synergistic with other checkpoint members in glioma. In general, high expression of PLK3 was associated with a malignant process and poor prognosis in glioma patients.</p><p><strong>Conclusion: </strong>Our findings indicated that PLK3 expression level was highly correlated to the malignancy of gliomas, and we validated that PLK3 could promote the GBM progress <i>in vitro</i> and <i>in vivo</i>. Furthermore, PLK3 played important roles in Tcell and neutrophil immune response in glioma. Besides, the conspicuous association between PLK3 and other immune checkpoints was also observed. Crucially, high-level PLK3 expression was revealed to be related to poor clinical prognosis. These results demonstrated that PLK3 may serve as a prognostic biomarker and a potential target for glioma.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 2","pages":"431-442"},"PeriodicalIF":2.0,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11753997/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143059940","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Apigenin facilitates apoptosis of acute lymphoblastic leukemia cells via AMP-activated protein kinase-mediated ferroptosis.
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2025-01-16 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.049757
Cancan He, Tingting Zhang, Wei Xiong, Shengyu Wang, Xin Sun

Background: The outcomes of pediatric patients with acute lymphoblastic leukemia (ALL) remain far less than favorable. While apigenin is an anti-cancer agent, studies on the mechanism by which it regulates ALL cell cycle progression are inadequate. Ferroptosis and AMP-activated protein kinase (AMPK) signaling are important processes for ALL patients. However, it remains unclear whether apigenin works by affecting AMPK and apoptosis.

Materials and methods: SUP-B15 and T-cell Jurkat ALL cells were treated with apigenin, and cell viability and apoptosis were measured using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assays, respectively. The thiobarbituric acid-reactive substances (TBARS) assay was used to evaluate lipid peroxidation. Intracellular Fe2+ levels were measured using a commercial kit. Corresponding proteins were detected by western blotting.

Results: Results showed that apigenin reduced cell viability and the levels of Ki67 and proliferating cell nuclear antigen (PCNA) expression in a concentration-dependent manner in both types of ALL cells. Apigenin also exerted anti-apoptotic effects on SUP-B15 and Jurkat cells. Apigenin activated AMP-activated protein kinase (AMPK) signaling and induced ferroptosis, and those effects were attenuated by inhibition of AMPK. Eventually, the reduced cell proliferation and increased cell apoptosis caused by apigenin in ALL cells were partly abolished by AMPK inhibition.

Conclusion: In summary, apigenin exerted anti-leukemia activity in ALL cells, and that effect was partially achieved by activation of AMPK signaling. Our findings suggest apigenin as a potential drug for treatment of ALL.

{"title":"Apigenin facilitates apoptosis of acute lymphoblastic leukemia cells via AMP-activated protein kinase-mediated ferroptosis.","authors":"Cancan He, Tingting Zhang, Wei Xiong, Shengyu Wang, Xin Sun","doi":"10.32604/or.2024.049757","DOIUrl":"10.32604/or.2024.049757","url":null,"abstract":"<p><strong>Background: </strong>The outcomes of pediatric patients with acute lymphoblastic leukemia (ALL) remain far less than favorable. While apigenin is an anti-cancer agent, studies on the mechanism by which it regulates ALL cell cycle progression are inadequate. Ferroptosis and AMP-activated protein kinase (AMPK) signaling are important processes for ALL patients. However, it remains unclear whether apigenin works by affecting AMPK and apoptosis.</p><p><strong>Materials and methods: </strong>SUP-B15 and T-cell Jurkat ALL cells were treated with apigenin, and cell viability and apoptosis were measured using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assays, respectively. The thiobarbituric acid-reactive substances (TBARS) assay was used to evaluate lipid peroxidation. Intracellular Fe<sup>2+</sup> levels were measured using a commercial kit. Corresponding proteins were detected by western blotting.</p><p><strong>Results: </strong>Results showed that apigenin reduced cell viability and the levels of Ki67 and proliferating cell nuclear antigen (PCNA) expression in a concentration-dependent manner in both types of ALL cells. Apigenin also exerted anti-apoptotic effects on SUP-B15 and Jurkat cells. Apigenin activated AMP-activated protein kinase (AMPK) signaling and induced ferroptosis, and those effects were attenuated by inhibition of AMPK. Eventually, the reduced cell proliferation and increased cell apoptosis caused by apigenin in ALL cells were partly abolished by AMPK inhibition.</p><p><strong>Conclusion: </strong>In summary, apigenin exerted anti-leukemia activity in ALL cells, and that effect was partially achieved by activation of AMPK signaling. Our findings suggest apigenin as a potential drug for treatment of ALL.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 2","pages":"421-429"},"PeriodicalIF":2.0,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11753985/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143047406","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
KHSRP promotes cancer stem cell maintenance, tumorigenesis, and suppresses anti-tumor immunity in gastric cancer.
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2025-01-16 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.058273
Yaru DU, Zhihui Pei, Shuqin Hu, Chuanwen Liao, Shuhao Liu

Objectives: KH-type splicing regulatory protein (KHSRP) is an RNA-binding protein involved in several cellular processes, including nuclear splicing, mRNA localization, and cytoplasmic degradation. While KHSRP's role has been studied in other cancers, its specific involvement in gastric cancer remains poorly understood. This study aims to explore KHSRP expression in gastric cancer and its potential effects on tumor progression and immune response.

Methods: KHSRP expression in gastric cancer tissues and normal tissues was analyzed using data from The Cancer Genome Atlas (TCGA) database. The correlation between KHSRP expression, patient survival, and immune response was also assessed. Immunohistochemistry was performed to evaluate KHSRP expression in gastric cancer tissues. Gain- and loss-of-function experiments were conducted to assess KHSRP's effects on gastric cancer cell proliferation, stemness, and migration. Furthermore, the impact of KHSRP silencing on tumor volume and immune cell infiltration was evaluated in a C3H/He mouse xenograft model.

Results: KHSRP was found to be overexpressed in gastric cancer tissues compared to normal tissues, with a positive correlation to tumor stage and a negative correlation with patient prognosis. Functional assays revealed that KHSRP promotes gastric cancer cell proliferation, enhances cancer stem cell properties, and increases migratory capabilities in vitro. In vivo, KHSRP silencing led to a significant reduction in tumor volume and increased immune cell infiltration in the mouse xenograft model.

Conclusions: KHSRP acts as an oncogene in gastric cancer by promoting tumorigenesis and suppressing anti-tumor immune responses. Its overexpression is associated with poor prognosis, making KHSRP a potential prognostic marker and therapeutic target in gastric cancer.

{"title":"KHSRP promotes cancer stem cell maintenance, tumorigenesis, and suppresses anti-tumor immunity in gastric cancer.","authors":"Yaru DU, Zhihui Pei, Shuqin Hu, Chuanwen Liao, Shuhao Liu","doi":"10.32604/or.2024.058273","DOIUrl":"10.32604/or.2024.058273","url":null,"abstract":"<p><strong>Objectives: </strong>KH-type splicing regulatory protein (KHSRP) is an RNA-binding protein involved in several cellular processes, including nuclear splicing, mRNA localization, and cytoplasmic degradation. While KHSRP's role has been studied in other cancers, its specific involvement in gastric cancer remains poorly understood. This study aims to explore KHSRP expression in gastric cancer and its potential effects on tumor progression and immune response.</p><p><strong>Methods: </strong>KHSRP expression in gastric cancer tissues and normal tissues was analyzed using data from The Cancer Genome Atlas (TCGA) database. The correlation between KHSRP expression, patient survival, and immune response was also assessed. Immunohistochemistry was performed to evaluate KHSRP expression in gastric cancer tissues. Gain- and loss-of-function experiments were conducted to assess KHSRP's effects on gastric cancer cell proliferation, stemness, and migration. Furthermore, the impact of KHSRP silencing on tumor volume and immune cell infiltration was evaluated in a C3H/He mouse xenograft model.</p><p><strong>Results: </strong>KHSRP was found to be overexpressed in gastric cancer tissues compared to normal tissues, with a positive correlation to tumor stage and a negative correlation with patient prognosis. Functional assays revealed that KHSRP promotes gastric cancer cell proliferation, enhances cancer stem cell properties, and increases migratory capabilities <i>in vitro</i>. <i>In vivo</i>, KHSRP silencing led to a significant reduction in tumor volume and increased immune cell infiltration in the mouse xenograft model.</p><p><strong>Conclusions: </strong>KHSRP acts as an oncogene in gastric cancer by promoting tumorigenesis and suppressing anti-tumor immune responses. Its overexpression is associated with poor prognosis, making KHSRP a potential prognostic marker and therapeutic target in gastric cancer.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 2","pages":"309-325"},"PeriodicalIF":2.0,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11753988/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143047292","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Exosomal miR-224-3p promotes lymphangiogenesis and lymph node metastasis by targeting GSK3B in gastric cancer.
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2025-01-16 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.050431
Zhengyang Zhou, Lei Qiao, Tongtong Wang, Wen Pan, Jingjing Duan, Haiyang Zhang, Ting Deng, Y I Ba, Y I He

Background: Patients with gastric cancer (GC) are prone to lymph node metastasis (LNM), which is an important factor for recurrence and poor prognosis of GC. Nowadays, more and more studies have confirmed that exosomes can participate in tumor lymphangiogenesis. An in-depth exploration of the pathological mechanism in the process of LNM in GC may provide effective targets and improve the diagnosis and treatment effect.

Materials and methods: We used sequencing analysis of collected serum to screen out exo-miRNA related to LNM in GC. ELISA, qRT-PCR, Western Blot, RNA pull-down assay, Transwell assay, animal experiments, and other experiments were used to verify the results.

Results: In this study, we screened out miR-224-3p related to GC progression and LNM in a vascular endothelial growth Factor C (VEGFC)-independent manner. We found that exo-miR-224-3p derived from GC cells could enter human lymphatic endothelial cells (HLECs) and promote the tube formation and migration of HLECs. In addition, it was revealed that miR-224-3p could bind to the 3'UTR region of GSK3B mRNA. Then, we proved that inhibiting the expression of GSK3B could suppress the phosphorylation of β-catenin and promote the transcription of PROX1, thus leading to tumor lymphangiogenesis. Furthermore, it was also found that hnRNPA1 mediated the sorting of miR-224-3p into exosomes, and the high expression of PKM2 promoted the secretion of exo-miR-224-3p.

Conclusions: Our discovery of the exo-miR-224-3p/GSK3B/β-catenin/PROX1 axis may provide a new direction for the clinical treatment of GC.

{"title":"Exosomal miR-224-3p promotes lymphangiogenesis and lymph node metastasis by targeting GSK3B in gastric cancer.","authors":"Zhengyang Zhou, Lei Qiao, Tongtong Wang, Wen Pan, Jingjing Duan, Haiyang Zhang, Ting Deng, Y I Ba, Y I He","doi":"10.32604/or.2024.050431","DOIUrl":"10.32604/or.2024.050431","url":null,"abstract":"<p><strong>Background: </strong>Patients with gastric cancer (GC) are prone to lymph node metastasis (LNM), which is an important factor for recurrence and poor prognosis of GC. Nowadays, more and more studies have confirmed that exosomes can participate in tumor lymphangiogenesis. An in-depth exploration of the pathological mechanism in the process of LNM in GC may provide effective targets and improve the diagnosis and treatment effect.</p><p><strong>Materials and methods: </strong>We used sequencing analysis of collected serum to screen out exo-miRNA related to LNM in GC. ELISA, qRT-PCR, Western Blot, RNA pull-down assay, Transwell assay, animal experiments, and other experiments were used to verify the results.</p><p><strong>Results: </strong>In this study, we screened out miR-224-3p related to GC progression and LNM in a vascular endothelial growth Factor C (VEGFC)-independent manner. We found that exo-miR-224-3p derived from GC cells could enter human lymphatic endothelial cells (HLECs) and promote the tube formation and migration of HLECs. In addition, it was revealed that miR-224-3p could bind to the 3'UTR region of GSK3B mRNA. Then, we proved that inhibiting the expression of GSK3B could suppress the phosphorylation of β-catenin and promote the transcription of PROX1, thus leading to tumor lymphangiogenesis. Furthermore, it was also found that hnRNPA1 mediated the sorting of miR-224-3p into exosomes, and the high expression of PKM2 promoted the secretion of exo-miR-224-3p.</p><p><strong>Conclusions: </strong>Our discovery of the exo-miR-224-3p/GSK3B/β-catenin/PROX1 axis may provide a new direction for the clinical treatment of GC.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 2","pages":"327-345"},"PeriodicalIF":2.0,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11753999/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143046518","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Circular RNA circAGAP1 promotes sunitinib sensitivity in renal cell carcinoma via sponging multiple PDGFR-targeted miRNAs. 环状 RNA circAGAP1 通过疏导多个 PDGFR 靶向 miRNA 促进肾细胞癌中舒尼替尼的敏感性。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2025-01-16 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.047698
Q I Lv, Gangmin Wang, Y I Hong, Tianyi Zhu, Shuang Qin, Saifei Sun, Yuting Wang, Yaohua Liu, Qing Zhang, Chunhui Ma, Peijun Wang

Background: Sunitinib resistance is a major challenge in advanced renal cell carcinoma (RCC). Clinically, elucidating the underlying mechanisms and developing practical countermeasures for sunitinib resistance in RCC is desirable. In previous studies, we found that circAGAP1 expression was significantly upregulated in clear cell RCC (ccRCC) and was strongly associated with poor prognosis. However, the role of circAGAP1 in sunitinib resistance in ccRCC remains unclear.

Methods: We used public databases for bioinformatics analysis to identify the binding targets of circAGAP1. Additionally, the effects of circAGAP1 on the proliferation, clonogenesis, apoptosis, and migration of ccRCC cells were analyzed using quantitative real-time PCR, cell counting kit-8 assays, migration and apoptosis assays, and colony formation assays. Furthermore, RNA immunoprecipitation, dual-luciferase reporter, and fluorescence in situ hybridization assays were used to explore the molecular mechanism.

Results: In this study, circAGAP1 exhibited higher expression in sunitinib-sensitive ccRCC cells and inhibited the clonogenesis, proliferation, and migration of ccRCC cells after sunitinib treatment. Mechanical studies revealed that circAGAP1 regulated the expression of sunitinib target platelet-derived growth factor receptor by acting as a microRNA sponge that suppresses miR-149-5p, miR-455-5p, and miR-15a-5p simultaneously. Overexpression of these three miRNAs reversed circAGAP1-mediated sunitinib sensitivity in ccRCC.

Conclusions: In summary, our findings indicate that circAGAP1 may serve as a promising biomarker to predict sunitinib sensibility and a therapeutic target in ccRCC.

{"title":"Circular RNA circAGAP1 promotes sunitinib sensitivity in renal cell carcinoma via sponging multiple PDGFR-targeted miRNAs.","authors":"Q I Lv, Gangmin Wang, Y I Hong, Tianyi Zhu, Shuang Qin, Saifei Sun, Yuting Wang, Yaohua Liu, Qing Zhang, Chunhui Ma, Peijun Wang","doi":"10.32604/or.2024.047698","DOIUrl":"10.32604/or.2024.047698","url":null,"abstract":"<p><strong>Background: </strong>Sunitinib resistance is a major challenge in advanced renal cell carcinoma (RCC). Clinically, elucidating the underlying mechanisms and developing practical countermeasures for sunitinib resistance in RCC is desirable. In previous studies, we found that circAGAP1 expression was significantly upregulated in clear cell RCC (ccRCC) and was strongly associated with poor prognosis. However, the role of circAGAP1 in sunitinib resistance in ccRCC remains unclear.</p><p><strong>Methods: </strong>We used public databases for bioinformatics analysis to identify the binding targets of circAGAP1. Additionally, the effects of circAGAP1 on the proliferation, clonogenesis, apoptosis, and migration of ccRCC cells were analyzed using quantitative real-time PCR, cell counting kit-8 assays, migration and apoptosis assays, and colony formation assays. Furthermore, RNA immunoprecipitation, dual-luciferase reporter, and fluorescence <i>in situ</i> hybridization assays were used to explore the molecular mechanism.</p><p><strong>Results: </strong>In this study, circAGAP1 exhibited higher expression in sunitinib-sensitive ccRCC cells and inhibited the clonogenesis, proliferation, and migration of ccRCC cells after sunitinib treatment. Mechanical studies revealed that circAGAP1 regulated the expression of sunitinib target platelet-derived growth factor receptor by acting as a microRNA sponge that suppresses miR-149-5p, miR-455-5p, and miR-15a-5p simultaneously. Overexpression of these three miRNAs reversed circAGAP1-mediated sunitinib sensitivity in ccRCC.</p><p><strong>Conclusions: </strong>In summary, our findings indicate that circAGAP1 may serve as a promising biomarker to predict sunitinib sensibility and a therapeutic target in ccRCC.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 2","pages":"407-420"},"PeriodicalIF":2.0,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11754003/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143047436","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
EMP2 promotes hepatocellular carcinoma proliferation and invasion by activating cellular autophagy. EMP2 通过激活细胞自噬促进肝细胞癌的增殖和侵袭。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2025-01-16 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.043948
Haiying Pang, Fengbo Wu, Y U Zhang, Nan Zhang, Chunting Wang, Qiu Li, G U He, Peng Zhang

Background: EMP2 is a tumor-associated membrane protein belonging to the GAS-3/PMP22 gene family. EMP2 expression demonstrates significant tissue specificity and heterogeneity in various human tissues and tumor tissues, where it may play a role in either promoting or inhibiting tumor growth. This study aimed to investigate the expression level, biological functions, and molecular mechanisms of EMP2 in liver cancer.

Methods: we analyzed the mRNA expression levels of EMPs family genes in hepatocellular carcinoma (HCC) tissues and normal liver tissues based on the TCGA database and immunohistochemical analysis of tissue microarrays. Subsequently, we constructed HCC cell lines with either knockdown or overexpression of EMP2 to examine the biological functions and molecular mechanisms of EMP2 in tumorigenesis in vivo and in vitro.

Results: Bioinformatic and immunohistochemical analysis of tissue microarrays have confirmed the significant upregulation of EMP2 in HCC tissues. In vitro and in vivo studies have shown that downregulation of EMP2 results in a moderate reduction in the proliferation and invasive capacity of HCC cells. Conversely, overexpression of EMP2 enhances the invasive capacity of HCC cells and induces autophagy. Initial investigations into the molecular mechanisms underlying EMP2-mediated enhancement of HCC cell invasion have revealed the dual regulation of EMP2-induced autophagy and the integrin pathway, which synergistically influence the invasive and metastatic potential of HCC cells.

Conclusion: EMP2 holds promise as a diagnostic marker for HCC metastasis and a potential target for targeted therapy.

{"title":"EMP2 promotes hepatocellular carcinoma proliferation and invasion by activating cellular autophagy.","authors":"Haiying Pang, Fengbo Wu, Y U Zhang, Nan Zhang, Chunting Wang, Qiu Li, G U He, Peng Zhang","doi":"10.32604/or.2024.043948","DOIUrl":"10.32604/or.2024.043948","url":null,"abstract":"<p><strong>Background: </strong>EMP2 is a tumor-associated membrane protein belonging to the GAS-3/PMP22 gene family. EMP2 expression demonstrates significant tissue specificity and heterogeneity in various human tissues and tumor tissues, where it may play a role in either promoting or inhibiting tumor growth. This study aimed to investigate the expression level, biological functions, and molecular mechanisms of EMP2 in liver cancer.</p><p><strong>Methods: </strong>we analyzed the mRNA expression levels of EMPs family genes in hepatocellular carcinoma (HCC) tissues and normal liver tissues based on the TCGA database and immunohistochemical analysis of tissue microarrays. Subsequently, we constructed HCC cell lines with either knockdown or overexpression of EMP2 to examine the biological functions and molecular mechanisms of EMP2 in tumorigenesis <i>in vivo</i> and <i>in vitro</i>.</p><p><strong>Results: </strong>Bioinformatic and immunohistochemical analysis of tissue microarrays have confirmed the significant upregulation of EMP2 in HCC tissues. <i>In vitro</i> and <i>in vivo</i> studies have shown that downregulation of EMP2 results in a moderate reduction in the proliferation and invasive capacity of HCC cells. Conversely, overexpression of EMP2 enhances the invasive capacity of HCC cells and induces autophagy. Initial investigations into the molecular mechanisms underlying EMP2-mediated enhancement of HCC cell invasion have revealed the dual regulation of EMP2-induced autophagy and the integrin pathway, which synergistically influence the invasive and metastatic potential of HCC cells.</p><p><strong>Conclusion: </strong>EMP2 holds promise as a diagnostic marker for HCC metastasis and a potential target for targeted therapy.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 2","pages":"443-464"},"PeriodicalIF":2.0,"publicationDate":"2025-01-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11754002/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143046336","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Comprehensive molecular characterization to predict immunotherapy response in advanced biliary tract cancer: a phase II trial of pembrolizumab. 综合分子表征预测晚期胆道癌免疫治疗反应:派姆单抗II期试验
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-12-20 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.049054
Ryul Kim, Joo Kyung Park, Minsuk Kwon, Minae An, Jung Yong Hong, Joon Oh Park, Sung Hee Lim, Seung Tae Kim

Background: Immune checkpoint inhibitors (ICIs) are effective in a subset of patients with metastatic solid tumors. However, the patients who would benefit most from ICIs in biliary tract cancer (BTC) are still controversial.

Materials and methods: We molecularly characterized tissues and blood from 32 patients with metastatic BTC treated with the ICI pembrolizumab as second-line therapy.

Results: All patients had microsatellite stable (MSS) type tumors. Three of the 32 patients achieved partial response (PR), with an objective response rate (ORR) of 9.4% (95% confidence interval [CI], 2.0-25.2) and nine showed stable disease (SD), exhibiting a disease control rate (DCR) of 37.5% (95% CI, 21.1-56.3). For the 31 patients who had access to PD-1 ligand 1 (PD-L1) combined positive score (CPS) testing (cut-off value ≥1%), the ORR was not different between those who had PD-L1-positive (PD-L1+; 1/11, 9.1%) and PDL1-(2/20, 10.0%) tumors (p = 1.000). The tumor mutational burden (TMB) of PD-L1+ BTC was comparable to that of PD-L1-BTC (p = 0.630). TMB and any exonic somatic mutations were also not predictive of pembrolizumab response. Molecular analysis of blood and tumor samples demonstrated a relatively high natural killer (NK) cell proportion in the peripheral blood before pembrolizumab treatment in patients who achieved tumor response. Moreover, the tumors of these patients presented high enrichment scores for NK cells, antitumor cytokines, and Th1 signatures, and a low enrichment score for cancer-associated fibroblasts.

Conclusions: This study shows the molecular characteristics associated with the efficacy of pembrolizumab in BTC of the MSS type.

背景:免疫检查点抑制剂(ICIs)对转移性实体瘤患者有效。然而,在胆道肿瘤(BTC)中,哪些患者从ICIs中获益最大仍存在争议。材料和方法:我们对32例接受ICI派姆单抗作为二线治疗的转移性BTC患者的组织和血液进行了分子表征。结果:所有患者均为微卫星稳定型(MSS)肿瘤。32例患者中有3例达到部分缓解(PR),客观缓解率(ORR)为9.4%(95%可信区间[CI] 2.0 ~ 25.2), 9例病情稳定(SD),疾病控制率(DCR)为37.5% (95% CI 21.1 ~ 56.3)。对于31例获得PD-1配体1 (PD-L1)联合阳性评分(CPS)检测(临界值≥1%)的患者,PD-L1阳性(PD-L1+;1/11, 9.1%)和PDL1(2/20, 10.0%)肿瘤(p = 1.000)。PD-L1+ BTC的肿瘤突变负荷(TMB)与PD-L1-BTC相当(p = 0.630)。TMB和任何外显子体细胞突变也不能预测派姆单抗的反应。血液和肿瘤样本的分子分析表明,在获得肿瘤反应的患者接受派姆单抗治疗前,外周血中自然杀伤(NK)细胞比例相对较高。此外,这些患者的肿瘤表现出NK细胞、抗肿瘤细胞因子和Th1特征的高富集评分,而癌症相关成纤维细胞的低富集评分。结论:本研究显示了派姆单抗治疗MSS型BTC的相关分子特征。
{"title":"Comprehensive molecular characterization to predict immunotherapy response in advanced biliary tract cancer: a phase II trial of pembrolizumab.","authors":"Ryul Kim, Joo Kyung Park, Minsuk Kwon, Minae An, Jung Yong Hong, Joon Oh Park, Sung Hee Lim, Seung Tae Kim","doi":"10.32604/or.2024.049054","DOIUrl":"10.32604/or.2024.049054","url":null,"abstract":"<p><strong>Background: </strong>Immune checkpoint inhibitors (ICIs) are effective in a subset of patients with metastatic solid tumors. However, the patients who would benefit most from ICIs in biliary tract cancer (BTC) are still controversial.</p><p><strong>Materials and methods: </strong>We molecularly characterized tissues and blood from 32 patients with metastatic BTC treated with the ICI pembrolizumab as second-line therapy.</p><p><strong>Results: </strong>All patients had microsatellite stable (MSS) type tumors. Three of the 32 patients achieved partial response (PR), with an objective response rate (ORR) of 9.4% (95% confidence interval [CI], 2.0-25.2) and nine showed stable disease (SD), exhibiting a disease control rate (DCR) of 37.5% (95% CI, 21.1-56.3). For the 31 patients who had access to PD-1 ligand 1 (PD-L1) combined positive score (CPS) testing (cut-off value ≥1%), the ORR was not different between those who had PD-L1-positive (PD-L1+; 1/11, 9.1%) and PDL1-(2/20, 10.0%) tumors (<i>p</i> = 1.000). The tumor mutational burden (TMB) of PD-L1+ BTC was comparable to that of PD-L1-BTC (<i>p</i> = 0.630). TMB and any exonic somatic mutations were also not predictive of pembrolizumab response. Molecular analysis of blood and tumor samples demonstrated a relatively high natural killer (NK) cell proportion in the peripheral blood before pembrolizumab treatment in patients who achieved tumor response. Moreover, the tumors of these patients presented high enrichment scores for NK cells, antitumor cytokines, and Th1 signatures, and a low enrichment score for cancer-associated fibroblasts.</p><p><strong>Conclusions: </strong>This study shows the molecular characteristics associated with the efficacy of pembrolizumab in BTC of the MSS type.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 1","pages":"57-65"},"PeriodicalIF":2.0,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11671410/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142903381","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
STIL enhances the development of lung adenocarcinoma by regulating the glycolysis pathway. STIL通过调节糖酵解途径促进肺腺癌的发展。
IF 2 4区 医学 Q3 ONCOLOGY Pub Date : 2024-12-20 eCollection Date: 2025-01-01 DOI: 10.32604/or.2024.048562
Lei Wang, Xianjin Xie

Background: To investigate SCL/TAL 1 interrupting locus (STIL)'s role and prognostic significance in lung adenocarcinoma (LUAD) progression, we examined STIL and E2 promoter binding factor 1 (E2F1) expression and their impacts on LUAD prognosis using Gene Expression Profiling Interactive Analysis (GEPIA).

Methods: Functional assays including CCK-8, wound-healing, 5-ethynyl-2-deoxyuridine (EdU), Transwell assays, and flow cytometry, elucidated STIL and E2F1's effects on cell viability, proliferation, apoptosis, and migration. Gene set enrichment analysis (GSEA) identified potential pathways, while metabolic assays assessed glucose metabolism.

Results: Our findings reveal that STIL and E2F1 are overexpressed in LUAD, correlating with adverse outcomes. It enhances cell proliferation, migration, and invasion, and suppresses apoptosis, activating downstream of E2F1. Silencing E2F1 reversed the promotion effect of the STIL overexpression on cell viability and invasiveness. Importantly, STIL modulates glycolysis, influencing glucose consumption, lactate production, and energy balance in LUAD cells.

Conclusion: Our model, incorporating STIL, age, and disease stage, robustly predicts patient prognosis, underscored STIL's pivotal role in LUAD pathogenesis through metabolic reprogramming. This comprehensive approach not only confirms STIL's prognostic value but also highlights its potential as a therapeutic target in LUAD.

背景:为了研究SCL/TAL 1中断位点(STIL)在肺腺癌(LUAD)进展中的作用和预后意义,我们采用基因表达谱交互分析(GEPIA)检测了STIL和E2启动子结合因子1 (E2F1)的表达及其对LUAD预后的影响。方法:通过CCK-8、创面愈合、5-乙基-2-脱氧尿苷(EdU)、Transwell实验和流式细胞术等功能检测,阐明了STIL和E2F1对细胞活力、增殖、凋亡和迁移的影响。基因集富集分析(GSEA)确定了潜在的途径,而代谢试验评估了葡萄糖代谢。结果:我们的研究结果表明,STIL和E2F1在LUAD中过表达,与不良结局相关。它增强细胞增殖、迁移和侵袭,抑制凋亡,激活下游E2F1。沉默E2F1逆转了STIL过表达对细胞活力和侵袭性的促进作用。重要的是,STIL调节糖酵解,影响LUAD细胞的葡萄糖消耗、乳酸生成和能量平衡。结论:我们的模型结合了STIL、年龄和疾病分期,可以可靠地预测患者预后,强调了STIL通过代谢重编程在LUAD发病机制中的关键作用。这种综合方法不仅证实了STIL的预后价值,而且强调了其作为LUAD治疗靶点的潜力。
{"title":"<i>STIL</i> enhances the development of lung adenocarcinoma by regulating the glycolysis pathway.","authors":"Lei Wang, Xianjin Xie","doi":"10.32604/or.2024.048562","DOIUrl":"10.32604/or.2024.048562","url":null,"abstract":"<p><strong>Background: </strong>To investigate SCL/TAL 1 interrupting locus (<i>STIL</i>)'s role and prognostic significance in lung adenocarcinoma (LUAD) progression, we examined <i>STIL</i> and E2 promoter binding factor 1 (E2F1) expression and their impacts on LUAD prognosis using Gene Expression Profiling Interactive Analysis (GEPIA).</p><p><strong>Methods: </strong>Functional assays including CCK-8, wound-healing, 5-ethynyl-2-deoxyuridine (EdU), Transwell assays, and flow cytometry, elucidated <i>STIL</i> and E2F1's effects on cell viability, proliferation, apoptosis, and migration. Gene set enrichment analysis (GSEA) identified potential pathways, while metabolic assays assessed glucose metabolism.</p><p><strong>Results: </strong>Our findings reveal that <i>STIL</i> and E2F1 are overexpressed in LUAD, correlating with adverse outcomes. It enhances cell proliferation, migration, and invasion, and suppresses apoptosis, activating downstream of E2F1. Silencing E2F1 reversed the promotion effect of the <i>STIL</i> overexpression on cell viability and invasiveness. Importantly, <i>STIL</i> modulates glycolysis, influencing glucose consumption, lactate production, and energy balance in LUAD cells.</p><p><strong>Conclusion: </strong>Our model, incorporating <i>STIL</i>, age, and disease stage, robustly predicts patient prognosis, underscored <i>STIL</i>'s pivotal role in LUAD pathogenesis through metabolic reprogramming. This comprehensive approach not only confirms <i>STIL</i>'s prognostic value but also highlights its potential as a therapeutic target in LUAD.</p>","PeriodicalId":19537,"journal":{"name":"Oncology Research","volume":"33 1","pages":"123-132"},"PeriodicalIF":2.0,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11671407/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142903361","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Oncology Research
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1