Pub Date : 2024-11-11DOI: 10.1186/s12935-024-03547-3
Chih-Chia Yu, Hon-Yi Lin, Michael W Y Chan, Shu-Fen Wu, Wen-Yen Chiou, Moon-Sing Lee, Chen-Lin Chi, Ru-Inn Lin, Feng-Chun Hsu, Hsuan-Ju Yang, Liang-Cheng Chen, Chia-Hui Chew, Shih-Kai Hung
Purpose: We tested whether the PARP inhibitor, Olaparib, can effectively enhance radiosensitivity while inhibiting OSCC growth and metastasis in vitro and in vivo. Patient samples were used for survival validation.
Methods: The present study investigated the effect of Olaparib and ionizing radiation (IR) on clonogenic, migratory, and invasive ability in human IR-sensitive (OML1) and IR-resistant (OML1-R) OSCC cell lines. We next explored the underlying mechanism with ELISA and a Western blotting assay. Two in vivo mouse models were established to investigate the efficacy of Olaparib combined with radiotherapy (RT) on local tumor growth and lung metastasis. IL-17 A expression was confirmed in tissue specimens of OSCC patients by immunohistochemistry.
Results: We found that Olaparib, in combination with IR, substantially inhibited cell growth, migration, and invasion in vitro. Mechanistically, the Olaparib treatment significantly reduced the secretion of IL-17 A in irradiated OSCC cells by attenuating NF-κB and p38 activity. Consistently, Olaparib enhanced the radiosensitivity and, with RT, synergistically reduced both tumor growth and lung metastasis in mice. In addition, OSCC patients with high IL-17 A expression were substantially associated with an increased risk of lymph node involvement and worse survival.
Conclusions: This study has highlighted that Olaparib displays radiosensitizing and antimetastatic effects by inhibiting the IL-17 A-dependent signal. Remarkably, Olaparib could provide a remarkable anticancer efficacy to improve treatment response in OSCC patients with recurrent/metastatic disease after RT.
{"title":"Olaparib enhancing radiosensitization and anti-metastatic effect of oral cancer by targeting IL-17A signal.","authors":"Chih-Chia Yu, Hon-Yi Lin, Michael W Y Chan, Shu-Fen Wu, Wen-Yen Chiou, Moon-Sing Lee, Chen-Lin Chi, Ru-Inn Lin, Feng-Chun Hsu, Hsuan-Ju Yang, Liang-Cheng Chen, Chia-Hui Chew, Shih-Kai Hung","doi":"10.1186/s12935-024-03547-3","DOIUrl":"10.1186/s12935-024-03547-3","url":null,"abstract":"<p><strong>Purpose: </strong>We tested whether the PARP inhibitor, Olaparib, can effectively enhance radiosensitivity while inhibiting OSCC growth and metastasis in vitro and in vivo. Patient samples were used for survival validation.</p><p><strong>Methods: </strong>The present study investigated the effect of Olaparib and ionizing radiation (IR) on clonogenic, migratory, and invasive ability in human IR-sensitive (OML1) and IR-resistant (OML1-R) OSCC cell lines. We next explored the underlying mechanism with ELISA and a Western blotting assay. Two in vivo mouse models were established to investigate the efficacy of Olaparib combined with radiotherapy (RT) on local tumor growth and lung metastasis. IL-17 A expression was confirmed in tissue specimens of OSCC patients by immunohistochemistry.</p><p><strong>Results: </strong>We found that Olaparib, in combination with IR, substantially inhibited cell growth, migration, and invasion in vitro. Mechanistically, the Olaparib treatment significantly reduced the secretion of IL-17 A in irradiated OSCC cells by attenuating NF-κB and p38 activity. Consistently, Olaparib enhanced the radiosensitivity and, with RT, synergistically reduced both tumor growth and lung metastasis in mice. In addition, OSCC patients with high IL-17 A expression were substantially associated with an increased risk of lymph node involvement and worse survival.</p><p><strong>Conclusions: </strong>This study has highlighted that Olaparib displays radiosensitizing and antimetastatic effects by inhibiting the IL-17 A-dependent signal. Remarkably, Olaparib could provide a remarkable anticancer efficacy to improve treatment response in OSCC patients with recurrent/metastatic disease after RT.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"24 1","pages":"373"},"PeriodicalIF":5.3,"publicationDate":"2024-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11552144/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615321","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-11DOI: 10.1186/s12935-024-03563-3
Hui Shen, Qianrui Kou, Linxin Shao, Jing Zhang, Fang Li
Ubiquitination is a prevalent post-translational modification that plays a crucial role in a wide range of pathophysiological processes, including cell proliferation, apoptosis, autophagy, immune response, and DNA damage repair. Among the enzymes involved in ubiquitination, E3 ubiquitin ligases are particularly significant, serving as key regulators of numerous diseases, including tumours. This review focuses on HECW2 (HECT, C2, and WW domain-containing E3 ubiquitin protein ligase 2, also known as NEDL2), providing a comprehensive overview of its interactors and its pathological roles in tumorous cancer and other diseases. The insights gained from this review may contribute to the development of novel treatment strategies for various diseases, particularly tumours.
{"title":"E3 ubiquitin ligase HECW2: a promising target for tumour therapy.","authors":"Hui Shen, Qianrui Kou, Linxin Shao, Jing Zhang, Fang Li","doi":"10.1186/s12935-024-03563-3","DOIUrl":"10.1186/s12935-024-03563-3","url":null,"abstract":"<p><p>Ubiquitination is a prevalent post-translational modification that plays a crucial role in a wide range of pathophysiological processes, including cell proliferation, apoptosis, autophagy, immune response, and DNA damage repair. Among the enzymes involved in ubiquitination, E3 ubiquitin ligases are particularly significant, serving as key regulators of numerous diseases, including tumours. This review focuses on HECW2 (HECT, C2, and WW domain-containing E3 ubiquitin protein ligase 2, also known as NEDL2), providing a comprehensive overview of its interactors and its pathological roles in tumorous cancer and other diseases. The insights gained from this review may contribute to the development of novel treatment strategies for various diseases, particularly tumours.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"24 1","pages":"374"},"PeriodicalIF":5.3,"publicationDate":"2024-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11556196/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615306","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-11DOI: 10.1186/s12935-024-03566-0
Jieqiong Li, Nan Hao, Juan Han, Mi Zhang, Xiaomei Li, Nan Yang
{"title":"Retraction Note: ZKSCAN3 drives tumor metastasis via integrin β4/FAK/AKT mediated epithelial-mesenchymal transition in hepatocellular carcinoma.","authors":"Jieqiong Li, Nan Hao, Juan Han, Mi Zhang, Xiaomei Li, Nan Yang","doi":"10.1186/s12935-024-03566-0","DOIUrl":"10.1186/s12935-024-03566-0","url":null,"abstract":"","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"24 1","pages":"375"},"PeriodicalIF":5.3,"publicationDate":"2024-11-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11556177/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142635892","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-10DOI: 10.1186/s12935-024-03554-4
Yoel G Montoyo-Pujol, José J Ponce, Silvia Delgado-García, Tina A Martín, Hortensia Ballester, Elena Castellón-Molla, Angela Ramos-Montoya, Inmaculada Lozano-Cubo, J Miguel Sempere-Ortells, Gloria Peiró
Background: Breast cancer (BC) is the most common cancer in women and the leading cause of cancer-related death worldwide. This heterogeneous disease has been historically considered a non-immunogenic type of cancer. However, recent advances in immunotherapy have increased the interest in knowing the role of the immune checkpoints (IC) and other immune regulation pathways in this neoplasia.
Methods: In this retrospective study, we evaluated the correlation of mRNA expression of CTLA-4, PDCD1 (PD1), CD274 (PD-L1), PDCD1LG2 (PD-L2), CD276 (B7-H3), JAK2, and FOXO1 with clinicopathological factors and BC patient's outcome by real-time quantitative polymerase chain reaction (qPCR).
Results: Our results showed that immunoregulatory gene expression depends on BC immunophenotype being CTLA-4 and PDCD1 (PD1) overexpressed on triple-negative/basal-like (TN/BL) and luminal B/HER2-positive phenotypes, respectively, and CD276 (B7-H3), JAK2 and FOXO1 associated with both luminal A and luminal B/HER2-negative tumors. In addition, we found that these genes can also be related to aggressive and non-aggressive clinicopathological characteristics in BC. Finally, survival analysis showed that CTLA-4 expression levels emerge as a significant independent factor of good prognosis in BC patients, especially in the HER2-enriched subtype.
Conclusion: Considering all these data, we can conclude that the expression of immunoregulatory genes depends on tumor phenotype and has potential clinical implications in BC patients.
背景:乳腺癌(BC)是女性最常见的癌症,也是全球癌症相关死亡的主要原因。这种异质性疾病历来被认为是一种非免疫原性癌症。然而,免疫疗法的最新进展增加了人们了解免疫检查点(IC)和其他免疫调节途径在这种肿瘤中的作用的兴趣:在这项回顾性研究中,我们通过实时定量聚合酶链反应(qPCR)评估了CTLA-4、PDCD1(PD1)、CD274(PD-L1)、PDCD1LG2(PD-L2)、CD276(B7-H3)、JAK2和FOXO1的mRNA表达与临床病理因素和BC患者预后的相关性:结果表明,免疫调节基因的表达取决于BC免疫表型,CTLA-4和PDCD1(PD1)分别在三阴性/基底样(TN/BL)和管腔B/HER2阳性表型中过表达,而CD276(B7-H3)、JAK2和FOXO1与管腔A和管腔B/HER2阴性肿瘤相关。此外,我们还发现这些基因还与 BC 的侵袭性和非侵袭性临床病理特征有关。最后,生存分析表明,CTLA-4表达水平是BC患者预后良好的重要独立因素,尤其是在HER2富集亚型中:综合上述数据,我们可以得出结论:免疫调节基因的表达取决于肿瘤表型,对BC患者具有潜在的临床意义。
{"title":"High CTLA-4 gene expression is an independent good prognosis factor in breast cancer patients, especially in the HER2-enriched subtype.","authors":"Yoel G Montoyo-Pujol, José J Ponce, Silvia Delgado-García, Tina A Martín, Hortensia Ballester, Elena Castellón-Molla, Angela Ramos-Montoya, Inmaculada Lozano-Cubo, J Miguel Sempere-Ortells, Gloria Peiró","doi":"10.1186/s12935-024-03554-4","DOIUrl":"10.1186/s12935-024-03554-4","url":null,"abstract":"<p><strong>Background: </strong>Breast cancer (BC) is the most common cancer in women and the leading cause of cancer-related death worldwide. This heterogeneous disease has been historically considered a non-immunogenic type of cancer. However, recent advances in immunotherapy have increased the interest in knowing the role of the immune checkpoints (IC) and other immune regulation pathways in this neoplasia.</p><p><strong>Methods: </strong>In this retrospective study, we evaluated the correlation of mRNA expression of CTLA-4, PDCD1 (PD1), CD274 (PD-L1), PDCD1LG2 (PD-L2), CD276 (B7-H3), JAK2, and FOXO1 with clinicopathological factors and BC patient's outcome by real-time quantitative polymerase chain reaction (qPCR).</p><p><strong>Results: </strong>Our results showed that immunoregulatory gene expression depends on BC immunophenotype being CTLA-4 and PDCD1 (PD1) overexpressed on triple-negative/basal-like (TN/BL) and luminal B/HER2-positive phenotypes, respectively, and CD276 (B7-H3), JAK2 and FOXO1 associated with both luminal A and luminal B/HER2-negative tumors. In addition, we found that these genes can also be related to aggressive and non-aggressive clinicopathological characteristics in BC. Finally, survival analysis showed that CTLA-4 expression levels emerge as a significant independent factor of good prognosis in BC patients, especially in the HER2-enriched subtype.</p><p><strong>Conclusion: </strong>Considering all these data, we can conclude that the expression of immunoregulatory genes depends on tumor phenotype and has potential clinical implications in BC patients.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"24 1","pages":"371"},"PeriodicalIF":5.3,"publicationDate":"2024-11-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11552348/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615317","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-09DOI: 10.1186/s12935-024-03549-1
Xin Ge, Zichu Shen, Yuxin Yin
Long non-coding RNAs (lncRNAs) are emerging as crucial regulators of gene expression through diverse mechanisms, including regulation of protein localization, sequestration of miRNAs, recruitment of chromatin modifiers, and modulation of signaling pathways. Accumulating evidence highlights their pivotal roles in tumor initiation, progression, and the development of therapeutic resistance. In this review, we comprehensively summarized the existing literature to identify lncRNAs associated with treatment responses in non-small cell lung cancer (NSCLC). Specifically, we categorized these lncRNAs based on their mechanisms of action in mediating resistance to chemotherapy, targeted therapy, and radiotherapy. Our analysis revealed that aberrant expression of various lncRNAs contributes to the development, metastasis, and therapeutic resistance in NSCLC, ultimately leading to poor clinical outcomes. By elucidating the intricate mechanisms through which lncRNAs modulate therapeutic responses, this review aims to provide mechanistic insights into the heterogeneous treatment outcomes observed in NSCLC patients and unveil potential therapeutic targets for overcoming drug resistance.
{"title":"Comprehensive review of LncRNA-mediated therapeutic resistance in non-small cell lung cancer.","authors":"Xin Ge, Zichu Shen, Yuxin Yin","doi":"10.1186/s12935-024-03549-1","DOIUrl":"10.1186/s12935-024-03549-1","url":null,"abstract":"<p><p>Long non-coding RNAs (lncRNAs) are emerging as crucial regulators of gene expression through diverse mechanisms, including regulation of protein localization, sequestration of miRNAs, recruitment of chromatin modifiers, and modulation of signaling pathways. Accumulating evidence highlights their pivotal roles in tumor initiation, progression, and the development of therapeutic resistance. In this review, we comprehensively summarized the existing literature to identify lncRNAs associated with treatment responses in non-small cell lung cancer (NSCLC). Specifically, we categorized these lncRNAs based on their mechanisms of action in mediating resistance to chemotherapy, targeted therapy, and radiotherapy. Our analysis revealed that aberrant expression of various lncRNAs contributes to the development, metastasis, and therapeutic resistance in NSCLC, ultimately leading to poor clinical outcomes. By elucidating the intricate mechanisms through which lncRNAs modulate therapeutic responses, this review aims to provide mechanistic insights into the heterogeneous treatment outcomes observed in NSCLC patients and unveil potential therapeutic targets for overcoming drug resistance.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"24 1","pages":"369"},"PeriodicalIF":5.3,"publicationDate":"2024-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11549762/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615301","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-09DOI: 10.1186/s12935-024-03534-8
Tao Wang, Shu Wang, Zhuolin Li, Jie Xie, Kuiying Du, Jing Hou
Background: Breast cancer remains a leading cause of mortality among women worldwide, necessitating innovative prognostic models to enhance treatment strategies.
Methods: Our study retrospectively enrolled 9,439 breast cancer patients from 12 independent datasets and single-cell data from 12 patients (64,308 cells). Moverover, 30 in-house clinical cohort were collected for validation. We employed a comprehensive approach by combining ten distinct machine learning algorithms across 108 different combinations to scrutinize 88 pre-existing signatures of breast cancer. To affirm the efficacy of our developed model, immunohistochemistry assays were performed. Additionally, we investigated various potential immunotherapeutic and chemotherapeutic interventions.
Results: This study introduces an Artificial Intelligence-aided Redox Signature (AIARS) as a novel prognostic tool, leveraging machine learning to identify critical redox-related gene signatures in breast cancer. Our results demonstrate that AIARS significantly outperforms existing prognostic models in predicting breast cancer outcomes, offering a robust tool for personalized treatment planning. Validation through immunohistochemistry assays on samples from 30 patients corroborated our results, underscoring the model's applicability on a wider scale. Furthermore, the analysis revealed that patients with low AIARS expression levels are more responsive to immunotherapy. Conversely, those exhibiting high AIARS were found to be more susceptible to certain chemotherapeutic agents, including vincristine.
Conclusions: Our study underscores the importance of redox biology in breast cancer prognosis and introduces a powerful machine learning-based tool, the AIARS, for personalized treatment strategies. By providing a more nuanced understanding of the redox landscape in breast cancer, the AIARS paves the way for the development of redox-targeted therapies, promising to enhance patient outcomes significantly. Future work will focus on clinical validation and exploring the mechanistic roles of identified genes in cancer biology.
{"title":"Machine learning unveils key Redox signatures for enhanced breast Cancer therapy.","authors":"Tao Wang, Shu Wang, Zhuolin Li, Jie Xie, Kuiying Du, Jing Hou","doi":"10.1186/s12935-024-03534-8","DOIUrl":"10.1186/s12935-024-03534-8","url":null,"abstract":"<p><strong>Background: </strong>Breast cancer remains a leading cause of mortality among women worldwide, necessitating innovative prognostic models to enhance treatment strategies.</p><p><strong>Methods: </strong>Our study retrospectively enrolled 9,439 breast cancer patients from 12 independent datasets and single-cell data from 12 patients (64,308 cells). Moverover, 30 in-house clinical cohort were collected for validation. We employed a comprehensive approach by combining ten distinct machine learning algorithms across 108 different combinations to scrutinize 88 pre-existing signatures of breast cancer. To affirm the efficacy of our developed model, immunohistochemistry assays were performed. Additionally, we investigated various potential immunotherapeutic and chemotherapeutic interventions.</p><p><strong>Results: </strong>This study introduces an Artificial Intelligence-aided Redox Signature (AIARS) as a novel prognostic tool, leveraging machine learning to identify critical redox-related gene signatures in breast cancer. Our results demonstrate that AIARS significantly outperforms existing prognostic models in predicting breast cancer outcomes, offering a robust tool for personalized treatment planning. Validation through immunohistochemistry assays on samples from 30 patients corroborated our results, underscoring the model's applicability on a wider scale. Furthermore, the analysis revealed that patients with low AIARS expression levels are more responsive to immunotherapy. Conversely, those exhibiting high AIARS were found to be more susceptible to certain chemotherapeutic agents, including vincristine.</p><p><strong>Conclusions: </strong>Our study underscores the importance of redox biology in breast cancer prognosis and introduces a powerful machine learning-based tool, the AIARS, for personalized treatment strategies. By providing a more nuanced understanding of the redox landscape in breast cancer, the AIARS paves the way for the development of redox-targeted therapies, promising to enhance patient outcomes significantly. Future work will focus on clinical validation and exploring the mechanistic roles of identified genes in cancer biology.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"24 1","pages":"368"},"PeriodicalIF":5.3,"publicationDate":"2024-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11549853/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142615318","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-11-08DOI: 10.1186/s12935-024-03561-5
Rui Hou, Luo Jiang
{"title":"Retraction Note: LINC00115 promotes stemness and inhibits apoptosis of ovarian cancer stem cells by upregulating SOX9 and inhibiting the Wnt/β-catenin pathway through competitively binding to microRNA-30a.","authors":"Rui Hou, Luo Jiang","doi":"10.1186/s12935-024-03561-5","DOIUrl":"10.1186/s12935-024-03561-5","url":null,"abstract":"","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"24 1","pages":"367"},"PeriodicalIF":5.3,"publicationDate":"2024-11-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11545768/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142635889","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Glioblastoma (GBM) is the most lethal and common primary tumor of central nervous system with a poor prognosis. Glioma stem cells (GSCs) are particularly significant in GBM proliferation, invasion, self-renewal and recurrence. Circular RNAs (circRNAs) play important roles in various physiological and pathological processes, including regulating the biological behavior of GBM. Therefore, discovering novel circRNAs related to GSCs may contribute to a promising approach for treatment of GBM. Herein, we find out a novel circRNA termed circZEB1 with a high expression in glioma. Limb-bud and heart (LBH) is a transcription cofactor and promotes glioma stem cell tumorigenicity in our study. Mechanistically, circZEB1 can upregulate the expression of transcription cofactor LBH via sponging miR-128-3p in GSCs. LBH can facilitate the expression of tumor necrosis factor-α (TNF-α), thus activating the NF-κB signaling pathway to promote the glioma progression. Meanwhile, LBH can also upregulate the RNA binding protein Fused in Sarcoma (FUS) expression, which can bind to and maintain the stability of circZEB1. A positive feedback loop is formed among FUS, circZEB1, miR-128-3p and LBH in GSCs. Our study uncovers a critical role of circZEB1 and provides a novel biomarker for treating GBM.
{"title":"FUS/circZEB1/miR-128-3p/LBH feedback loop contributes to the malignant phenotype of GSCs via TNF-α-mediated NF-κB signaling pathway.","authors":"Guoqing Zhang, Yang Jiang, Zhichao Wang, Zhengting Guo, Jinpeng Hu, Xinqiao Li, Yongfeng Wang, Zhitao Jing","doi":"10.1186/s12935-024-03526-8","DOIUrl":"10.1186/s12935-024-03526-8","url":null,"abstract":"<p><p>Glioblastoma (GBM) is the most lethal and common primary tumor of central nervous system with a poor prognosis. Glioma stem cells (GSCs) are particularly significant in GBM proliferation, invasion, self-renewal and recurrence. Circular RNAs (circRNAs) play important roles in various physiological and pathological processes, including regulating the biological behavior of GBM. Therefore, discovering novel circRNAs related to GSCs may contribute to a promising approach for treatment of GBM. Herein, we find out a novel circRNA termed circZEB1 with a high expression in glioma. Limb-bud and heart (LBH) is a transcription cofactor and promotes glioma stem cell tumorigenicity in our study. Mechanistically, circZEB1 can upregulate the expression of transcription cofactor LBH via sponging miR-128-3p in GSCs. LBH can facilitate the expression of tumor necrosis factor-α (TNF-α), thus activating the NF-κB signaling pathway to promote the glioma progression. Meanwhile, LBH can also upregulate the RNA binding protein Fused in Sarcoma (FUS) expression, which can bind to and maintain the stability of circZEB1. A positive feedback loop is formed among FUS, circZEB1, miR-128-3p and LBH in GSCs. Our study uncovers a critical role of circZEB1 and provides a novel biomarker for treating GBM.</p>","PeriodicalId":9385,"journal":{"name":"Cancer Cell International","volume":"24 1","pages":"365"},"PeriodicalIF":5.3,"publicationDate":"2024-11-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11545228/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142602812","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}