1
|
Wang W, Liu Y, Wu J. The roles of lncRNAs in the development of drug resistance of oral cancers. Biomed Pharmacother 2024; 180:117458. [PMID: 39413618 DOI: 10.1016/j.biopha.2024.117458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/11/2024] [Accepted: 09/19/2024] [Indexed: 10/18/2024] Open
Abstract
Oral cancers are a significant global health concern, with a high incidence of treatment failure primarily due to the development of drug resistance. Long non-coding RNAs (lncRNAs) have emerged as critical regulators of gene expression, playing pivotal roles in various cellular processes, including tumor progression and response to therapy. This review explores the multifaceted roles of lncRNAs in the development of drug resistance in oral cancers. We highlight the mechanisms by which lncRNAs modulate drug efflux, apoptosis, epithelial-mesenchymal transition (EMT), and other pathways associated with chemoresistance. Key lncRNAs implicated in resistance to commonly used chemotherapeutic agents in oral cancers are discussed, along with their potential as therapeutic targets. Understanding the involvement of lncRNAs in drug resistance mechanisms offers promising avenues for overcoming treatment barriers and improving patient outcomes. This review underscores the need for further research to elucidate the precise roles of lncRNAs in oral cancer resistance and their translation into clinical interventions.
Collapse
Affiliation(s)
- Wenjing Wang
- Department of Stomatology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 43400, China
| | - Yi Liu
- Department of Stomatology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 43400, China
| | - Jianan Wu
- Experimental and Practical Teaching Center, Hubei College of Chinese Medicine, Jingzhou, Hubei 434000, China.
| |
Collapse
|
2
|
Chen KL, Huang SW, Yao JJ, He SW, Gong S, Tan XR, Liang YL, Li JY, Huang SY, Li YQ, Zhao Y, Qiao H, Xu S, Zang S, Ma J, Liu N. LncRNA DYNLRB2-AS1 promotes gemcitabine resistance of nasopharyngeal carcinoma by inhibiting the ubiquitination degradation of DHX9 protein. Drug Resist Updat 2024; 76:101111. [PMID: 38908233 DOI: 10.1016/j.drup.2024.101111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/24/2024]
Abstract
Gemcitabine (GEM) based induction chemotherapy is a standard treatment for locoregionally advanced nasopharyngeal carcinoma (NPC). However, approximately 15 % of patients are still resistant to GEM-containing chemotherapy, which leads to treatment failure. Nevertheless, the underlying mechanisms of GEM resistance remain poorly understood. Herein, based on a microarray analysis, we identified 221 dysregulated lncRNAs, of which, DYNLRB2-AS1 was one of the most upregulated lncRNAs in GEM-resistance NPC cell lines. DYNLRB2-AS1 was shown to function as contain an oncogenic lncRNA that promoted NPC GEM resistance, cell proliferation, but inhibited cell apoptosis. Mechanistically, DYNLRB2-AS1 could directly bind to the DHX9 protein and prevent its interaction with the E3 ubiquitin ligase PRPF19, and thus blocking PRPF19-mediated DHX9 degradation, which ultimately facilitated the repair of DNA damage in the presence of GEM. Clinically, higher DYNLRB2-AS1 expression indicated an unfavourable overall survival of NPC patients who received induction chemotherapy. Overall, this study identified the oncogenic lncRNA DYNLRB2-AS1 as an independent prognostic biomarker for patients with locally advanced NPC and as a potential therapeutic target for overcoming GEM chemoresistance in NPC.
Collapse
Affiliation(s)
- Kai-Lin Chen
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Sai-Wei Huang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ji-Jin Yao
- Department of Head and Neck Oncology, the Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Shi-Wei He
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Sha Gong
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xi-Rong Tan
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ye-Lin Liang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jun-Yan Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Sheng-Yan Huang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ying-Qin Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yin Zhao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Han Qiao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Sha Xu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Shengbing Zang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| | - Jun Ma
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| | - Na Liu
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| |
Collapse
|
3
|
Chen HB, Gong XY, Shen WH, Zhu ZH, Chen X. Exosomal AC068768.1 enhances the proliferation, migration, and invasion of laryngeal squamous cell carcinoma through miR-139-5p/NOTCH1 axis. Heliyon 2024; 10:e36358. [PMID: 39258189 PMCID: PMC11386030 DOI: 10.1016/j.heliyon.2024.e36358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/12/2024] Open
Abstract
Objective Long non-coding RNAs (lncRNAs) are closely associated with the pathogenesis of laryngeal squamous cell carcinoma (LSCC). This study aimed to investigate the roles of AC068768.1 in LSCC. Methods Exosomes were extracted by ultracentrifugation and identified by transmission electron microscopy (TEM) assay. The expression levels of mRNA and miRNA were determined by real-time quantitative polymerase chain reaction (RT-qPCR). Cellular functions were assesses through immunofluorescence, flow cytometry, colony formation, wound healing and transwell assays. Chromatin immunoprecipitation (ChIP) and luciferase assays were conducted to verify the binding of AC068768.1 by signal transducer and activator of transcription 3 (STAT3). Xenograft assays were performed to confirm the roles of AC068768.1 in LSCC, and hematoxylin-eosin (HE) staining was applied for histological analysis. Results LSCC cell-derived exosomes induced M2-like tumor-associated macrophages (TAM2) polarization, which promoted the proliferation, migration, and invasion of LSCCs. Knockdown of exosomal AC068768.1 inhibited M2 polarization and suppressed LSCC aggressiveness both in vitro and in vivo. Moreover, AC068768.1 sponged miR-139-5p, inducing the upregulation of neurogenic locus notch homolog protein 1 (NOTCH1). LSCCs adapted to TAM2 polarization in the tumor microenvironment via AC068768.1-mediated activation of the NOTCH1 pathway. Additionally, NOTCH1 activated STAT3. Conclusion The AC068768.1/miR-139-5p/NOTCH1/STAT3 axis promotes the metastasis of LSCC. This finding may provide a novel target for LSCC therapy.
Collapse
Affiliation(s)
- Hai-Bin Chen
- Department of Otorhinolaryngology, The First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Xiao-Yang Gong
- Department of Otorhinolaryngology, The First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Wen-Hao Shen
- Department of Otorhinolaryngology, The First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Zi-Hang Zhu
- Department of Otorhinolaryngology, The First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - Xi Chen
- Department of Otorhinolaryngology, The First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, China
| |
Collapse
|
4
|
Wang Q, Li H, Wu T, Yu B, Cong H, Shen Y. Nanodrugs based on co-delivery strategies to combat cisplatin resistance. J Control Release 2024; 370:14-42. [PMID: 38615892 DOI: 10.1016/j.jconrel.2024.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 03/24/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
Cisplatin (CDDP), as a broad-spectrum anticancer drug, is able to bind to DNA and inhibit cell division. Despite the widespread use of cisplatin since its discovery, cisplatin resistance developed during prolonged chemotherapy, similar to other small molecule chemotherapeutic agents, severely limits its clinical application. Cisplatin resistance in cancer cells is mainly caused by three reasons: DNA repair, decreased cisplatin uptake/increased efflux, and cisplatin inactivation. In earlier combination therapies, the emergence of multidrug resistance (MDR) in cancer cells prevented the achievement of the desired therapeutic effect even with the accurate combination of two chemotherapeutic drugs. Therefore, combination therapy using nanocarriers for co-delivery of drugs is considered to be ideal for alleviating cisplatin resistance and reducing cisplatin-related toxicity in cancer cells. This article provides an overview of the design of cisplatin nano-drugs used to combat cancer cell resistance, elucidates the mechanisms of action of cisplatin and the pathways through which cancer cells develop resistance, and finally discusses the design of drugs and related carriers that can synergistically reduce cancer resistance when combined with cisplatin.
Collapse
Affiliation(s)
- Qiubo Wang
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Hui Li
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Taixia Wu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China
| | - Bing Yu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China.
| | - Hailin Cong
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China; School of Materials Science and Engineering, Shandong University of Technology, Zibo 255000, China.
| | - Youqing Shen
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao 266071, China; Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bio-nanoengineering, and Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| |
Collapse
|
5
|
Kim SH, Kim HJ, Kim YJ, Kim YH, Park HR. LncRNA EIF3J-DT promotes chemoresistance in oral squamous cell carcinoma. Oral Dis 2024. [PMID: 38817073 DOI: 10.1111/odi.14987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 04/23/2024] [Accepted: 04/29/2024] [Indexed: 06/01/2024]
Abstract
OBJECTIVES This study aimed to screen oral squamous cell carcinoma (OSCC) diagnostic and prognostic candidates and investigate the potential functions and mechanisms of candidates in the chemoresistance of OSCC cell lines. MATERIALS AND METHODS Differential expression profiling of lncRNA was performed in a large cohort of OSCC patients from the Cancer Genome Atlas database to identify OSCC diagnostic and prognostic candidates. Taxol resistance in OSCC cell lines was analyzed using MTT assay. OSCC cell lines transfected with EIF3J-DT pcDNA or siRNA were used to determine its regulatory effects on apoptosis, cell cycle distribution and autophagy using flow cytometry and western blot. RESULTS We identified EIF3J-DT as a candidate for OSCC diagnosis and prognosis. The expression level of EIF3J-DT in OSCC cell lines correlates with taxol resistance. EIF3J-DT silencing attenuated taxol resistance, and EIF3J-DT overexpression enhanced taxol resistance in OSCC cell lines. Silencing of EIF3J-DT reduced taxol resistance by inducing apoptosis, cell cycle arrest, and ATG14-mediated autophagy inhibition in OSCC cell lines. CONCLUSIONS We found that EIF3J-DT induced chemoresistance by regulating apoptosis, cell cycle, and autophagy in OSCC cell lines, which EIF3J-DT might provide a novel therapeutic approach for OSCC as well as a diagnostic and prognostic factor.
Collapse
Affiliation(s)
- Seon Hyun Kim
- Periodontal Disease Signaling Network Research Center (MRC), School of Dentistry, Pusan National University, Yangsan, Korea
| | - Hye Jung Kim
- Periodontal Disease Signaling Network Research Center (MRC), School of Dentistry, Pusan National University, Yangsan, Korea
| | - Yeong Joo Kim
- Interdisciplinary Program of Genomic Data Science, Pusan National University, Yangsan, Korea
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Korea
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan, Korea
| | - Yun Hak Kim
- Periodontal Disease Signaling Network Research Center (MRC), School of Dentistry, Pusan National University, Yangsan, Korea
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Korea
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan, Korea
| | - Hae Ryoun Park
- Periodontal Disease Signaling Network Research Center (MRC), School of Dentistry, Pusan National University, Yangsan, Korea
- Department of Oral Pathology, Dental & Life Science Institute, School of Dentistry, Pusan National University, Yangsan, Korea
| |
Collapse
|
6
|
Wu YL, Liu W, Zhao T, Jin J. P4HA2 contributes to head and neck squamous cell carcinoma progression and EMT through PI3K/AKT signaling pathway. Med Oncol 2024; 41:163. [PMID: 38777998 PMCID: PMC11111551 DOI: 10.1007/s12032-024-02358-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/14/2024] [Indexed: 05/25/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) can be defined as a deadly illness with a dismal prognosis in advanced stages. Therefore, we seek to examine P4HA2 expression and effect in HNSCC, along with the underlying mechanisms. This study utilized integrated bioinformatics analyses to evaluate the P4HA2 expression pattern, prognostic implication, and probable function in HNSCC. The study conducted various in vitro experiments, including colony formation, CCK-8, flow cytometry, wound healing, and transwell assays, on the human HNSCC cell line CAL-27 to examine the involvement of P4HA2 in HNSCC progression. Moreover, western blotting was used to investigate epithelial-mesenchymal transition (EMT) markers and PI3K/AKT pathway markers to elucidate the underlying mechanisms. P4HA2 expression was significantly enhanced in HNSCC, and its overexpression was correlated to tumor aggressiveness and a poor prognosis in patients. Based on in vitro experiments, the overexpressed P4HA2 enhanced cell proliferation, migration, invasion, as well as EMT while reducing apoptosis, whereas P4HA2 silencing exhibited the reverse effect. P4HA2 overexpression enhanced PI3K/AKT phosphorylation in HNSCC cells. Moreover, LY294002 was observed to counteract the effects of upregulated P4HA2 on proliferation, migration, invasion, and EMT in HNSCC. Collectively, we indicated that P4HA2 promoted HNSCC progression and EMT via PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Yan-Ling Wu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, Guangdong, China
| | - Wan Liu
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, Guangdong, China
| | - Tingting Zhao
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, Guangdong, China
| | - Jing Jin
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, Guangdong, China.
| |
Collapse
|
7
|
Zhou J, Zheng H, Zhang H, Yu W, Li B, Ye L, Wang L. MCM5 is a Novel Therapeutic Target for Glioblastoma. Onco Targets Ther 2024; 17:371-381. [PMID: 38765057 PMCID: PMC11100520 DOI: 10.2147/ott.s457600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/08/2024] [Indexed: 05/21/2024] Open
Abstract
Objective MCM5 is a DNA licensing factor involved in cell proliferation and has been previously established as an excellent biomarker in a number of malignancies. Nevertheless, the role of MCM5 in GBM has not been fully clarified. The present study aimed to investigate the potential roles of MCM5 in the treatment of GBM and to elucidate its underlying mechanism, which is beneficial for developing new therapeutic strategies and predicting prognosis. Methods Firstly, we obtained transcriptomic and proteomic data from the TCGA and CPTAC databases on glioma patients. Employing the DeSeq2 R package, we then identified genes with joint differential expression in GBM tissues subjected to chemotherapy. To develop a prognostic risk score model, we performed univariate and multivariate Cox regression analyses. In vitro knockdown and overexpression of MCM5 were used to further investigate the biological functions of GBM cells. Additionally, we also delved into the upstream regulation of MCM5, revealing associations with several transcription factors. Finally, we investigated differences in immune cell infiltration and drug sensitivity across diverse risk groups identified in the prognostic risk model. Results In this study, the chemotherapy-treated GBM samples exhibited consistent alterations in 46 upregulated and 94 downregulated genes at both the mRNA and protein levels. Notably, MCM5 emerged as a gene with prognostic significance as well as potential therapeutic relevance. In vitro experiments subsequently validated the role of increased MCM5 expression in promoting GBM cell proliferation and resistance to TMZ. Correlations with transcription factors such as CREB1, CTCF, NFYB, NRF1, PBX1, TEAD1, and USF1 were discovered during upstream regulatory analysis, enriching our understanding of MCM5 regulatory mechanisms. The study additionally delves into immune cell infiltration and drug sensitivity, providing valuable insights for personalized treatment approaches. Conclusion This study identifies MCM5 as a key player in GBM, demonstrating its prognostic significance and potential therapeutic relevance by elucidating its role in promoting cell proliferation and resistance to chemotherapy.
Collapse
Affiliation(s)
- Jian Zhou
- Hyperbaric Oxygen Department, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Medical School, Shenzhen, 518055, People’s Republic of China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, People’s Republic of China
| | - Housheng Zheng
- Hyperbaric Oxygen Department, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Medical School, Shenzhen, 518055, People’s Republic of China
| | - Huiru Zhang
- Hyperbaric Oxygen Department, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Medical School, Shenzhen, 518055, People’s Republic of China
| | - Wenqiang Yu
- Hyperbaric Oxygen Department, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Medical School, Shenzhen, 518055, People’s Republic of China
| | - Baoer Li
- Hyperbaric Oxygen Department, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Medical School, Shenzhen, 518055, People’s Republic of China
| | - Liang Ye
- Hyperbaric Oxygen Department, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Medical School, Shenzhen, 518055, People’s Republic of China
| | - Lu Wang
- Hyperbaric Oxygen Department, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Medical School, Shenzhen, 518055, People’s Republic of China
| |
Collapse
|
8
|
Gong H, Liu Z, Yuan C, Luo Y, Chen Y, Zhang J, Cui Y, Zeng B, Liu J, Li H, Deng Z. Identification of cuproptosis-related lncRNAs with the significance in prognosis and immunotherapy of oral squamous cell carcinoma. Comput Biol Med 2024; 171:108198. [PMID: 38417385 DOI: 10.1016/j.compbiomed.2024.108198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 01/22/2024] [Accepted: 02/18/2024] [Indexed: 03/01/2024]
Abstract
Cuproptosis, a recently characterized programmed cell death mechanism, has emerged as a potential contributor to tumorigenesis, metastasis, and immune modulation. Long non-coding RNAs (lncRNAs) have demonstrated diverse regulatory roles in cancer and hold promise as biomarkers. However, the involvement and prognostic significance of cuproptosis-related lncRNAs (CRLs) in oral squamous cell carcinoma (OSCC) remain poorly understood. Based on TCGA-OSCC data, we integrated single-sample gene set enrichment analysis (ssGSEA), the LASSO algorithm, and the tumor immune dysfunction and exclusion (TIDE) algorithm. We identified 11 CRLs through differential expression, Spearman correlation, and univariate Cox regression analyses. Two distinct CRL-related subtypes were unveiled, delineating divergent survival patterns, tumor microenvironments (TME), and mutation profiles. A robust CRL-based signature (including AC107027.3, AC008011.2, MYOSLID, AC005785.1, AC019080.5, AC020558.2, AC025265.1, FAM27E3, and LINC02367) prognosticated OSCC outcomes, immunotherapy responses, and anti-tumor strategies. Superior predictive power compared to other lncRNA models was demonstrated. Functional assessments confirmed the influence of FAM27E3, LINC02367, and MYOSLID knockdown on OSCC cell behaviors. Remarkably, the CRLs-based signature maintained stability across OSCC patient subgroups, underscoring its clinical potential for survival prediction. This study elucidates CRLs' roles in TME of OSCC and establishes a potential signature for precision therapy.
Collapse
Affiliation(s)
- Han Gong
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Zhaolong Liu
- Hunan Key Laboratory of Oral Health Research, Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Chunhui Yuan
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Ying Luo
- Hunan Key Laboratory of Oral Health Research, Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Yuhan Chen
- Hunan Key Laboratory of Oral Health Research, Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Junyi Zhang
- Hunan Key Laboratory of Oral Health Research, Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Yiteng Cui
- Hunan Key Laboratory of Oral Health Research, Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Bin Zeng
- School of Stomatology, Changsha Medical University, Changsha, Hunan, China
| | - Jing Liu
- Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Hui Li
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Zhiyuan Deng
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; Hunan Key Laboratory of Oral Health Research, Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China; School of Stomatology, Changsha Medical University, Changsha, Hunan, China.
| |
Collapse
|
9
|
Yu Y, Wang J, Guo Q, Luo H. LINC01134: a pivotal oncogene with promising predictive maker and therapeutic target in hepatocellular carcinoma. Front Oncol 2024; 14:1265762. [PMID: 38450182 PMCID: PMC10915649 DOI: 10.3389/fonc.2024.1265762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 01/29/2024] [Indexed: 03/08/2024] Open
Abstract
Hepatocellular carcinoma (HCC) represents a leading and fatal malignancy within the gastrointestinal tract. Recent advancements highlight the pivotal role of long non-coding RNAs (lncRNAs) in diverse biological pathways and pathologies, particularly in tumorigenesis. LINC01134, a particular lncRNA, has attracted considerable attention due to its oncogenic potential in hepatoma. Current research underscores LINC01134's potential in augmenting the onset and progression of HCC, with notable implications in drug resistance. This review comprehensively explores the molecular functions and regulatory mechanisms of LINC01134 in HCC, offering a fresh perspective for therapeutic interventions. By delving into LINC01134's multifaceted roles, we aim to foster novel strategies in HCC management.
Collapse
Affiliation(s)
- Yutian Yu
- Department of Spleen and Stomach Diseases, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, China
| | - Jialing Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Qingfa Guo
- Second Clinical Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Hongliang Luo
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
10
|
Park SJ, Greer PL, Lee N. From odor to oncology: non-canonical odorant receptors in cancer. Oncogene 2024; 43:304-318. [PMID: 38087050 DOI: 10.1038/s41388-023-02908-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 01/31/2024]
Abstract
Odorant receptors, traditionally associated with olfaction as chemoreceptors, have been increasingly recognized for their presence and diverse functions in various non-nasal tissues throughout the body. Beyond their roles in sensory perception, emerging evidence suggests a compelling interplay between odorant receptors and cancer progression as well. Alongside the canonical GPCR odorant receptors, dysregulation of non-canonical odorant receptors such as trace amine-associated receptors (TAARs), formyl peptide receptors (FPRs), and membrane-spanning 4A family (MS4As) has been observed in various cancer types, suggesting their contributions to cancer progression. The roles of these non-canonical chemoreceptors in cancer are complex, with some receptors promoting tumorigenesis and others acting as tumor-suppressing factors upon activation, depending on the cancer type. These findings shed light on the potential of non-canonical odorant receptors as therapeutic targets and prognostic markers in cancer, inviting further exploration to unravel their precise mechanisms of action and implications in cancer biology. In this review, we provide a comprehensive overview of the intricate relationships between these chemoreceptors and various types of cancer, potentially paving the way for innovative odor-based therapeutics. Ultimately, this review discusses the potential development of novel therapeutic strategies targeting these non-canonical chemoreceptors.
Collapse
Affiliation(s)
- Sung Jin Park
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Paul L Greer
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Namgyu Lee
- Department of Biomedical Science and Engineering, Dankook University, Cheonan, 31116, Republic of Korea.
| |
Collapse
|
11
|
Wang R, Yang Y, Wang L, Shi Q, Ma H, He S, Feng L, Fang J. SOX2-OT Binds with ILF3 to Promote Head and Neck Cancer Progression by Modulating Crosstalk between STAT3 and TGF-β Signaling. Cancers (Basel) 2023; 15:5766. [PMID: 38136312 PMCID: PMC10742126 DOI: 10.3390/cancers15245766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 12/01/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Long non-coding RNA (lncRNA) is involved in the progression of head and neck squamous cell carcinoma (HNSCC). The molecular mechanism of lncRNA SOX2-OT in HNSCC remains unclear. Therefore, we aimed to elucidate the oncogenic role of SOX2-OT in HNSCC. QRT-PCR analysis was performed in 61 pairs of HNSCC cancer tissues, adjacent normal tissues, and 68 plasma samples confirmed that lncRNA SOX2-OT was overexpressed in cancer tissues and plasma samples, which served as a poor prognostic factor for HNSCC. The FISH assay demonstrated that SOX2-OT was localized in the nucleus and cytoplasm of HNSCC cell lines. Further, the cell function assay confirmed that SOX2-OT promoted cell proliferation and metastasis in vitro and in vivo. RNA pulldown and RIP assay results revealed that SOX2-OT bonds with ILF3 in HNSCC, and the rescue assay confirmed that SOX2-OT played an oncogenic role depending on ILF3 protein expression. Ingenuity pathway analysis and Western blotting indicated that SOX2-OT regulated HNSCC progression by promoting STAT3 phosphorylation and modulating the crosstalk between STAT3 and TGF-β signaling. These results reveal evidence for the role of SOX2-OT in HNSCC progression and metastasis by binding to ILF3, which may serve as a therapeutic target and prognostic biomarker in HNSCC.
Collapse
Affiliation(s)
- Ru Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Tong Ren Hospital, Capital Medical University, 1 Dongjiaominxiang Street, Beijing 100730, China; (R.W.); (Y.Y.); (L.W.); (Q.S.); (H.M.); (S.H.); (L.F.)
- Key Laboratory of Otorhinolaryngology, Head and Neck Surgery, Beijing Institute of Otorhinolaryngology, Beijing 100730, China
| | - Yifan Yang
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Tong Ren Hospital, Capital Medical University, 1 Dongjiaominxiang Street, Beijing 100730, China; (R.W.); (Y.Y.); (L.W.); (Q.S.); (H.M.); (S.H.); (L.F.)
- Key Laboratory of Otorhinolaryngology, Head and Neck Surgery, Beijing Institute of Otorhinolaryngology, Beijing 100730, China
| | - Lingwa Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Tong Ren Hospital, Capital Medical University, 1 Dongjiaominxiang Street, Beijing 100730, China; (R.W.); (Y.Y.); (L.W.); (Q.S.); (H.M.); (S.H.); (L.F.)
- Key Laboratory of Otorhinolaryngology, Head and Neck Surgery, Beijing Institute of Otorhinolaryngology, Beijing 100730, China
| | - Qian Shi
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Tong Ren Hospital, Capital Medical University, 1 Dongjiaominxiang Street, Beijing 100730, China; (R.W.); (Y.Y.); (L.W.); (Q.S.); (H.M.); (S.H.); (L.F.)
- Key Laboratory of Otorhinolaryngology, Head and Neck Surgery, Beijing Institute of Otorhinolaryngology, Beijing 100730, China
| | - Hongzhi Ma
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Tong Ren Hospital, Capital Medical University, 1 Dongjiaominxiang Street, Beijing 100730, China; (R.W.); (Y.Y.); (L.W.); (Q.S.); (H.M.); (S.H.); (L.F.)
- Key Laboratory of Otorhinolaryngology, Head and Neck Surgery, Beijing Institute of Otorhinolaryngology, Beijing 100730, China
| | - Shizhi He
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Tong Ren Hospital, Capital Medical University, 1 Dongjiaominxiang Street, Beijing 100730, China; (R.W.); (Y.Y.); (L.W.); (Q.S.); (H.M.); (S.H.); (L.F.)
- Key Laboratory of Otorhinolaryngology, Head and Neck Surgery, Beijing Institute of Otorhinolaryngology, Beijing 100730, China
| | - Ling Feng
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Tong Ren Hospital, Capital Medical University, 1 Dongjiaominxiang Street, Beijing 100730, China; (R.W.); (Y.Y.); (L.W.); (Q.S.); (H.M.); (S.H.); (L.F.)
- Key Laboratory of Otorhinolaryngology, Head and Neck Surgery, Beijing Institute of Otorhinolaryngology, Beijing 100730, China
| | - Jugao Fang
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Tong Ren Hospital, Capital Medical University, 1 Dongjiaominxiang Street, Beijing 100730, China; (R.W.); (Y.Y.); (L.W.); (Q.S.); (H.M.); (S.H.); (L.F.)
- Key Laboratory of Otorhinolaryngology, Head and Neck Surgery, Beijing Institute of Otorhinolaryngology, Beijing 100730, China
| |
Collapse
|
12
|
Zhang X, Chen X, Sun D, Song N, Li M, Zheng W, Yu Y, Ding G, Jiang Y. ENAH-202 promotes cancer progression in oral squamous cell carcinoma by regulating ZNF502/VIM axis. Cancer Med 2023; 12:20892-20905. [PMID: 37902191 PMCID: PMC10709750 DOI: 10.1002/cam4.6652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/31/2023] Open
Abstract
BACKGROUND We aimed to demonstrate the regulatory effect of long non-coding RNA (lncRNA) ENAH-202 on oral squamous cell carcinoma (OSCC) development as well as its molecular mechanism. METHODS We detected ENAH-202 expression in OSCC tissues and cell lines by quantitative real-time PCR (qPCR). The biological function of ENAH-202 was assessed in vitro and in vivo using CCK-8, colony formation assays, transwell assays, xenograft formation, and tail vein injection. The further molecular mechanism by which ENAH-202 promoted OSCC progression was identified using RNA pull-down, LS-MS/MS analysis, RNA immunoprecipitation (RIP), and chromatin immunoprecipitation (ChIP) assays. RESULTS ENAH-202 was significantly upregulated in OSCC tissues and cells. ENAH-202 promoted OSCC cell proliferation, migration, and invasion in vitro and in vivo. The expression of enabled homolog (ENAH) and epithelial-to-mesenchymal transition (EMT)-related proteins was changed with the expression of ENAH-202. Moreover, ENAH-202 promoted the transcription of Vimentin (VIM) by binding with ZNF502, which can help ENAH-202 promote OSCC progression. CONCLUSIONS ENAH-202 facilitated OSCC cell proliferation and metastasis by regulating ZNF502/VIM axis, which played an important role in OSCC progression.
Collapse
Affiliation(s)
- Xinyue Zhang
- School of StomatologyWeifang Medical UniversityWeifangShandongChina
- Weifang Key Laboratory of Oral BiomedicineWeifang Medical UniversityWeifangShandongChina
| | - Xi Chen
- School of StomatologyWeifang Medical UniversityWeifangShandongChina
- Weifang Key Laboratory of Oral BiomedicineWeifang Medical UniversityWeifangShandongChina
| | - Dongyuan Sun
- School of StomatologyWeifang Medical UniversityWeifangShandongChina
- Weifang Key Laboratory of Oral BiomedicineWeifang Medical UniversityWeifangShandongChina
| | - Ning Song
- School of StomatologyWeifang Medical UniversityWeifangShandongChina
- Weifang Key Laboratory of Oral BiomedicineWeifang Medical UniversityWeifangShandongChina
| | - Minmin Li
- School of StomatologyWeifang Medical UniversityWeifangShandongChina
- Weifang Key Laboratory of Oral BiomedicineWeifang Medical UniversityWeifangShandongChina
| | - Wentian Zheng
- School of StomatologyWeifang Medical UniversityWeifangShandongChina
- Weifang Key Laboratory of Oral BiomedicineWeifang Medical UniversityWeifangShandongChina
| | - Yang Yu
- School of StomatologyWeifang Medical UniversityWeifangShandongChina
- Weifang Key Laboratory of Oral BiomedicineWeifang Medical UniversityWeifangShandongChina
| | - Gang Ding
- School of StomatologyWeifang Medical UniversityWeifangShandongChina
- Weifang Key Laboratory of Oral BiomedicineWeifang Medical UniversityWeifangShandongChina
| | - Yingying Jiang
- School of StomatologyWeifang Medical UniversityWeifangShandongChina
- Weifang Key Laboratory of Oral BiomedicineWeifang Medical UniversityWeifangShandongChina
| |
Collapse
|
13
|
Masrour M, Khanmohammadi S, Fallahtafti P, Rezaei N. Long non-coding RNA as a potential diagnostic biomarker in head and neck squamous cell carcinoma: A systematic review and meta-analysis. PLoS One 2023; 18:e0291921. [PMID: 37733767 PMCID: PMC10513217 DOI: 10.1371/journal.pone.0291921] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/09/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is a group of malignancies arising from the epithelium of the head and neck. Despite efforts in treatment, results have remained unsatisfactory, and the death rate is high. Early diagnosis of HNSCC has clinical importance due to its high rates of invasion and metastasis. This systematic review and meta-analysis evaluated the diagnostic accuracy of lncRNAs in HNSCC patients. METHODS PubMed, ISI, SCOPUS, and EMBASE were searched for original publications published till April 2023 using MeSH terms and free keywords "long non-coding RNA" and "head and neck squamous cell carcinoma" and their expansions. The Reitsma bivariate random effect model pooled diagnostic test performance for studies that reported specificity and sensitivity; diagnostic AUC values from all trials were meta-analyzed using the random effects model with the inverse variance method. RESULTS The initial database search yielded 3209 articles, and 25 studies met our criteria. The cumulative sensitivity and specificity for lncRNAs in the diagnosis of HNSCC were 0.74 (95%CI: 0.68-0.7 (and 0.79 (95%CI: 0.74-0.83), respectively. The pooled AUC value for all specimen types was found to be 0.83. Using the inverse variance method, 71 individual lncRNAs yielded a pooled AUC of 0.77 (95%CI: 0.74-0.79). Five studies reported on the diagnostic accuracy of the MALAT1 lncRNA with a pooled AUC value of 0.83 (95%CI: 0.73-0.94). CONCLUSIONS LncRNAs could be used as diagnostic biomarkers for HNSCC, but further investigation is needed to validate clinical efficacy and elucidate mechanisms. High-throughput sequencing and bioinformatics should be used to ascertain expression profiles.
Collapse
Affiliation(s)
- Mahdi Masrour
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shaghayegh Khanmohammadi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | | | - Nima Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Zhou W, Feng Y, Lin C, CHAO CK, He Z, Zhao S, Xue J, Zhao X, Cao W. Yin Yang 1-Induced Long Noncoding RNA DUXAP9 Drives the Progression of Oral Squamous Cell Carcinoma by Blocking CDK1-Mediated EZH2 Degradation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207549. [PMID: 37401236 PMCID: PMC10477890 DOI: 10.1002/advs.202207549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 04/04/2023] [Indexed: 07/05/2023]
Abstract
LncRNAs play a critical role in oral squamous cell carcinoma (OSCC) progression. However, the function and detailed molecular mechanism of most lncRNAs in OSCC are not fully understood. Here, a novel nuclear-localized lncRNA, DUXAP9 (DUXAP9), that is highly expressed in OSCC is identified. A high level of DUXAP9 is positively associated with lymph node metastasis, poor pathological differentiation, advanced clinical stage, worse overall survival, and worse disease-specific survival in OSCC patients. Overexpression of DUXAP9 significantly promotes OSCC cell proliferation, migration, invasion, and xenograft tumor growth and metastasis, and upregulates N-cadherin, Vimentin, Ki67, PCNA, and EZH2 expression and downregulates E-cadherin in vitro and in vivo, whereas knockdown of DUXAP9 remarkably suppresses OSCC cell proliferation, migration, invasion, and xenograft tumor growth in vitro and in vivo in an EZH2-dependent manner. Yin Yang 1 (YY1) is found to activate the transcriptional expression of DUXAP9 in OSCC. Furthermore, DUXAP9 physically interacts with EZH2 and inhibits EZH2 degradation via the suppression of EZH2 phosphorylation, thereby blocking EZH2 translocation from the nucleus to the cytoplasm. Thus, DUXAP9 can serve as a promising target for OSCC therapy.
Collapse
Affiliation(s)
- Wenkai Zhou
- Department of Oral and Maxillofacial & Head and Neck OncologyShanghai Ninth People's HospitalCollege of StomatologyShanghai Jiao Tong University School of MedicineShanghai200011China
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai200011China
| | - Yisheng Feng
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai200011China
| | - Chengzhong Lin
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai200011China
- The 2nd Dental CenterShanghai Ninth People's HospitalCollege of StomatologyShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityShanghai200011China
| | - Chi Kuan CHAO
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai200011China
| | - Ziqi He
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai200011China
| | - Shiyao Zhao
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai200011China
| | - Jieyuan Xue
- Department of CardiologyShanghai Chest HospitalShanghai Jiao Tong UniversityShanghai200030China
| | - Xu‐Yun Zhao
- Department of Biochemistry and Molecular Cell BiologyShanghai Key Laboratory for Tumor Microenvironment and InflammationKey Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Wei Cao
- Department of Oral and Maxillofacial & Head and Neck OncologyShanghai Ninth People's HospitalCollege of StomatologyShanghai Jiao Tong University School of MedicineShanghai200011China
- National Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai200011China
| |
Collapse
|
15
|
Zheng Z, Wang X, Chen D. Proteasome inhibitor MG132 enhances the sensitivity of human OSCC cells to cisplatin via a ROS/DNA damage/p53 axis. Exp Ther Med 2023; 25:224. [PMID: 37123203 PMCID: PMC10133788 DOI: 10.3892/etm.2023.11924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 02/27/2023] [Indexed: 05/02/2023] Open
Abstract
Cis-diamine-dichloroplatinum II (cisplatin, CDDP) is a key chemotherapeutic regimen in the treatment of oral squamous cell carcinoma (OSCC). However, the therapeutic efficacy of cisplatin in OSCC may be hampered by chemoresistance. Therefore, the development of novel combination therapy strategies to overcome the limitations of CDDP is of great importance. The proteasome inhibitor MG132 exhibits anti-cancer properties against various types of cancer. However, our knowledge of its anti-cancer effects in combination with CDDP in OSCC cells remains limited. In the current study, the synergetic effects of MG132 and CDDP were evaluated in the human CAL27 OSCC cell line. CAL27 cells were treated with CDDP alone or in combination with MG132. The results showed that MG132 significantly reduced cell viability in a dose-dependent manner. Additionally, cell viability was significantly reduced in CAL27 cells treated with 0.2 µM MG132 and 2 µM CDDP compared with cells treated with MG132 or CDDP alone. In addition, MG132 significantly enhanced the CDDP-induced generation of intracellular reactive oxygen species and DNA damage in OSCC cells. Furthermore, treatment with CDDP or MG132 alone notably inhibited colony formation and proliferation of OSCC cells. However, co-treatment of OSCC cells with MG132 and CDDP further hampered colony formation and proliferation compared with cells treated with either MG132 or CDDP alone. Finally, in cells co-treated with MG132 and CDDP, the expression of p53 was markedly elevated and the p53-mediated apoptotic pathway was further activated compared with cells treated with MG132 or CDDP alone, as shown by the enhanced cell apoptosis, Bax upregulation, and Bcl-2 downregulation. Overall, the results of the current study support the synergistic anti-cancer effects of a combination of MG132 and CDDP against OSCC, thus suggesting that the combination of MG132 and CDDP may be a promising therapeutic strategy for the management of OSCC.
Collapse
Affiliation(s)
- Zheng Zheng
- Department of Stomatology, The First People's Hospital of Nantong, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226000, P.R. China
| | - Xiang Wang
- Department of Stomatology, The First People's Hospital of Nantong, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226000, P.R. China
- Correspondence to: Dr Donglei Chen or Dr Xiang Wang, Department of Stomatology, The First People's Hospital of Nantong, Affiliated Hospital 2 of Nantong University, 6 Haierxiang Road, Nantong, Jiangsu 226000, P.R. China
| | - Donglei Chen
- Department of Stomatology, The First People's Hospital of Nantong, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226000, P.R. China
- Correspondence to: Dr Donglei Chen or Dr Xiang Wang, Department of Stomatology, The First People's Hospital of Nantong, Affiliated Hospital 2 of Nantong University, 6 Haierxiang Road, Nantong, Jiangsu 226000, P.R. China
| |
Collapse
|
16
|
Zhang Z, Kim BS, Han W, Chen X, Yan Y, Lin L, Chai G. Identifying Oxidized Lipid Metabolism-Related LncRNAs as Prognostic Biomarkers of Head and Neck Squamous Cell Carcinoma. J Pers Med 2023; 13:jpm13030488. [PMID: 36983670 PMCID: PMC10054813 DOI: 10.3390/jpm13030488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
The relationship between oxidized lipid metabolism and the immunological function of cancer is well known. However, the functions and regulatory mechanisms of lncRNAs associated with oxidized lipid metabolism in head and neck squamous cell carcinoma (HNSCC) remain to be fully elucidated. In this study, we established an oxidized lipid metabolism-related lncRNA prognostic signature to assess the prognosis and immune infiltration of HNSCC patients. The HNSCC transcriptome was obtained from The Cancer Genome Atlas. The choice of the target genes with a relevance score greater than 10 was performed via a correlation analysis by GeneCards. Patients were categorized by risk score and generated with multivariate Cox regression, which was then validated and evaluated using the Kaplan–Meier analysis and time-dependent receiver operating characteristics (ROC). A nomogram was constructed by combining the risk score with the clinical data. We constructed a risk score with 24 oxidized lipid metabolism-related lncRNAs. The areas’ 1-, 2-, and 3-year OS under the ROC curve (AUC) were 0.765, 0.724, and 0.724, respectively. Furthermore, the nomogram clearly distinguished the survival probabilities of patients in high- and low-risk groups, between which substantial variations were revealed by immune infiltration analysis. The results supported the fact that oxidized lipid metabolism-related lncRNAs might predict prognoses and assist with differentiating amid differences in immune infiltration in HNSCC.
Collapse
|
17
|
Li S, Shen S, Ge W, Cen Y, Zhang S, Cheng X, Wang X, Xie X, Lu W. Long non-coding RNA SLC25A21-AS1 inhibits the development of epithelial ovarian cancer by specifically inducing PTBP3 degradation. Biomark Res 2023; 11:12. [PMID: 36717926 PMCID: PMC9885650 DOI: 10.1186/s40364-022-00432-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/03/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Epithelial ovarian cancer (EOC) is a highly prevalent disease that rapidly metastasizes and has poor prognosis. Most women are in the middle or late stages when diagnosed and have low survival rates. Recently, long non-coding RNAs (lncRNAs) were recognized to play pivotal roles in the development of EOC. METHODS The expression of SLC25A21 antisense RNA 1 (SLC25A21-AS1) and Polypyrimidine Tract Binding Protein 3 (PTBP3) in EOC cells was assessed via qPCR. The proliferation activity of these cells was detected by EdU and Cell counting kit-8 (CCK8) assays, while the death rate of apoptotic cells and the cell cycle were detected by flow cytometry. Detection of cell transfer rate by transwell assay. Protein expression was measured through western blotting. Interactions between SLC25A21-AS1 and PTBP3 were detected through RNA immunoprecipitation (RIP), IF-FISH co-localization experiments and electrophoretic mobility shift assay (EMSA). The in vivo importance of SLC25A21-AS1 as a tumor suppressor modulator was assessed using murine xenograft models. RESULTS The lncRNA SLC25A21-AS1 has negligible expression in ovarian cancer tissues compared with that in normal ovarian tissues. A series of functional experiments revealed that the upregulation of SLC25A21-AS1 markedly blocked the proliferation and metastasis of EOC cells in vitro, while its downregulation had the opposite effect. Overexpression of SLC25A21-AS1 in a nude mouse model of EOC in vivo resulted in slower tumor growth and weakened metastatic potential. Moreover, SLC25A21-AS1 reduced the protein stability of PTBP3 and promoted its degradation. A series of subsequent experiments found that SLC25A21-AS1 inhibits EOC cell proliferation and metastasis by modulating PTBP3 through the ubiquitin-proteasome pathway and that the combination of SLC25A21-AS1 and PTBP3 provides the necessary conditions for the for the function to be realized. CONCLUSIONS Our research reveals the effect of SLC25A21-AS1 in EOC development and suggests SLC25A21-AS1 can serve as a prognostic target by promoting the degradation of PTBP3 to improve patient survival.
Collapse
Affiliation(s)
- Sihui Li
- Women's Reproductive Health Laboratory of Zhejiang Province; Women's Hospital; School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Shizhen Shen
- Women's Reproductive Health Laboratory of Zhejiang Province; Women's Hospital; School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Wanzhong Ge
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
- Division of Human Reproduction and Developmental Genetics, Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
- Institute of Genetics, Zhejiang University, Hangzhou, 310058, China
| | - Yixuan Cen
- Women's Reproductive Health Laboratory of Zhejiang Province; Women's Hospital; School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Songfa Zhang
- Department of Gynecologic Oncology; Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Xiaodong Cheng
- Women's Reproductive Health Laboratory of Zhejiang Province; Women's Hospital; School of Medicine, Zhejiang University, Hangzhou, 310006, China
- Department of Gynecologic Oncology; Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Xinyu Wang
- Women's Reproductive Health Laboratory of Zhejiang Province; Women's Hospital; School of Medicine, Zhejiang University, Hangzhou, 310006, China
- Department of Gynecologic Oncology; Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Xing Xie
- Women's Reproductive Health Laboratory of Zhejiang Province; Women's Hospital; School of Medicine, Zhejiang University, Hangzhou, 310006, China
- Department of Gynecologic Oncology; Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Weiguo Lu
- Women's Reproductive Health Laboratory of Zhejiang Province; Women's Hospital; School of Medicine, Zhejiang University, Hangzhou, 310006, China.
- Department of Gynecologic Oncology; Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China.
- Cancer Center, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
18
|
Hui Y, Xu Z, Li J, Kuang L, Zhong Y, Tang Y, Wei J, Zhou H, Zheng T. Nonenzymatic function of DPP4 promotes diabetes-associated cognitive dysfunction through IGF-2R/PKA/SP1/ERp29/IP3R2 pathway-mediated impairment of Treg function and M1 microglia polarization. Metabolism 2023; 138:155340. [PMID: 36302455 DOI: 10.1016/j.metabol.2022.155340] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/16/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Impairment of regulatory T (Treg) cells function is implicated in the pathogenesis of immune imbalance-mediated cognitive impairment. A complete understanding of whether and how this imbalance affect cognitive function in type 2 diabetes is lacking, and the driver affecting this imbalance remains unknown. METHODS We examined the impact of enzymatic and non-enzymatic function of DPP4 on Treg cell impairment, microglia polarization and diabetes-associated cognitive defects and identified its underlying mechanism in type 2 diabetic patients with cognitive impairment and in db/db mice. RESULTS We report that DPP4 binds to IGF2-R on Treg cell surface and activates PKA/SP1 signaling, which upregulate ERp29 expression and promote its binding to IP3R2, thereby inhibiting IP3R2 degradation and promoting mitochondria-associated ER membrane formation and mitochondria calcium overload in Tregs. This, in turn, impairs Tregs function and polarizes microglia toward a pro-inflammatory phenotype in the hippocampus and finally leads to neuroinflammation and cognitive impairment in type 2 diabetes. Importantly, inhibiting DPP4 enzymatic activity in type 2 diabetic patients or mutating DPP4 enzymatic active site in db/db mice did not reverse these changes. However, IGF-2R knockdown or blockade ameliorated these effects both in vivo and in vitro. CONCLUSION These findings highlight the nonenzymatic role of DPP4 in impairing Tregs function, which may facilitate the design of novel immunotherapies for diabetes-associated cognitive impairment.
Collapse
Affiliation(s)
- Ya Hui
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541199, PR China; Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi 541199, PR China
| | - Zhiqiang Xu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541199, PR China; Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi 541199, PR China
| | - Jiaxiu Li
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541199, PR China; Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi 541199, PR China; Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, Guangxi 541199, PR China
| | - Liuyu Kuang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541199, PR China
| | - Yuanmei Zhong
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541199, PR China
| | - Yunyun Tang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541199, PR China
| | - Junjie Wei
- Lingui Clinical Medical College, Guilin Medical University, Guilin, Guangxi 541199, PR China
| | - Huimin Zhou
- Department of General Medicine, Guilin Medical University, Guilin, Guangxi 541199, PR China
| | - Tianpeng Zheng
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541199, PR China; Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi 541199, PR China; Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, Guangxi 541199, PR China; Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541199, PR China.
| |
Collapse
|
19
|
Mirzaei S, Paskeh MDA, Entezari M, Mirmazloomi SR, Hassanpoor A, Aboutalebi M, Rezaei S, Hejazi ES, Kakavand A, Heidari H, Salimimoghadam S, Taheriazam A, Hashemi M, Samarghandian S. SOX2 function in cancers: Association with growth, invasion, stemness and therapy response. Biomed Pharmacother 2022; 156:113860. [DOI: 10.1016/j.biopha.2022.113860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/30/2022] [Accepted: 10/08/2022] [Indexed: 11/29/2022] Open
|
20
|
LncRNA HOXA-AS2 Promotes Temozolomide Resistance in Glioblastoma by Regulated miR-302a-3p/IGF1 Axis. Genet Res (Camb) 2022; 2022:3941952. [PMID: 36479381 PMCID: PMC9705095 DOI: 10.1155/2022/3941952] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 11/23/2022] Open
Abstract
Background Glioblastoma (GBM) is a highly prevalent brain tumor characterized by high rates of morbidity, recurrence, and mortality. While temozolomide (TMZ) is commonly used as a first-line treatment for this cancer, the emergence of TMZ resistance limits its utility. The long noncoding RNA HOXA-AS2 reportedly drives GBM progression, but whether it can influence therapeutic resistance to TMZ has yet to be established. Methods HOXA-AS2 expression was analyzed in TMZ-resistant and sensitive GBM tissue samples and cell lines by qPCR. A siRNA-based approach was used to knock down HOXA-AS2 in GBM cells, after which TMZ resistance was tested. Bioinformatics approaches were used to predict miRNA binding targets of HOXA-AS2, after which a series of luciferase reporter assay and rescue experiments with appropriate miRNA inhibitor/mimic constructs were performed to validate these predictions and to clarify the ability of HOXA-AS2 to regulate chemoresistant activity. Results TMZ-resistant GBM patients and cell lines exhibited increased HOXA-AS2 expression that was correlated with worse overall survival. Knocking down HOXA-AS2 increased the sensitivity of resistant GBM cells to TMZ. miR-302a-3p was identified as a HOXA-AS2 target confirmed through luciferase reporter assays and rescue experiments, and IGF1 was further identified as a confirmed miR-302a-3p target. In addition, HOXA-AS2 knockdown resulted in a corresponding drop in IGF1 expression consistent with indirect regulation mediated by miR-302a-3p. Conclusion In summary, these results highlight the role of HOXA-AS2 as a driver of TMZ resistance in GBM through its ability to regulate the miR-302a-3p/IGF1 signaling axis, highlighting this pathway as a promising target for the diagnosis, therapeutic sensitization, and/or treatment of affected patients.
Collapse
|
21
|
Zhao Y, Zhao J, Zhong M, Zhang Q, Yan F, Feng Y, Guo Y. The expression and methylation of PITX genes is associated with the prognosis of head and neck squamous cell carcinoma. Front Genet 2022; 13:982241. [PMID: 36204311 PMCID: PMC9530742 DOI: 10.3389/fgene.2022.982241] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/07/2022] [Indexed: 12/24/2022] Open
Abstract
Background: The PITX gene family, comprising PITX1, PITX2, and PITX3, is critical in organogenesis and has been evolutionary conserved in animals. PITX genes are associated with the advanced progression and poor prognosis of multiple cancers. However, the relationship between the PITX genes and head and neck squamous cell carcinoma (HNSC) has not been reported. Methods: We used data from The Cancer Genome Atlas (TCGA) to analyze the association between PITX mRNA expression and clinicopathological parameters of patients with HNSC. The prognostic value of PITX genes was evaluated using the Kaplan-Meier plotter. Multivariate Cox analysis was used to screen out prognosis-associated genes to identify better prognostic indicators. The potential roles of PITX1 and PITX2 in HNSC prognosis were investigated using the protein-protein interaction (PPI) network, Gene Ontology (GO) analysis, and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. The correlation between PITX1 and PITX2 expression or methylation and immune cell infiltration was evaluated using the tumor-immune system interaction database (TISIDB). MethSurv was used to identify DNA methylation and its effect on HNSC prognosis. Results:PITX genes expression was correlated with different cancers. PITX1 and PITX2 expression was lower in the patients with HNSC. In HNSC, PITX1 expression was significantly related to the clinical stage, histologic grade, and N stage, while PITX2 expression was only significantly related to the histologic grade. The high expression of PITX3 was significantly related to the histologic grade, T stage, and N stage. Survival analysis revealed that PITX genes had prognostic value in HNSC, which was supported by multivariate Cox analysis. PPI network and enrichment analysis showed that the genes interacting with PITX1 and PITX2 belonged predominantly to signaling pathways associated with DNA binding and transcription. Of the CpG DNA methylation sites in PITX1 and PITX2, 28 and 22 were related to the prognosis of HNSC, respectively. Additionally, PITX1 and PITX2 expression and methylation was associated with tumor-infiltrating lymphocytes (TILs). Conclusion: The PITX genes were differentially expressed in patients with HNSC, highlighting their essential role in DNA methylation and tumor-infiltrating immune cell regulation, as well as overall prognostic value in HNSC.
Collapse
Affiliation(s)
- Yaqiong Zhao
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jie Zhao
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Mengmei Zhong
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qian Zhang
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Fei Yan
- Hunan Key Laboratory of Oral Health Research and Hunan 3D Printing Engineering Research Center of Oral Care and Hunan Clinical Research Center of Oral Major Diseases and Oral Health and Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, Changsha, China
| | - Yunzhi Feng
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yue Guo
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Yue Guo,
| |
Collapse
|
22
|
Li S, Xie X, Peng F, Du J, Peng C. Regulation of temozolomide resistance via lncRNAs: Clinical and biological properties of lncRNAs in gliomas (Review). Int J Oncol 2022; 61:101. [PMID: 35796022 PMCID: PMC9291250 DOI: 10.3892/ijo.2022.5391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/10/2022] [Indexed: 11/05/2022] Open
Abstract
Gliomas are a primary types of intracranial malignancies and are characterized by a poor prognosis due to aggressive recurrence profiles. Temozolomide (TMZ) is an auxiliary alkylating agent that is extensively used in conjunction with surgical resection and forms the mainstay of clinical treatment strategies for gliomas. However, the frequent occurrence of TMZ resistance in clinical practice limits its therapeutic efficacy. Accumulating evidence has demonstrated that long non‑coding RNAs (lncRNAs) can play key and varied roles in glioma progression. lncRNAs have been reported to inhibit glioma progression by targeting various signaling pathways. In addition, the differential expression of lncRNAs has also been found to mediate the resistance of glioma to several chemotherapeutic agents, particularly to TMZ. The present review article therefore summarizes the findings of previous studies in an aim to report the significance and function of lncRNAs in regulating the chemoresistance of gliomas. The present review may provide further insight into the clinical treatment of gliomas.
Collapse
Affiliation(s)
- Sui Li
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of The Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiaofang Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, P.R. China
| | - Fu Peng
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of The Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Correspondence to: Dr Fu Peng or Professor Junrong Du, Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of The Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, 17 Renmin South Road, Chengdu, Sichuan 610041, P.R. China, E-mail: , E-mail:
| | - Junrong Du
- Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of The Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, P.R. China
- Correspondence to: Dr Fu Peng or Professor Junrong Du, Department of Pharmacology, Key Laboratory of Drug-Targeting and Drug Delivery System of The Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, 17 Renmin South Road, Chengdu, Sichuan 610041, P.R. China, E-mail: , E-mail:
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, P.R. China
| |
Collapse
|
23
|
Eptaminitaki GC, Stellas D, Bonavida B, Baritaki S. Long Non-coding RNAs (lncRNAs) signaling in Cancer Chemoresistance: From Prediction to Druggability. Drug Resist Updat 2022; 65:100866. [DOI: 10.1016/j.drup.2022.100866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 11/03/2022]
|
24
|
Meng Z, Yang W, Zhu L, Liu W, Wang Y. A novel necroptosis-related LncRNA signature for prediction of prognosis and therapeutic responses of head and neck squamous cell carcinoma. Front Pharmacol 2022; 13:963072. [PMID: 36016575 PMCID: PMC9395581 DOI: 10.3389/fphar.2022.963072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Long non-coding RNAs (lncRNAs) play an essential role in the occurrence and prognosis of tumors, and it has great potential as biomarkers of tumors. However, the roles of Necroptosis-related lncRNA (NRLs) in Head and neck squamous cell carcinoma (HNSCC) remain elusive. Methods: We comprehensively analyzed the gene expression and clinical information of 964 HNSCC in four cohorts. LASSO regression was utilized to construct a necroptosis-related lncRNA prognosis signature (NLPS). We used univariate and multivariate regression to assess the independent prognostic value of NLPS. Based on the optimal cut-off, patients were divided into high- and low-risk groups. In addition, the immune profile, multi-omics alteration, and pharmacological landscape of NLPS were further revealed. Results: A total of 21 NRLs associated with survival were identified by univariate regression in four cohorts. We constructed and validated a best prognostic model (NLPS). Compared to the low-risk group, patients in the high group demonstrated a more dismal prognosis. After adjusting for clinical features by multivariate analysis, NLPS still displayed independent prognostic value. Additionally, further analysis found that patients in the low-risk group showed more abundant immune cell infiltration and immunotherapy response. In contrast, patients in the high-risk group were more sensitive to multiple chemotherapeutic agents. Conclusion: As a promising tool, the establishment of NLPS provides guidance and assistance in the clinical management and personalized treatment of HNSCC.
Collapse
Affiliation(s)
| | | | | | | | - Yudong Wang
- Department of Maxillofacial Surgery, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
25
|
The Long Noncoding Transcript HNSCAT1 Activates KRT80 and Triggers Therapeutic Efficacy in Head and Neck Squamous Cell Carcinoma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4156966. [PMID: 35965679 PMCID: PMC9371835 DOI: 10.1155/2022/4156966] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/04/2022] [Indexed: 12/23/2022]
Abstract
Head and neck squamous carcinoma (HNSC) is the most prevalent malignancy of the head and neck regions. Long noncoding RNAs (lncRNAs) are vital in tumorigenesis regulation. However, the role of lncRNAs in HNSC requires further exploration. Herein, through bioinformatic assays using The Cancer Genome Atlas (TCGA) datasets, rapid amplification of cDNA ends (RACE) assays, and RNA-FISH, we revealed that a novel cytoplasmic transcript, HNSC-associated transcript 1 (HNSCAT1, previously recognized as linc01269), was downregulated in tumor samples and advanced tumor stages and was also associated with favorable outcomes in HNSC. Overexpression of HNSCAT1 triggered treatment efficacy in HNSCs both in vivo and in vitro. More importantly, through high-throughput transcriptome analysis (RNA-seq, in NODE database, OEZ007550), we identified KRT80, a tumor suppressor in HNSC, as the target of HNSCAT1. KRT80 expression was modulated by lncRNA HNSCAT1 and presented a positive correlation in tumor samples (R = 0.52, p < 0.001). Intriguingly, we identified that miR-1245 simultaneously interacts with KRT80 and HNSCAT1, which bridges the regulatory function between KRT80 and HNSCAT1. Conclusively, our study demonstrated that lncRNA HNSCAT1 functions as a necessary tumor inhibitor in HNSC, which provides a novel mechanism of lncRNA function and provides alternative targets for the diagnosis and treatment of HNSC.
Collapse
|
26
|
Peña-Flores JA, Bermúdez M, Ramos-Payán R, Villegas-Mercado CE, Soto-Barreras U, Muela-Campos D, Álvarez-Ramírez A, Pérez-Aguirre B, Larrinua-Pacheco AD, López-Camarillo C, López-Gutiérrez JA, Garnica-Palazuelos J, Estrada-Macías ME, Cota-Quintero JL, Barraza-Gómez AA. Emerging role of lncRNAs in drug resistance mechanisms in head and neck squamous cell carcinoma. Front Oncol 2022; 12:965628. [PMID: 35978835 PMCID: PMC9376329 DOI: 10.3389/fonc.2022.965628] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/01/2022] [Indexed: 12/12/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) originates in the squamous cell lining the mucosal surfaces of the head and neck region, including the oral cavity, nasopharynx, tonsils, oropharynx, larynx, and hypopharynx. The heterogeneity, anatomical, and functional characteristics of the patient make the HNSCC a complex and difficult-to-treat disease, leading to a poor survival rate and a decreased quality of life due to the loss of important physiologic functions and aggressive surgical injury. Alteration of driver-oncogenic and tumor-suppressing lncRNAs has recently been recently in HNSCC to obtain possible biomarkers for diagnostic, prognostic, and therapeutic approaches. This review provides current knowledge about the implication of lncRNAs in drug resistance mechanisms in HNSCC. Chemotherapy resistance is a major therapeutic challenge in HNSCC in which lncRNAs are implicated. Lately, it has been shown that lncRNAs involved in autophagy induced by chemotherapy and epithelial–mesenchymal transition (EMT) can act as mechanisms of resistance to anticancer drugs. Conversely, lncRNAs involved in mesenchymal–epithelial transition (MET) are related to chemosensitivity and inhibition of invasiveness of drug-resistant cells. In this regard, long non-coding RNAs (lncRNAs) play a pivotal role in both processes and are important for cancer detection, progression, diagnosis, therapy response, and prognostic values. As the involvement of more lncRNAs is elucidated in chemoresistance mechanisms, an improvement in diagnostic and prognostic tools could promote an advance in targeted and specific therapies in precision oncology.
Collapse
Affiliation(s)
- José A. Peña-Flores
- Faculty of Odontology, Autonomous University of Chihuahua, Chihuahua, Mexico
| | - Mercedes Bermúdez
- Faculty of Odontology, Autonomous University of Chihuahua, Chihuahua, Mexico
- *Correspondence: Mercedes Bermúdez,
| | - Rosalío Ramos-Payán
- Faculty of Biological and Chemical Sciences, Autonomous University of Sinaloa, Culiacán, Mexico
| | | | - Uriel Soto-Barreras
- Faculty of Odontology, Autonomous University of Chihuahua, Chihuahua, Mexico
| | | | | | | | | | | | - Jorge A. López-Gutiérrez
- Faculty of Biological and Chemical Sciences, Autonomous University of Sinaloa, Culiacán, Mexico
- Faculty of Biology, Autonomous University of Sinaloa, Culiacán, Mexico
| | | | | | - Juan L. Cota-Quintero
- Faculty of Biology, Autonomous University of Sinaloa, Culiacán, Mexico
- Faculty of Odontology , Autonomous University of Sinaloa, Culiacán, Mexico
| | | |
Collapse
|
27
|
Chen X, Liu Y, Sun D, Sun R, Wang X, Li M, Song N, Ying J, Guo T, Jiang Y. Long noncoding RNA lnc-H2AFV-1 promotes cell growth by regulating aberrant m6A RNA modification in HNSCC. Cancer Sci 2022; 113:2071-2084. [PMID: 35403343 PMCID: PMC9207353 DOI: 10.1111/cas.15366] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/28/2022] [Accepted: 04/03/2022] [Indexed: 11/29/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the most common malignant tumor in the oral and maxillofacial regions, and long noncoding RNAs (lncRNAs) play crucial roles in the occurrence and progression of HNSCC. The lncRNA lnc-H2AFV-1 was found to be upregulated in HNSCC tissues; however, the function of lnc-H2AFV-1 in regulating HNSCC proliferation and the potential molecular mechanism is unclear. The present study evaluated the expression of lnc-H2AFV-1 in HNSCC tissues using quantitative real-time PCR (qPCR) and associated abundant lnc-H2AFV-1 expression with tumor size. Functionally, lnc-H2AFV-1 significantly promoted the proliferation of HNSCC cells in vitro and in vivo. Quantified N6-methyladenosine (m6A) RNA methylation and dot blot assays revealed that total m6A methylation in HNSCC cells was accompanied by lnc-H2AFV-1 expression. Western blotting showed that the expression of methyltransferase-like (METTL) 3 and METTL14 was consistent with that of lnc-H2AFV-1, whereas the expression of demethylase fat mass and obesity-associated (FTO) was contrary to that of lnc-H2AFV-1. Methylated RNA immunoprecipitation sequencing (MeRIP-seq) and MeRIP-qPCR revealed that lnc-H2AFV-1 overexpression led to the elevated expression and maximal m6A methylation of intraflagellar transport (IFT) 80 in HNSCC. In addition, METTL3/14 knockdown decreased IFT80 expression. Thus, our findings suggested that lnc-H2AFV-1 might be a biomarker that alters m6A modification by regulating the m6A methylases METTL3/14 and FTO, and then mediating downstream target IFT80 to promote HNSCC progression.
Collapse
Affiliation(s)
- Xi Chen
- School of Stomatology, Weifang Medical University, Weifang, Shandong, China
| | - Yunxia Liu
- School of Stomatology, Weifang Medical University, Weifang, Shandong, China.,Department of Dentistry, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Dongyuan Sun
- School of Stomatology, Weifang Medical University, Weifang, Shandong, China
| | - Rongqi Sun
- School of Stomatology, Weifang Medical University, Weifang, Shandong, China
| | - Xiaoxiao Wang
- School of Stomatology, Weifang Medical University, Weifang, Shandong, China.,Department of Dentistry, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Minmin Li
- School of Stomatology, Weifang Medical University, Weifang, Shandong, China
| | - Ning Song
- School of Stomatology, Weifang Medical University, Weifang, Shandong, China
| | - Jicheng Ying
- School of Stomatology, Weifang Medical University, Weifang, Shandong, China
| | - Tao Guo
- Department of Pathophysiology, School of Basic Medical Sciences, Weifang Medical University, Weifang, Shandong, China
| | - Yingying Jiang
- School of Stomatology, Weifang Medical University, Weifang, Shandong, China.,Department of Dentistry, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China.,Department of Oral and Maxillofacial-Head & Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
28
|
Impact of Non-Coding RNAs on Chemotherapeutic Resistance in Oral Cancer. Biomolecules 2022; 12:biom12020284. [PMID: 35204785 PMCID: PMC8961659 DOI: 10.3390/biom12020284] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 02/04/2023] Open
Abstract
Drug resistance in oral cancer is one of the major problems in oral cancer therapy because therapeutic failure directly results in tumor recurrence and eventually in metastasis. Accumulating evidence has demonstrated the involvement of non-coding RNAs (ncRNAs), such as microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), in processes related to the development of drug resistance. A number of studies have shown that ncRNAs modulate gene expression at the transcriptional or translational level and regulate biological processes, such as epithelial-to-mesenchymal transition, apoptosis, DNA repair and drug efflux, which are tightly associated with drug resistance acquisition in many types of cancer. Interestingly, these ncRNAs are commonly detected in extracellular vesicles (EVs) and are known to be delivered into surrounding cells. This intercellular communication via EVs is currently considered to be important for acquired drug resistance. Here, we review the recent advances in the study of drug resistance in oral cancer by mainly focusing on the function of ncRNAs, since an increasing number of studies have suggested that ncRNAs could be therapeutic targets as well as biomarkers for cancer diagnosis.
Collapse
|