1
|
Li C, Zhang ED, Yu R, Yuan B, Yang Y, Zeng Z, Huang H. Comprehensive multi-omics analysis showed that CDC6 is a potential prognostic and immunotherapy biomarker for multiple cancer types including HCC. Transl Oncol 2025; 53:102314. [PMID: 39904279 PMCID: PMC11846587 DOI: 10.1016/j.tranon.2025.102314] [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: 09/12/2024] [Revised: 01/07/2025] [Accepted: 01/30/2025] [Indexed: 02/06/2025] Open
Abstract
BACKGROUND Cell division cycle 6 (CDC6) is a member of the AAA+ ATPase family and has chaperone-like activity. Many studies have shown that CDC6 plays an important role in cancer development and progression. METHODS Explored CDC6 mRNA and protein expression in normal human tissues and tumors using TCGA, GTEx, and HPA. The role of CDC6 in cancer was analyzed using multiple web platforms and software, including R, cBioPortal, UALCAN, SangerBox and others. Finally, CCK-8, EdU assays and Transwell assays were used to verify the effects of CDC6 knockdown on HCC cell proliferation, migration, and invasion. RESULTS CDC6 expression was upregulated in most cancers and was associated with poorer prognosis. RNA methylation may play an important role in CDC6 epigenetic modification. CDC6 was significantly positively associated with CD4+ Th2 cells and MDSC in a variety of tumors. Furthermore, immunomodulatory genes are strongly associated with CDC6 expression in most tumor types. CDC6 has higher predictive value than B. Clonality and TMB, and its expression is significantly positively correlated with TMB/MSI and DNAss/RNAss, and is closely related to cell cycle events. Down-regulation of CDC6 can inhibit proliferation, migration and invasion of HCC cells. CONCLUSIONS CDC6 is associated with the occurrence and progression of multiple cancer types by regulating the cell cycle. It holds promise as a diagnostic and prognostic biomarker for cancer, and offers potential in immunomodulatory and targeted therapies.
Collapse
Affiliation(s)
- Chenxuan Li
- Organ Transplantation Center, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming 650032, Yunnnan, China
| | - En-di Zhang
- Organ Transplantation Center, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming 650032, Yunnnan, China
| | - Rui Yu
- Organ Transplantation Center, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming 650032, Yunnnan, China
| | - Bo Yuan
- Organ Transplantation Center, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming 650032, Yunnnan, China
| | - Yunxin Yang
- Organ Transplantation Center, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming 650032, Yunnnan, China
| | - Zhong Zeng
- Organ Transplantation Center, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming 650032, Yunnnan, China.
| | - Hanfei Huang
- Organ Transplantation Center, The First Affiliated Hospital of Kunming Medical University, 295 Xichang Road, Kunming 650032, Yunnnan, China.
| |
Collapse
|
2
|
Zhao H, Sun H, Fang J, Yuan G, Sun S, Gu Y, Zhou X. CDC6 overexpression contributes to the malignant phenotype of glioma via IL6/JAK2/STAT3 signaling. Am J Cancer Res 2024; 14:3372-3387. [PMID: 39113855 PMCID: PMC11301287 DOI: 10.62347/dota1781] [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: 02/18/2024] [Accepted: 07/05/2024] [Indexed: 08/10/2024] Open
Abstract
Glioma, a prevalent primary tumor of the central nervous system, is targeted by molecular therapies aiming to intervene in specific genes and signaling pathways to inhibit tumor growth and spread. Our previous bioinformatics study revealed that significant CDC6 overexpression in gliomas was closely correlated with poor patient prognosis. Through qPCR, western blotting, and immunohistochemistry, we will further validate CDC6 expression in clinical glioma specimens, while the effects of silencing and overexpressing CDC6 in the U87 and LN229 glioma cell lines on malignancy will be assessed through MTS, EdU, transwell, and migration assays. Luciferase reporter assays, ChIP, qPCR, and western blotting were used to explore the upstream and downstream molecular mechanisms of CDC6. Our study confirmed the abnormal overexpression of CDC6 in gliomas, particularly in glioblastomas. CDC6 promotes glioma cell activity, proliferation, invasion, and migration by activating the IL6-mediated JAK2/STAT3 signaling pathway. The transcription Factor E2F8 directly regulates CDC6 transcription, playing a crucial role in its abnormal overexpression in gliomas. This research provides vital evidence supporting CDC6 as a molecular target for glioma therapy.
Collapse
Affiliation(s)
- Hao Zhao
- Department of Neurosurgery, Zibo Central HospitalZibo 255036, Shandong, China
| | - Hu Sun
- Department of Neurosurgery, Zibo Central HospitalZibo 255036, Shandong, China
| | - Jing Fang
- Department of Paediatric Neurology, Zibo Central HospitalZibo 255036, Shandong, China
| | - Guang Yuan
- Department of Neurosurgery, Zibo Central HospitalZibo 255036, Shandong, China
| | - Shuo Sun
- Department of Neurosurgery, Zibo Central HospitalZibo 255036, Shandong, China
| | - Yinghao Gu
- Department of Neurosurgery, Zibo Central HospitalZibo 255036, Shandong, China
| | - Xiaojun Zhou
- Department of Paediatric Neurology, Zibo Central HospitalZibo 255036, Shandong, China
| |
Collapse
|
3
|
Qian M, Liang X, Zeng Q, Zhang C, He N, Ma J. SMU1 Knockdown Suppresses Gastric Carcinoma Growth, Migration, and Invasion and Modulates the Cell Cycle. Cancer Control 2024; 31:10732748241281716. [PMID: 39236066 PMCID: PMC11378178 DOI: 10.1177/10732748241281716] [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] [Indexed: 09/07/2024] Open
Abstract
INTRODUCTION The role of SMU1 in DNA replication and RNA splicing is well-established, yet its specific function and dysregulated mechanisms in gastric cancer (GC) remain inadequately elucidated. This study seeks to investigate the potential oncogenic and progression-promoting effects of SMU1 in GC, with the ultimate goal of informing novel approaches for treatment and diagnosis. METHODS The study investigated the expression levels of SMU1 in GC and adjacent normal tissues by analyzing data from the TCGA (27 tissue pairs) and GEO (47 tissue pairs) databases. Immunohistochemistry was used to examine 277 tumor tissue and adjacent non-tumor tissue spots from GC tissue chips, along with relevant follow-up information. The study further assessed the proliferation, invasion, and migration capabilities of cells by manipulating SMU1 expression levels and conducting various assays, including CCK-8, EdU incorporation, colony formation, transwells, flow cytometry, and subcutaneous tumorigenesis assays. RESULTS Our study revealed a significant upregulation of SMU1 mRNA and protein levels in GC tissues compared to adjacent tissues. Univariate and multivariate Cox analysis demonstrated that elevated levels of SMU1 were independent prognostic factors for GC prognosis (P = 0.036). Additionally, median survival analysis indicated a significant association between high SMU1 expression and poor prognosis in GC patients (P = 0.0002). In experiments conducted both in vivo and in vitro, it was determined that elevated levels of SMU1 can enhance the proliferation, invasion, and migration of GC cells, whereas suppression of SMU1 can impede the progression of GC by modulating the G1/S checkpoint of the cell cycle. CONCLUSIONS Our research introduces the novel idea that SMU1 could serve as a prognostic marker for GC progression, influencing cell proliferation through cell cycle activation. These results offer valuable insights into the understanding, diagnosis, and management of gastric carcinoma.
Collapse
Affiliation(s)
- Meirui Qian
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| | - Xue Liang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| | - Qingmei Zeng
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
| | - Chen Zhang
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Nan He
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Jing Ma
- Department of Gastroenterology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, China
| |
Collapse
|
4
|
Yin LK, Yuan HY, Liu JJ, Xu XL, Wang W, Bai XY, Wang P. Identification of survival-associated biomarkers based on three datasets by bioinformatics analysis in gastric cancer. World J Clin Cases 2023; 11:4763-4787. [PMID: 37584004 PMCID: PMC10424043 DOI: 10.12998/wjcc.v11.i20.4763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/11/2023] [Accepted: 06/06/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is one of the most common malignant tumors with poor prognosis in terms of advanced stage. However, the survival-associated biomarkers for GC remains unclear. AIM To investigate the potential biomarkers of the prognosis of patients with GC, so as to provide new methods and strategies for the treatment of GC. METHODS RNA sequencing data from The Cancer Genome Atlas (TCGA) database of STAD tumors, and microarray data from Gene Expression Omnibus (GEO) database (GSE19826, GSE79973 and GSE29998) were obtained. The differentially expressed genes (DEGs) between GC patients and health people were picked out using R software (x64 4.1.3). The intersections were underwent between the above obtained co-expression of differential genes (co-DEGs) and the DEGs of GC from Gene Expression Profiling Interactive Analysis database, and Gene Ontology (GO) analysis, Kyoto Encyclopedia of Gene and Genome (KEGG) pathway analysis, Gene Set Enrichment Analysis (GSEA), Protein-protein Interaction (PPI) analysis and Kaplan-Meier Plotter survival analysis were performed on these DEGs. Using Immunohistochemistry (IHC) database of Human Protein Atlas (HPA), we verified the candidate Hub genes. RESULTS With DEGs analysis, there were 334 co-DEGs, including 133 up-regulated genes and 201 down-regulated genes. GO enrichment analysis showed that the co-DEGs were involved in biological process, cell composition and molecular function pathways. KEGG enrichment analysis suggested the co-DEGs pathways were mainly enriched in ECM-receptor interaction, protein digestion and absorption pathways, etc. GSEA pathway analysis showed that co-DEGs mainly concentrated in cell cycle progression, mitotic cell cycle and cell cycle pathways, etc. PPI analysis showed 84 nodes and 654 edges for the co-DEGs. The survival analysis illustrated 11 Hub genes with notable significance for prognosis of patients were screened. Furtherly, using IHC database of HPA, we confirmed the above candidate Hub genes, and 10 Hub genes that associated with prognosis of GC were identified, namely BGN, CEP55, COL1A2, COL4A1, FZD2, MAOA, PDGFRB, SPARC, TIMP1 and VCAN. CONCLUSION The 10 Hub genes may be the potential biomarkers for predicting the prognosis of GC, which can provide new strategies and methods for the diagnosis and treatment of GC.
Collapse
Affiliation(s)
- Long-Kuan Yin
- Department of Gastrointestinal Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
- Sichuan Key Laboratory of Medical Imaging, North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Hua-Yan Yuan
- Department of Gastrointestinal Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Jian-Jun Liu
- Department of Gastrointestinal Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Xiu-Lian Xu
- Department of Gastrointestinal Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Wei Wang
- Department of Gastrointestinal Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Xiang-Yu Bai
- Department of Gastrointestinal Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
- Sichuan Key Laboratory of Medical Imaging, North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Pan Wang
- Department of Gastrointestinal Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
- Sichuan Key Laboratory of Medical Imaging, North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| |
Collapse
|
5
|
Xu T, Xie M, Jing X, Jiang H, Wu X, Wang X, Shu Y. Loss of miR-26b-5p promotes gastric cancer progression via miR-26b-5p-PDE4B/CDK8-STAT3 feedback loop. J Transl Med 2023; 21:77. [PMID: 36737782 PMCID: PMC9898947 DOI: 10.1186/s12967-023-03933-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 01/26/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Chronic inflammation is a well-known risk factor for the development of gastric cancer (GC). Nevertheless, the molecular mechanisms underlying inflammation-related GC progression are incompletely defined. METHODS Bioinformatic analysis was performed based on data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), and the expression of miR-26b-5p in GC cells and tissues was validated by quantitative real-time PCR (qRT-PCR). Cell proliferation was examined through Cell Counting Kit-8 (CCK8), 5-Ethynyl-2'-deoxyuridine (EdU), colony formation, flow cytometry, and tumor xenografts. Correlation between miR-26b-5p and Cyclin dependent kinase 8 (CDK8) or Phosphodiesterase 4B (PDE4B) was analyzed by dual-luciferase reporter assays, qRT-PCR, and Western blot. The effect of miR-26b-5p on the Signal transducer and activator of transcription 3 (STAT3) pathway was investigated using Western blot, immunofluorescence (IF), and immunohistochemistry (IHC). The impact of STAT3 on miR-26b-5p was determined by dual-luciferase reporter assays and qRT-PCR. RESULTS The expression of miR-26b-5p was significantly downregulated in Helicobacter Pylori (H. pylori)-infected GC cells. The decreased expression of miR-26b-5p was also detected in GC cells and tissues compared to normal gastric epithelium cells (GES1) and normal adjacent gastric tissues. The low expression of miR-26b-5p promoted GC proliferation in vitro and in vivo and was related to the poor outcome of GC patients. In terms of mechanism, miR-26b-5p directly targeted PDE4B and CDK8, resulting in decreased phosphorylation and nuclear translocation of STAT3, which was associated with the regulation of GC proliferation by miR-26b-5p. Notably, miR-26b-5p was transcriptionally suppressed by STAT3, thus forming the miR-26b-5p-PDE4B/CDK8-STAT3 positive feedback loop. CONCLUSION The newly identified miR-26b-5p-PDE4B/CDK8-STAT3 feedback loop plays an important role in inflammation-related GC progression and may serve as a promising therapeutic target for GC.
Collapse
Affiliation(s)
- Tingting Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Oncology, Gusu School, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Mengyan Xie
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinming Jing
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Huning Jiang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xi Wu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinzhu Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
- Department of Oncology, Gusu School, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China.
- Department of Oncology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
6
|
Yuehui Z, Hosoki M, Oshima M, Tajima T, Miyagi M, Raman S, Raju R, Matsuka Y. Identification of microRNA Signatures in Peripheral Blood of Young Women as Potential Biomarkers for Metal Allergy. Biomedicines 2023; 11:biomedicines11020277. [PMID: 36830814 PMCID: PMC9953729 DOI: 10.3390/biomedicines11020277] [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: 12/15/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/20/2023] Open
Abstract
MicroRNA (miRNA) is a short (19-24 nucleotide) endogenous non-protein RNA that exists in the body and controls the translation process from genes to proteins. It has become useful as a diagnostic tool and a potential treatment target in cancer research. To explore the function of miRNA in contact dermatitis, female participants with a positive metal allergy diagnosis (n = 3) were enrolled along with additional female participants with no medical history of metal allergy (n = 3). A patch test was performed on each participant. Peripheral blood was collected from all the participants before the patch test and at days 3 and 7 after starting the patch test. After total RNA was obtained from peripheral blood leukocytes and cDNA was generated, microarray analysis was performed to analyze the large-scale circulating miRNA profile. Real-time polymerase chain reaction (RT-PCR) was then used to clarify the overall target miRNA expression. Downregulation of hsa-let-7d-5p, hsa-miR-24-3p, hsa-miR-23b-3p, hsa-miR-26b-5p, and hsa-miR-150-5p was found on day 7. Certain miRNAs were confirmed using RT-PCR. These peripheral blood miRNAs could be diagnostic biomarkers for metal allergies.
Collapse
Affiliation(s)
- Zhang Yuehui
- Department of Stomatognathic Function and Occlusal Reconstruction, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 7708504, Japan
| | - Maki Hosoki
- Department of Stomatognathic Function and Occlusal Reconstruction, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 7708504, Japan
| | - Masamitsu Oshima
- Department of Stomatognathic Function and Occlusal Reconstruction, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 7708504, Japan
| | - Toyoko Tajima
- Department of Stomatognathic Function and Occlusal Reconstruction, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 7708504, Japan
| | - Mayu Miyagi
- Department of Stomatognathic Function and Occlusal Reconstruction, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 7708504, Japan
| | - Swarnalakshmi Raman
- Department of Stomatognathic Function and Occlusal Reconstruction, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 7708504, Japan
| | - Resmi Raju
- Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Yoshizo Matsuka
- Department of Stomatognathic Function and Occlusal Reconstruction, Graduate School of Biomedical Sciences, Tokushima University, Tokushima 7708504, Japan
- Correspondence:
| |
Collapse
|
7
|
Wang F, Zhao F, Zhang L, Xiong L, Mao Q, Liu Y, Qiu X, Wang X, Shui L, Chen X, Ren K, Shui P, Zhang Q, Deng Y, Li W, Xie X, Wu D, Li T, Lang J, Liu L, Chen H, Xu J, Bai S, Li Z, Yue Q, Chen N, Zhou B, Yi C, Wei Y, Fu Y, Luo Y, Gou Q, Liu L, Liu Y, Kang J, Wang J, Jing D, Zhang F, Yang X, Li X, Jiang T, Zhang Z, Zhou Y, Yi J. CDC6 is a prognostic biomarker and correlated with immune infiltrates in glioma. Mol Cancer 2022; 21:153. [PMID: 35879762 PMCID: PMC9316328 DOI: 10.1186/s12943-022-01623-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 07/12/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Cell division cycle 6 (CDC6) has been proven to be associated with the initiation and progression of human multiple tumors. However, it's role in glioma, which is ranked as one of the common primary malignant tumor in the central nervous system and is associated with high morbidity and mortality, is unclear. METHODS In this study, we explored CDC6 gene expression level in pan-cancer. Furthermore, we focused on the relationships between CDC6 expression, its prognostic value, potential biological functions, and immune infiltrates in glioma patients. We also performed vitro experiments to assess the effect of CDC6 expression on proliferative, apoptotic, migrant and invasive abilities of glioma cells. RESULTS As a result, CDC6 expression was upregulated in multiple types of cancer, including glioma. Moreover, high expression of CDC6 was significantly associated with age, IDH status, 1p/19q codeletion status, WHO grade and histological type in glioma (all p < 0.05). Meanwhile, high CDC6 expression was associated with poor overall survival (OS) in glioma patients, especially in different clinical subgroups. Furthermore, a univariate Cox analysis showed that high CDC6 expression was correlated with poor OS in glioma patients. Functional enrichment analysis indicated that CDC6 was mainly involved in pathways related to DNA transcription and cytokine activity, and Gene Set Enrichment Analysis (GSEA) revealed that MAPK pathway, P53 pathway and NF-κB pathway in cancer were differentially enriched in glioma patients with high CDC6 expression. Single-sample gene set enrichment analysis (ssGSEA) showed CDC6 expression in glioma was positively correlated with Th2 cells, Macrophages and Eosinophils, and negative correlations with plasmacytoid dendritic cells, CD8 T cells and NK CD56bright cells, suggesting its role in regulating tumor immunity. Finally, CCK8 assay, flow cytometry and transwell assays showed that silencing CDC6 could significantly inhibit proliferation, migration, invasion, and promoted apoptosis of U87 cells and U251 cells (p < 0.05). CONCLUSION In conclusion, high CDC6 expression may serve as a promising biomarker for prognosis and correlated with immune infiltrates, presenting to be a potential immune therapy target in glioma.
Collapse
Affiliation(s)
- Feng Wang
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China. .,Department of Cancer Center Head and Neck, West China Hospital, Sichuan University, Chengdu, China.
| | - Fen Zhao
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China.,Department of Oncology, Chengdu First People's Hospital, Chengdu, 610041, Sichuan Province, China
| | - Li Zhang
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China.,Department of Cancer Center Head and Neck, West China Hospital, Sichuan University, Chengdu, China
| | - Lai Xiong
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China
| | - Qing Mao
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China
| | - Yanhui Liu
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China
| | - Xiaoguang Qiu
- Department of Radiotherapy, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100050, China
| | - Xiang Wang
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China
| | - Lin Shui
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China
| | - Xi Chen
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China
| | - Kexing Ren
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China.,Department of Cancer Center Head and Neck, West China Hospital, Sichuan University, Chengdu, China
| | - Pixian Shui
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Qiongwen Zhang
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China.,Department of Cancer Center Head and Neck, West China Hospital, Sichuan University, Chengdu, China
| | - Yifei Deng
- Department of Radiotherapy, Chengdu Seventh Hospital, Chengdu, China
| | - Weimin Li
- Center for Precision Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoqi Xie
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China.
| | - Dengbin Wu
- Cancer Hospital, An Steel Group General Hospital, Anshan, Liao Ning, People's Republic of China.
| | - Tao Li
- Department of Radiotherapy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610041, Sichuan, China
| | - Jinyi Lang
- Department of Radiotherapy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610041, Sichuan, China
| | - Lei Liu
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China.,Department of Cancer Center Head and Neck, West China Hospital, Sichuan University, Chengdu, China
| | - Huaying Chen
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China.,Department of Cancer Center Head and Neck, West China Hospital, Sichuan University, Chengdu, China
| | - Jianguo Xu
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China
| | - Sen Bai
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China
| | - Zhiping Li
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China
| | - Qiang Yue
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China
| | - Ni Chen
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China
| | - Bingwen Zhou
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China
| | - Cheng Yi
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China.
| | - Yuquan Wei
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China
| | - Yuchuan Fu
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China
| | - Yong Luo
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China
| | - Qiheng Gou
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China.,Department of Cancer Center Head and Neck, West China Hospital, Sichuan University, Chengdu, China
| | - Lunxu Liu
- Department of Internal Medicine, West China Hospital Cancer Center Head And Neck, Sichuan University, Chengdu, China
| | - Yuanzhao Liu
- Department of Radiotherapy, Beijing Hospital, Beijing, People's Republic of China
| | - Jingbo Kang
- Department of Radiotherapy, The sixth Medical Center of PLA General Hospital, Beijing, People's Republic of China
| | - Junjie Wang
- Department of Radiation Oncology, Peking University Third Hospital, No. 49, Beijing, China
| | | | - Fuquan Zhang
- Department of Radiation Oncology, Beijing Union Medical College Hospital, Beijing, China
| | - Xiaoyan Yang
- Department of Radiotherapy, First Hospital of Shan Xi Medical, University, Taiyuan West, Taiyuan, China
| | - Xianfeng Li
- Department of Radiotherapy, First Hospital of Shan Xi Medical, University, Taiyuan West, Taiyuan, China
| | - Tao Jiang
- Department of Radiotherapy, Beijing Tian Tan Hospital, Capital Medical University, Beijing, 100050, China
| | - Zongcun Zhang
- Qing Dao Central Hospital, 127 Si Liu South Road, Shi Bei District, Qing Dao, Shan Dong Province, China
| | - Yizhi Zhou
- Shanghai High-Tech United Bio-Technological R&D Co., Ltd, Shanghai, China
| | - Junlin Yi
- Department of Radiation Oncology, National Cancer Center National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
8
|
Zhang LQ, Zhou SL, Li JK, Chen PN, Zhao XK, Wang LD, Li XL, Zhou FY. Identification of a seven-cell cycle signature predicting overall survival for gastric cancer. Aging (Albany NY) 2022; 14:3989-3999. [PMID: 35537781 PMCID: PMC9134949 DOI: 10.18632/aging.204060] [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: 01/15/2022] [Accepted: 04/25/2022] [Indexed: 11/25/2022]
Abstract
While genetic alterations in several regulators of the cell cycle have a significant impact on the gastric carcinogenesis process, the prognostic role of them remains to be further elucidated. The TCGA-STAD training set were downloaded and the mRNA expression matrix of cell cycle genes was extracted and corrected for further analysis after taking the intersection with GSE84437 dataset. Differentially expressed mRNAs were identified between tumor and normal tissue samples in TCGA-STAD. Univariate Cox regression analysis and lasso Cox regression model established a novel seven-gene cell cycle signature (including GADD45B, TFDP1, CDC6, CDC25A, CDC7, SMC1A and MCM3) for GC prognosis prediction. Patients in the high-risk group shown significantly poorer survival than patients in the low-risk group. The signature was found to be an independent prognostic factor for GC survival. Nomogram including the signature shown some clinical net benefit for overall survival prediction. The signature was further validated in the GSE84437 dataset. In tissue microarray, CDC6 and MCM3 protein expression were significant differences by the immunohistochemistry-based H-score between tumor tissues and adjacent tissues, and CDC6 is an independent prognostic factor for GC. Interestingly, our GSEA revealed that low-risk patients were more related to cell cycle pathways and might benefit more from therapies targeting cell cycle. Our study identified a novel robust seven-gene cell cycle signature for GC prognosis prediction that may serve as a beneficial complement to clinicopathological staging. The signature might provide potential biomarkers for the application of cell cycle regulators to therapies and treatment response prediction.
Collapse
Affiliation(s)
- Lian-Qun Zhang
- Department of Gastroenterology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou 450003, Henan, China
| | - Sheng-Li Zhou
- Department of Pathology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou 450003, Henan, China
| | - Jun-Kuo Li
- Department of Thoracic Surgery, Anyang Tumor Hospital, Anyang 455000, Henan, China
| | - Pei-Nan Chen
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450008, Henan, China
| | - Xue-Ke Zhao
- State Key Laboratory of Esophageal Cancer Prevention and Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou 450003, Henan, China
| | - Li-Dong Wang
- State Key Laboratory of Esophageal Cancer Prevention and Treatment and Henan Key Laboratory for Esophageal Cancer Research of the First Affiliated Hospital, Zhengzhou University, Zhengzhou 450003, Henan, China
| | - Xiu-Ling Li
- Department of Gastroenterology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou 450003, Henan, China
| | - Fu-You Zhou
- Department of Thoracic Surgery, Anyang Tumor Hospital, Anyang 455000, Henan, China
| |
Collapse
|
9
|
Mahabady MK, Mirzaei S, Saebfar H, Gholami MH, Zabolian A, Hushmandi K, Hashemi F, Tajik F, Hashemi M, Kumar AP, Aref AR, Zarrabi A, Khan H, Hamblin MR, Nuri Ertas Y, Samarghandian S. Noncoding RNAs and their therapeutics in paclitaxel chemotherapy: Mechanisms of initiation, progression, and drug sensitivity. J Cell Physiol 2022; 237:2309-2344. [PMID: 35437787 DOI: 10.1002/jcp.30751] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 12/16/2022]
Abstract
The identification of agents that can reverse drug resistance in cancer chemotherapy, and enhance the overall efficacy is of great interest. Paclitaxel (PTX) belongs to taxane family that exerts an antitumor effect by stabilizing microtubules and inhibiting cell cycle progression. However, PTX resistance often develops in tumors due to the overexpression of drug transporters and tumor-promoting pathways. Noncoding RNAs (ncRNAs) are modulators of many processes in cancer cells, such as apoptosis, migration, differentiation, and angiogenesis. In the present study, we summarize the effects of ncRNAs on PTX chemotherapy. MicroRNAs (miRNAs) can have opposite effects on PTX resistance (stimulation or inhibition) via influencing YES1, SK2, MRP1, and STAT3. Moreover, miRNAs modulate the growth and migration rates of tumor cells in regulating PTX efficacy. PIWI-interacting RNAs, small interfering RNAs, and short-hairpin RNAs are other members of ncRNAs regulating PTX sensitivity of cancer cells. Long noncoding RNAs (LncRNAs) are similar to miRNAs and can modulate PTX resistance/sensitivity by their influence on miRNAs and drug efflux transport. The cytotoxicity of PTX against tumor cells can also be affected by circular RNAs (circRNAs) and limitation is that oncogenic circRNAs have been emphasized and experiments should also focus on onco-suppressor circRNAs.
Collapse
Affiliation(s)
- Mahmood K Mahabady
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Hamidreza Saebfar
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad H Gholami
- Faculty of Veterinary Medicine, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Amirhossein Zabolian
- Resident of Orthopedics, Department of Orthopedics, School of Medicine, 5th Azar Hospital, Golestan University of Medical Sciences, Golestan, Iran
| | - Kiavash Hushmandi
- Division of Epidemiology, Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Farid Hashemi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Fatemeh Tajik
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Alan P Kumar
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Pharmacology, Cancer Science Institute of Singapore, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Amir R Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA.,Xsphera Biosciences Inc, Boston, Massachusetts, USA
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul, Turkey
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan, Pakistan
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey.,ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Turkey
| | - Saeed Samarghandian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
10
|
Song S, Wang Y, Liu P. DNA Replication Licensing Factors: Novel Targets for Cancer Therapy via Inhibiting the Stemness of Cancer Cells. Int J Biol Sci 2022; 18:1211-1219. [PMID: 35173548 PMCID: PMC8771848 DOI: 10.7150/ijbs.67529] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/17/2021] [Indexed: 11/19/2022] Open
Abstract
The replication licensing factors strictly regulate the DNA replication origin licensing process to guarantee the stability of the genome. Numerous experimental studies have recently demonstrated that the replication licensing factors as oncogenes are essential for the occurrence and development of cancers. Drug resistance, being one of the main characteristics of cancer stem cells, can cause a high recurrence rate and a low survival rate in patients with different cancers. However, the function of the replication licensing factors in cancer stemness remains unclear. The following article highlights the most recent research on DNA replication origin licensing factors in cancer and their function in anti-cancer drug resistance. Moreover, this article proposes a new perspective that replication licensing factors as chemotherapy shield affect anti-cancer drug resistance by promoting the stemness of cancer cells.
Collapse
Affiliation(s)
- Shaoran Song
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- The Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yaochun Wang
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- The Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Peijun Liu
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- The Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| |
Collapse
|
11
|
Yang C, Deng S. Hsa_circ_0017728 as an oncogene in gastric cancer by sponging miR-149 and modulating the IL-6/STAT3 pathway. Arch Med Sci 2022; 18:1558-1571. [PMID: 36457988 PMCID: PMC9710264 DOI: 10.5114/aoms.2019.87274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 06/03/2019] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Circular RNAs (circRNAs) have been identified as competing endogenous RNAs (ceRNAs) to mediate gene expression participating in the progression of multiple cancers, including gastric carcinoma (GC). However, the underlying molecular mechanisms by which circRNAs-modulated cell proliferation and apoptosis in GC had not been completely clarified. In our study, hsa_circ_0017728 as a potential oncogene competing endogenous RNA (ceRNA) was investigated in the progression and development of gastric carcinogenesis. MATERIAL AND METHODS High-throughput sequencing was used to determine differentially expressed circRNAs in GC tissues and corresponding non-cancerous tissues. The CCK-8 assay and Annexin V-fluorescein isothiocyanate/polyimide (Annexin V-FITC/PI) staining were performed to detect the cell viability and apoptosis in GC cells. In addition, gene expression and protein levels in GC tissues and cell lines were measured using RT-qPCR and western blotting, respectively. RESULTS Our results demonstrated that the hsa_circ_0017728 expression level was up-regulated in GC tissues and cell lines and closely associated with poor overall survival and pathological differentiation, higher TNM stage and lymph node metastasis. Knockdown of hsa_circ_0017728 had the ability to cause inhibition of cell proliferation and migration and elevate the cell apoptosis rate in GC cells. We also discovered that hsa_circ_0017728 might serve as a ceRNA to sponge miR-149 and indirectly regulated the IL-6/STAT3 signaling pathway in GC cell proliferation and apoptosis. CONCLUSIONS The regulatory network of hsa_circ_0017728/miR-149/IL-6/STAT3 cascade signaling might provide a better understanding of gastric carcinogenesis and progression.
Collapse
Affiliation(s)
- Chun Yang
- School of Medicine, University of Electronic Science and Technology of China; Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Shaoping Deng
- School of Medicine, University of Electronic Science and Technology of China; Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| |
Collapse
|
12
|
Qu J, Lin Z. Autophagy Regulation by Crosstalk between miRNAs and Ubiquitination System. Int J Mol Sci 2021; 22:ijms222111912. [PMID: 34769343 PMCID: PMC8585084 DOI: 10.3390/ijms222111912] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/27/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are non-coding single-stranded RNA molecules encoded by endogenous genes with ~22 nucleotides which are involved in the regulation of post-transcriptional gene expression. Ubiquitination and deubiquitination are common post-translational modifications in eukaryotic cells and important pathways in regulating protein degradation and signal transduction, in which E3 ubiquitin ligases and deubiquitinases (DUBs) play a decisive role. MiRNA and ubiquitination are involved in the regulation of most biological processes, including autophagy. Furthermore, in recent years, the direct interaction between miRNA and E3 ubiquitin ligases or deubiquitinases has attracted much attention, and the cross-talk between miRNA and ubiquitination system has been proved to play key regulatory roles in a variety of diseases. In this review, we summarized the advances in autophagy regulation by crosstalk between miRNA and E3 ubiquitin ligases or deubiquitinases.
Collapse
|
13
|
Zhou X, Xue D, Qiu J. Identification of biomarkers related to glycolysis with weighted gene co-expression network analysis in oral squamous cell carcinoma. Head Neck 2021; 44:89-103. [PMID: 34713497 DOI: 10.1002/hed.26910] [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: 11/16/2020] [Revised: 09/10/2021] [Accepted: 10/05/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is the most common tumor in the oral cavity and maxillofacial region. Increasing evidence suggests that aerobic glycolysis plays an important role in the occurrence, development, and prognosis of OSCC. Therefore, the identification of biomarkers related to glycolysis in OSCC represents considerable potential for improving its treatment. METHODS In the present study, a single-sample gene-set enrichment analysis (ssGSEA) algorithm with weighted gene co-expression network analysis (WGCNA) were used to quantify the degree of glycolysis and identify key modules with the greatest correlation with glycolysis. RESULTS Glycolytic scores significantly correlated with prognosis. In the key module 5 HUB genes were finally selected, which displayed a robust predictive effect. The expressions of key genes were associated with glycolysis. CONCLUSIONS The research comprehensively analyzed the glycolysis of OSCC and identified several biomarkers related to glycolysis. These biomarkers may represent potential therapeutic targets for future OSCC therapy.
Collapse
Affiliation(s)
- Xiongming Zhou
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Danfeng Xue
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jiaxuan Qiu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
14
|
Kong DG, Yao FZ. CDC6 is a possible biomarker for hepatocellular carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2021; 14:811-818. [PMID: 34367412 PMCID: PMC8339721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 05/17/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVE Research has proven that the expression of CDC6 is tightly related to tumorigenesis and progression of various tumors. However, the effects of CDC6 in hepatocellular carcinoma remain uncertain. The main purpose of this research is to explore this relationship. METHODS We assessed the expression levels of CDC6 in a serious of cancers from GEPIA database. The expression of CDC6 in hepatocellular carcinoma tissue and normal liver tissue was compared, and further assessed by immunohistochemical staining. Graphpad software was performed for data analysis, and t-test and χ2 analysis were used to investigate the role of CDC6 in hepatocellular carcinoma. RESULTS The expression level of CDC6 was significantly higher in malignant carcinoid, melanoma, urothelial tumor, and hepatocellular carcinoma in the GEPIA online database. It was related to clinical progression of hepatocellular carcinoma. We found that the expression of CDC6 was correlated with tumor size (P=0.018) and the number of tumor nodes (P=0.003), but not with age, gender and AFP value (P>0.05). CONCLUSIONS The expression level of CDC6 in hepatocellular carcinoma is related tightly to clinical findings. Detecting the expression of CDC6 might provide a new biomarker for patients with hepatocellular carcinoma.
Collapse
Affiliation(s)
- De-Gang Kong
- Department of Hepatobiliary and Pancreatic Surgery, The Secondary Hospital of Tianjin Medical University No. 23 Pingjiang Road, Hexi District, Tianjin 300211, China
| | - Fu-Zhou Yao
- Department of Hepatobiliary and Pancreatic Surgery, The Secondary Hospital of Tianjin Medical University No. 23 Pingjiang Road, Hexi District, Tianjin 300211, China
| |
Collapse
|
15
|
Jiang Y, Zhang X, Rong L, Hou Y, Song J, Zhang W, He M, Xie Y, Li Y, Song F. Integrative analysis of the gastric cancer long non-coding RNA-associated competing endogenous RNA network. Oncol Lett 2021; 21:456. [PMID: 33907566 PMCID: PMC8063256 DOI: 10.3892/ol.2021.12717] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/10/2021] [Indexed: 12/18/2022] Open
Abstract
Gastric cancer (GC) is a common type of cancer, and identification of novel diagnostic biomarkers associated with this disease is important. The present study aimed to identify novel diagnostic biomarkers associated with the prognosis of GC, using an integrated bioinformatics approach. Differentially expressed long non-coding RNAs (lncRNAs) associated with GC were identified using Gene Expression Omnibus datasets (GSE58828, GSE72305 and GSE99416) and The Cancer Genome Atlas database. A competing endogenous RNA network that incorporated five lncRNAs [long intergenic non-protein coding RNA 501 (LINC00501), LINC00365, SOX21 antisense divergent transcript 1 (SOX21-AS1), GK intronic transcript 1 (GK-IT1) and DLEU7 antisense RNA 1 (DLEU7-AS1)], 29 microRNAs and 114 mRNAs was constructed. Gene Ontology and protein-protein interaction network analyses revealed that these lncRNAs may be involved in 'biological regulation', 'metabolic process', 'cell communication', 'developmental process', 'cell proliferation', 'reproduction' and the 'cell cycle'. The results of receiver operating characteristic curve analysis demonstrated that LINC00501 (AUC=0.819), LINC00365 (AUC=0.580), SOX21-AS1 (AUC=0.736), GK-IT1 (AUC=0.823) and DLEU7-AS1 (AUC=0.932) had the potential to become valuable diagnostic biomarkers for GC. Associations with clinicopathological characteristics demonstrated that LINC00501 expression was significantly associated with sex (P=0.015) and tumor grade (P=0.022). Furthermore, LINC00365 expression was significantly associated with lymph node metastasis (P=0.025). Gene set enrichment analysis revealed that LINC00501, LINC00365 and SOX21-AS1 were enriched in signaling pathways associated with GC. Reverse transcription-quantitative PCR analysis demonstrated that LINC00501 expression (P=0.043) was significantly upregulated in GC tissues, whereas the expression levels of LINC00365 (P=0.033) and SOX21-AS1 (P=0.037) were significantly downregulated in GC tissues. Taken together, the results of the present study suggest that LINC00501, LINC00365, SOX21-AS1, GK-IT1 and DLEU7-AS1 may be used as novel diagnostic biomarkers for GC, and may be functionally associated with GC development and progression.
Collapse
Affiliation(s)
- Yuyou Jiang
- Basic Medical College, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xianqin Zhang
- School of Basic Medical Sciences, Chengdu Medical College, Chengdu, Sichuan 610500, P.R. China
| | - Li Rong
- Department of Infectious Disease, Chongqing Public Health Medical Center, Chongqing 400036, P.R. China
| | - Yi Hou
- Experimental Teaching and Management Center, Chongqing Medical University, Chongqing 401331, P.R. China
| | - Jing Song
- Basic Medical College, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Wanfeng Zhang
- Department of Bioinformatics, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Min He
- Basic Medical College, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yan Xie
- Basic Medical College, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yue Li
- Basic Medical College, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Fangzhou Song
- Basic Medical College, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
16
|
Yicong Y, Wang Y, Denglong W, Baoying H. Increased CDC6 Expression Associates With Poor Prognosis in Patients With Clear Cell Renal Cell Carcinoma. Front Oncol 2021; 11:666418. [PMID: 34136398 PMCID: PMC8202290 DOI: 10.3389/fonc.2021.666418] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/21/2021] [Indexed: 12/12/2022] Open
Abstract
Background CDC6 (Cell division control protein 6), located at chromosome 17q21.3, plays an important role in the early stage of DNA replication and has unique functions in various malignant tumors. Here, we evaluate the relationship between CDC6 expression and oncology outcomes in patients with clear cell renal cell carcinoma (ccRCC). Methods A retrospective analysis of 118 ccRCC patients in Affiliated Hospital of Nantong University from 2015 to 2017 was performed. Triplicate tissue microarrays (TMA) were prepared from formalin-fixed and paraffin-embedded specimens. Immunohistochemistry (IHC) was conducted to evaluate the relationship between CDC6 expression and standard pathological features and prognosis. The RNA sequencing data and corresponding clinical information were acquired from the TCGA database. GSEA was used to identify signal pathways related to CDC6. Cox regression analysis was used to assess independent prognostic factors. In addition, the relationship between CDC6 and immunity was also investigated. Results The results of Kaplan–Meier curve indicated that the OS of the patients with high expression of CDC6 was shorter than that of the patients with low CDC6 expression. Integrating the TCGA database and IHC staining, the results showed that CDC6 in ccRCC tissue was obviously up-regulated compared with adjacent normal kidney tissue. The results of Logistic regression analysis demonstrated that ccRCC patients with high expression of CDC6 are more likely to develop advanced disease than ccRCC patients with low CDC6 expression. The results of GSEA showed that the high expression of CDC6 was related to multiple signaling pathways. As for immunity, it was also related to TMB, immune checkpoint molecules, tumor microenvironment and immune infiltration. There were significantly correlations with CDC6 and immune cell infiltration levels and tumor microenvironment. The results of further results of the TCGA database showed that CDC6 was obviously related to immune checkpoint molecules and immune cells. Conclusions Increased expression of CDC6 is a potentially prognostic factor of poor prognosis in ccRCC patients.
Collapse
Affiliation(s)
- Yao Yicong
- School of Medicine, Tongji University, Shanghai, China.,Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Wang
- Department of Urology, Affiliated Hospital of Nantong University, Nantong, China
| | - Wu Denglong
- Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hu Baoying
- Department of Immunology, Medical College, Nantong University, Shanghai, China
| |
Collapse
|
17
|
Ji Y, Gao L, Zhang C, Sun X, Dai L, Ji Z, Zhang J, Zhang Z, Cao W, Zhao Y, Zhang L. Identification of the hub genes and prognostic indicators of gastric cancer and correlation of indicators with tumor-infiltrating immune cell levels. J Cancer 2021; 12:4025-4038. [PMID: 34093807 PMCID: PMC8176244 DOI: 10.7150/jca.52105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 04/25/2021] [Indexed: 12/29/2022] Open
Abstract
Aims: To identify the hub genes and prognostic indicators of gastric cancer (GC) and determine the correlation between prognostic indicators and the tumor-infiltrating immune cell levels so as to provide useful information for future GC diagnosis and treatment. Methods: The Cancer Genome Atlas (TCGA) stomach adenocarcinoma dataset and two microarray datasets were used to screen the overlapping differentially expressed genes (DEGs) between normal gastric and GC tissue samples. Hub genes were screened via protein-protein interaction networks and module analysis of the overlapping DEGs. Their expression was validated at the cell level and tissue level using the ONCOMINE database. The prognostic indicators of overall survival (OS) and disease-free survival was identified by Cox proportional hazards regression analysis based on tumor grade and cancer stage. The expression of hub genes was validated at the cell level. The correlation of prognostic indicators with the tumor-infiltrating immune cell levels was analyzed using Tumor IMmune Estimation Resource. Results: Ten hub genes, namely CDC6, CDC20, BUB1B, TOP2A, CDK1, AURKA, CCNA2, CCNB1, MAD2L1, and KIF11, were screened and their upregulation in the GC tissue was verified. Three prognostic factors, namely LUM, VCAN, and EFNA4, were identified; their expression was higher in GC cells than in normal cells. LUM, VCAN, and EFNA4 were correlated with tumor-infiltrating immune cell levels in GC. Significance: The identified hub genes and prognostic indicators of GC could be useful indicators for future GC diagnosis and treatment.
Collapse
Affiliation(s)
- Yun Ji
- BGI College, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450007, China.,Henan Institute of Medical and Pharmaceutical Science, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450052, China
| | - Lu Gao
- BGI College, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450007, China.,Henan Institute of Medical and Pharmaceutical Science, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450052, China
| | - Can Zhang
- BGI College, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450007, China.,Henan Institute of Medical and Pharmaceutical Science, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450052, China
| | - Xu Sun
- Integrated TCM and Western Medicine Department, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, China
| | - Liping Dai
- Henan Institute of Medical and Pharmaceutical Science, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450052, China
| | - Zhenyu Ji
- Henan Institute of Medical and Pharmaceutical Science, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450052, China
| | - Jianying Zhang
- Henan Institute of Medical and Pharmaceutical Science, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450052, China
| | - Zhida Zhang
- BGI College, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450007, China.,Henan Institute of Medical and Pharmaceutical Science, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450052, China
| | - Wei Cao
- Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou 450000, China
| | - Yang Zhao
- Henan Institute of Medical and Pharmaceutical Science, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450052, China
| | - Liguo Zhang
- Henan Institute of Medical and Pharmaceutical Science, Zhengzhou University, No. 40 Daxue Road, Zhengzhou 450052, China.,Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou 450000, China
| |
Collapse
|
18
|
MiR-27a-3p/miR-27b-3p Promotes Neurofibromatosis Type 1 via Targeting of NF1. J Mol Neurosci 2021; 71:2353-2363. [PMID: 33570696 DOI: 10.1007/s12031-020-01779-2] [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: 06/19/2020] [Accepted: 12/15/2020] [Indexed: 10/22/2022]
Abstract
The dysregulation of microRNAs (miRNAs) is a crucial molecular signature of disease development. The potential implication of miRNAs in neurofibromatosis type 1 (NF1) remains poorly investigated. The expression levels of miR-27a-3p, miR-27b-3p, and neurofibromin 1 (NF1) were detected by real-time quantitative polymerase chain reaction (RT-qPCR) analysis. The functional roles of miR-27a-3p and miR-27b-3p in NF1 were explored by CCK8 (Cell Counting Kit-8), 5-ethynyl-2'-deoxyuridine (EdU), terminal deoxynucleoitidyl transferase dUTP nick-end labeling (TUNEL), and transwell assays. Luciferase reporter, RNA pull-down, and RNA binding protein immunoprecipitation (RIP) assays were employed to study the probable target relationship between miRNA and messenger RNA (mRNA). MiR-27b-3p and miR-27a-3p were upregulated in dermal and plexiform human Schwann cells (HSC) from NF1 neurofibromas as well as cell lines of malignant peripheral nerve sheath tumors (MPNSTs). MiR-27a-3p/miR-27b-3p mimics promoted the proliferative, migratory, and invasive ability of dermal HSC and MPNST cell ST88-14, while inhibiting the apoptotic capacity. MiR-27a-3p/miR-27b-3p inhibitors elicited the opposite impacts on the above cellular behaviors in dermal HSC and ST88-14. Intriguingly, NF1 was revealed to be the target of both miR-27a-3p and miR-27b-3p, and was negatively modulated by them. MiR-27a-3p/miR-27b-3p upregulation suppressed the expression of NF1 in dermal HSC and ST88-14. Furthermore, NF1 depletion counterbalanced the functional alteration induced by miR-27a-3p/miR-27b-3p inhibition. Our study suggests that both miR-27b-3p and miR-27a-3p are involved in upstream molecular activity responsible for the depletion of NF1, representing promising targets for therapeutic application in NF1.
Collapse
|
19
|
Marengo B, Pulliero A, Corrias MV, Leardi R, Farinini E, Fronza G, Menichini P, Monti P, Monteleone L, Valenti GE, Speciale A, Perri P, Madia F, Izzotti A, Domenicotti C. Potential Role of miRNAs in the Acquisition of Chemoresistance in Neuroblastoma. J Pers Med 2021; 11:jpm11020107. [PMID: 33562297 PMCID: PMC7916079 DOI: 10.3390/jpm11020107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/20/2021] [Accepted: 02/04/2021] [Indexed: 02/06/2023] Open
Abstract
Neuroblastoma (NB) accounts for about 8–10% of pediatric cancers, and the main causes of death are the presence of metastases and the acquisition of chemoresistance. Metastatic NB is characterized by MYCN amplification that correlates with changes in the expression of miRNAs, which are small non-coding RNA sequences, playing a crucial role in NB development and chemoresistance. In the present study, miRNA expression was analyzed in two human MYCN-amplified NB cell lines, one sensitive (HTLA-230) and one resistant to Etoposide (ER-HTLA), by microarray and RT-qPCR techniques. These analyses showed that miRNA-15a, -16-1, -19b, -218, and -338 were down-regulated in ER-HTLA cells. In order to validate the presence of this down-regulation in vivo, the expression of these miRNAs was analyzed in primary tumors, metastases, and bone marrow of therapy responder and non-responder pediatric patients. Principal component analysis data showed that the expression of miRNA-19b, -218, and -338 influenced metastases, and that the expression levels of all miRNAs analyzed were higher in therapy responders in respect to non-responders. Collectively, these findings suggest that these miRNAs might be involved in the regulation of the drug response, and could be employed for therapeutic purposes.
Collapse
Affiliation(s)
- Barbara Marengo
- Department of Experimental Medicine, University of Genova, 16100 Genova, Italy; (L.M.); (G.E.V.); (A.I.); (C.D.)
- Correspondence: ; Tel.: +39-010-3538831
| | | | - Maria Valeria Corrias
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16100 Genova, Italy; (M.V.C.); (P.P.)
| | - Riccardo Leardi
- Department of Pharmacy, University of Genova, 16100 Genova, Italy; (R.L.); (E.F.)
| | - Emanuele Farinini
- Department of Pharmacy, University of Genova, 16100 Genova, Italy; (R.L.); (E.F.)
| | - Gilberto Fronza
- UOC Mutagenesis and Cancer Prevention, IRCCS Ospedale Policlinico San Martino, 16100 Genova, Italy; (G.F.); (P.M.); (P.M.); (A.S.)
| | - Paola Menichini
- UOC Mutagenesis and Cancer Prevention, IRCCS Ospedale Policlinico San Martino, 16100 Genova, Italy; (G.F.); (P.M.); (P.M.); (A.S.)
| | - Paola Monti
- UOC Mutagenesis and Cancer Prevention, IRCCS Ospedale Policlinico San Martino, 16100 Genova, Italy; (G.F.); (P.M.); (P.M.); (A.S.)
| | - Lorenzo Monteleone
- Department of Experimental Medicine, University of Genova, 16100 Genova, Italy; (L.M.); (G.E.V.); (A.I.); (C.D.)
| | - Giulia Elda Valenti
- Department of Experimental Medicine, University of Genova, 16100 Genova, Italy; (L.M.); (G.E.V.); (A.I.); (C.D.)
| | - Andrea Speciale
- UOC Mutagenesis and Cancer Prevention, IRCCS Ospedale Policlinico San Martino, 16100 Genova, Italy; (G.F.); (P.M.); (P.M.); (A.S.)
| | - Patrizia Perri
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, 16100 Genova, Italy; (M.V.C.); (P.P.)
| | - Francesca Madia
- Medical Genetics Unit, IRCCS Giannina Gaslini Institute, 16100 Genova, Italy;
| | - Alberto Izzotti
- Department of Experimental Medicine, University of Genova, 16100 Genova, Italy; (L.M.); (G.E.V.); (A.I.); (C.D.)
- UOC Mutagenesis and Cancer Prevention, IRCCS Ospedale Policlinico San Martino, 16100 Genova, Italy; (G.F.); (P.M.); (P.M.); (A.S.)
| | - Cinzia Domenicotti
- Department of Experimental Medicine, University of Genova, 16100 Genova, Italy; (L.M.); (G.E.V.); (A.I.); (C.D.)
| |
Collapse
|
20
|
Ghafouri-Fard S, Shoorei H, Abak A, Abbas Raza SH, Pichler M, Taheri M. Role of non-coding RNAs in modulating the response of cancer cells to paclitaxel treatment. Biomed Pharmacother 2020; 134:111172. [PMID: 33360156 DOI: 10.1016/j.biopha.2020.111172] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/12/2020] [Accepted: 12/15/2020] [Indexed: 02/07/2023] Open
Abstract
Paclitaxel is a chemotherapeutic substance that is administered for treatment of an extensive spectrum of human malignancies. In spite of its potent short-term effects against tumor cells, resistance to paclitaxel occurs in a number of patients precluding its long-term application in these patients. Non-coding RNAs have been shown to influence response of cancer cells to this chemotherapeutic agent via different mechanisms. Mechanistically, these transcripts regulate expression of several genes particularly those being involved in the apoptotic processes. Lots of in vivo and in vitro assays have demonstrated the efficacy of oligonucleotide-mediated microRNAs (miRNA)/ long non-coding RNAs (lncRNA) silencing in enhancement of response of cancer cells to paclitaxel. Therefore, targeted therapies against non-coding RNAs have been suggested as applicable modalities for combatting resistance to this agent. In the present review, we provide a summary of studies which assessed the role of miRNAs and lncRNAs in conferring resistance to paclitaxel.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Atefe Abak
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sayed Haidar Abbas Raza
- College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang, China
| | - Martin Pichler
- Research Unit of Non-Coding RNAs and Genome Editing in Cancer, Division of Clinical Oncology, Department of Internal Medicine, Comprehensive Cancer Center Graz, Medical University of Graz, 8036 Graz, Austria; Department of Experimental Therapeutics, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
21
|
Wang G, Xu G, Wang W. Long Noncoding RNA CDKN2B-AS1 Facilitates Lung Cancer Development Through Regulating miR-378b/NR2C2. Onco Targets Ther 2020; 13:10641-10649. [PMID: 33116641 PMCID: PMC7585785 DOI: 10.2147/ott.s261973] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 09/02/2020] [Indexed: 12/13/2022] Open
Abstract
Aim Long noncoding RNA (lncRNA) have proved to be important regulators in various diseases. CDKN2B-AS1 was a newly identified tumor-related lncRNA, and previous studies have reported its function in laryngeal squamous cancer and osteosarcoma. However, the function and mechanism of lncRNA CDKN2B-AS1 in lung cancer are still unknown. Methods Cell proliferation, invasion, migration and apoptosis were detected via CCK-8, transwell assay and Western blot. Bioinformatics analysis was used to predict the potential target of CDKN2B-AS1. A rescue experiment was performed to identify the relationship between CDKN2B-AS1 and miR-378b. Results The expression of lncRNA CDKN2B-AS1 was significantly upregulated in lung cancer tissues and cell lines. Overexpression of CDKN2B-AS1 promoted cell proliferation, invasion and reduced cell apoptosis. Knockdown of CDKN2B-AS1 inhibited cell proliferation, invasion and increased cell apoptosis. Bioinformatics analysis predicted that miR-378b was the direct target. We also provided evidence that NR2C2 was the target of miR-378b. The expression of NR2C2 was significantly upregulated in lung cancer tissues and cell lines. The rescue experiment further confirmed the relationship between CDKN2B-AS1 and miR-378b. Overexpression of miR-378b completely reversed the function of CDKN2B-AS1. Conclusion Taken together, our results comprehensively analyzed the function of CDKN2B-AS1 in lung cancer and provided a possible mechanism that CDKN2B-AS1 facilitates lung cancer development by regulating miR-378b and NR2C2. Thus, our study offers a potential therapeutic target for treating lung cancer.
Collapse
Affiliation(s)
- Guolei Wang
- Department of Thoracic Oncology, Henan Chest Hospital, Zhengzhou, Henan, People's Republic of China
| | - Guanghui Xu
- Department of Thoracic Oncology, Henan Chest Hospital, Zhengzhou, Henan, People's Republic of China
| | - Wenguang Wang
- Department of Thoracic Oncology, Henan Chest Hospital, Zhengzhou, Henan, People's Republic of China
| |
Collapse
|
22
|
Youn Y, Lee JC, Kim J, Kim JH, Hwang JH. Cdc6 disruption leads to centrosome abnormalities and chromosome instability in pancreatic cancer cells. Sci Rep 2020; 10:16518. [PMID: 33020506 PMCID: PMC7536414 DOI: 10.1038/s41598-020-73474-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
Cell division cycle 6 (Cdc6) plays key roles in regulating DNA replication, and activation and maintenance of cell cycle check points. In addition, Cdc6 exerts oncogenic properties via genomic instability associated with incomplete DNA replication. This study aimed to examine the effects of Cdc6 on pancreatic cancer (PC) cells. Our results showed that Cdc6 expression was higher in clinical PC specimens (based on analysis of the GEPIA database) and cell lines, and the high Cdc6 expression was associated with poorer survival in The Cancer Genome Atlas-PC cohort. In addition, Cdc6-depleted PC cells significantly inhibited cell proliferation and colony formation, delayed G2/M cell cycle progression, and increased expression of p-histone H3 and cyclin A2 levels. These observations could be explained by Cdc6 depletion leading to multipolar and split spindles via centrosome amplification and microtubule disorganization which eventually increases chromosome missegregation. Furthermore, Cdc6-depleted PC cells showed significantly increased apoptosis, which was consistent with increased caspase-9 and caspase-3 activation. Collectively, our results demonstrated that Cdc6-depleted PC cells are arrested in mitosis and eventually undergo cell death by induced multipolar spindles, centrosome aberrations, microtubule disorganization, and chromosome instability. In conclusion, Cdc6 may be a potential biomarker and therapeutic target for PC.
Collapse
Affiliation(s)
- Yuna Youn
- Department of Internal Medicine, Seoul National University Bundang Hospital, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea
| | - Jong-Chan Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jaihwan Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea
| | - Jae Hyeong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea.
| | - Jin-Hyeok Hwang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea. .,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| |
Collapse
|
23
|
Zhou J, Song Y, Gan W, Liu L, Chen G, Chen Z, Luo G, Zhang L, Zhang G, Wang P, Cao Y. Upregulation of COL8A1 indicates poor prognosis across human cancer types and promotes the proliferation of gastric cancer cells. Oncol Lett 2020; 20:34. [PMID: 32774507 PMCID: PMC7405348 DOI: 10.3892/ol.2020.11895] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 01/13/2020] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) was one of the most common types of the digestive system. COL8A1 was reported to be associated with cancer progression. The present study showed COL8A1 was overexpressed and correlated to shorter overall survival (OS) time across human cancer types. Specially, our results showed COL8A1 was up-regulated in advanced stage GC compared to low stage GC samples. Higher expression of COL8A1 was significantly correlated to shorter OS time in patients with GC. Bioinformatics analysis revealed COL8A1 was involved in regulating cell proliferation and metastasis. Experimental validations of COL8A1 showed that silencing of COL8A1 could significantly suppressed cell proliferation, migration and invasion in GC. These results provided a potential target for the clinical prognosis and treatment of gastric cancer.
Collapse
Affiliation(s)
- Jun Zhou
- Department of Gastrointestinal Surgery, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, P.R. China
| | - Yaning Song
- Department of Gastrointestinal Surgery, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, P.R. China
| | - Wei Gan
- Department of Gastrointestinal Surgery, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, P.R. China
| | - Liye Liu
- Department of Gastrointestinal Surgery, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, P.R. China
| | - Guibing Chen
- Department of Gastrointestinal Surgery, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, P.R. China
| | - Zhenyu Chen
- Department of Gastrointestinal Surgery, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, P.R. China
| | - Guode Luo
- Department of Gastrointestinal Surgery, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, P.R. China
| | - Lin Zhang
- Department of Gastrointestinal Surgery, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, P.R. China
| | - Guohu Zhang
- Department of Gastrointestinal Surgery, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, P.R. China
| | - Peihong Wang
- Department of Gastrointestinal Surgery, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, P.R. China
| | - Yongkuan Cao
- Department of Gastrointestinal Surgery, The General Hospital of Western Theater Command, Chengdu, Sichuan 610083, P.R. China
| |
Collapse
|
24
|
Non-coding RNAs in drug resistance of head and neck cancers: A review. Biomed Pharmacother 2020; 127:110231. [PMID: 32428836 DOI: 10.1016/j.biopha.2020.110231] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/30/2020] [Accepted: 05/03/2020] [Indexed: 02/06/2023] Open
Abstract
Head and neck cancer (HNC), which includes epithelial malignancies of the upper aerodigestive tract (oral cavity, oropharynx, pharynx, hypopharynx, larynx, and thyroid), are slowly but consistently increasing, while the overall survival rate remains unsatisfactory. Because of the multifunctional anatomical intricacies of the head and neck, disease progression and therapy-related side effects often severely affect the patient's appearance and self-image, as well as their ability to breathe, speak, and swallow. Patients with HNC require a multidisciplinary approach involving surgery, radiation therapy, and chemotherapeutics. Chemotherapy is an important part of the comprehensive treatment of tumors, especially advanced HNC, but drug resistance is the main cause of poor clinical efficacy. The most important determinant of this phenomenon is still largely unknown. Recent studies have shown that non-coding RNAs have a crucial role in HNC drug resistance. In addition, they can serve as biomarkers in the diagnosis, treatment, and prognosis of HNCs. In this review, we summarize the relationship between non-coding RNAs and drug resistance of HNC, and discuss their potential clinical application in overcoming HNC chemoresistance.
Collapse
|
25
|
Yang ZH, Li J, Chen WZ, Kong FS. Oncogenic gene RGC-32 is a direct target of miR-26b and facilitates tongue squamous cell carcinoma aggressiveness through EMT and PI3K/AKT signalling. Cell Biochem Funct 2020; 38:943-954. [PMID: 32325539 DOI: 10.1002/cbf.3520] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 02/08/2020] [Accepted: 02/12/2020] [Indexed: 11/12/2022]
Abstract
Growing data have recognized the significance of Response Gene to Complement (RGC)-32 in numerous tumour developments. Notwithstanding, the functional role and underlying mechanism of it in tongue squamous cell carcinoma (TSCC) remain enigmatic. Here, to identify the impact of RGC-32 in TSCC, its expression in multiple TSCC cells was measured and loss-of-function experiments in cell lines were performed to illuminate the function of it induced TSCC progression, via si-RNA knockdown, CCK-8, colony formation, wound-healing, transwell, flow cytometry and western blot assays. To clarify potential mechanism, expressions of hallmarks in epithelial-mesenchymal transition (EMT) process and PI3K/AKT signalling were assessed, and the upstream miR regulator of RGC-32 was predicted and verified by applying bioinformatic approaches and dual-luciferase reporter assay, respectively. Finally, the rescue experiments were applied to better elucidate the effect of miR-26b/RGC-32 axis in TSCC behaviours. As a result, RGC-32 was upregulated in TSCC cells and knocking down of it abrogated cell proliferation, trans-migration and invasion, whilst promoted apoptosis in TSCC, which was regulated through repressing EMT and inactivation of PI3K/AKT signalling. Subsequently, miR-26b was predicted and identified as an upstream regulator of RGC-32, and the pro-tumorigenic effect of RGC-32 was reversed by miR-26b overexpression. Collectively, our results demonstrated that RGC-32 facilitated TSCC progression, which was modulated by activations of PI3K/AKT pathway and EMT process, and reduction of its negative regulator of miR-26b. These findings highlight a novel role of miR-26b/RGC-32 axis in TSCC and underlying mechanism, encouraging a potent usage in TSCC treatment. SIGNIFICANCE OF THE STUDY: We first uncovered that Response Gene to Complement-32 played a significantly pro-tumorigenic role in tongue squamous cell carcinoma (TSCC), which was closely regulated by downregulation of miR-26b and activations of epithelial-mesenchymal transition process and PI3K/AKT signalling. These findings contribute to better understand the molecular mechanism in carcinogenesis of TSCC, and shed some light on promising strategy for TSCC therapeutics.
Collapse
Affiliation(s)
- Zhong-Heng Yang
- Department of Stomatology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Juan Li
- Department of Pathology, The Fourth Hospital of Jinan, Jinan, Shandong, China
| | - Wei-Zhi Chen
- Department of Radiology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Fan-Shuang Kong
- Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
26
|
Zhao W, Wang S, Qin T, Wang W. Circular RNA (circ-0075804) promotes the proliferation of retinoblastoma via combining heterogeneous nuclear ribonucleoprotein K (HNRNPK) to improve the stability of E2F transcription factor 3 E2F3. J Cell Biochem 2020; 121:3516-3525. [PMID: 32065448 DOI: 10.1002/jcb.29631] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 12/19/2019] [Indexed: 12/14/2022]
Abstract
It is growingly recognized that messenger RNAs (mRNAs) are important regulators of various cancers. However, there are few reporters about the function of E2F3 in retinoblastoma (RB), which needs more exploration. In addition, the circRNA circ-0075804 was derived from the E2F3 host gene. The purpose of the study is to figure out the role and molecular regulation mechanism of E2F3 and circ-0075804 in RB. The role of E2F3 in RB was determined through E2F3 silencing and loss of expression was evaluated by real-time quantitative polymerase chain reaction (RT-qPCR), Western blot, CCK-8, colony formation, and 5-ethynyl-2'-deoxyuridine assays. The interactions between E2F3 and circ-0075804 were validated through loss and gain function of circ-0075804. Besides, the role of circ-0075804 in RB was determined by several functional assays. And the binding ability between heterogeneous nuclear ribonucleoprotein K and circ-0075804 was verified by RNA pull-down, Western blot, and RT-qPCR assays. The expression of E2F3 was upregulated in RB cell lines. Furthermore, knockdown of E2F3 inhibited cell proliferation and induced cell apoptosis in RB. And circ-0075804 positively regulated the expression of E2F3. Moreover, circ-0075804 facilitated cell proliferation and suppressed cell apoptosis. Besides, HNRNPK could bind with circ-0075804 in RB. Finally, knockdown of E2F3 partly rescued the promoting role of circ-0075804 overexpression in RB. Overall, circ-0075804 promotes the proliferation of RB via combining HNRNPK to improve the stability of E2F3, which brings new light for treating RB.
Collapse
Affiliation(s)
- Wenbo Zhao
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuai Wang
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tingyu Qin
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenzhan Wang
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
27
|
Wang M, Liao Q, Zou P. PRKCZ-AS1 promotes the tumorigenesis of lung adenocarcinoma via sponging miR-766-5p to modulate MAPK1. Cancer Biol Ther 2020; 21:364-371. [PMID: 31939714 DOI: 10.1080/15384047.2019.1702402] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Lung adenocarcinoma (LUAD) is the most prevalent histological subclass of non-small cell lung cancer. Long non-coding RNAs (lncRNAs) have been recognized as the crucial regulatory factors in tumor development and progression. Nevertheless, limited research has been carried on the function of PRKCZ-AS1 in LUAD. In this study, the expression of PRKCZ-AS1 in LUAD tissues and cell lines was notably upregulated. Moreover, knockdown of PRKCZ-AS1 inhibited the proliferation and migration, but promoted apoptosis in LUAD cells. Furthermore, miR-766-5p could bind with PRKCZ-AS1. Besides, the expression miR-766-5p was negatively regulated by PRKCZ-AS1 expression in LUAD cells. Furtherly, PRKCZ-AS1 expression positively regulated the expression of MAPK1. Similarly, the expression of MAPK1 was negatively regulated by miR-766-5p expression. Moreover, the binding ability between miR-766-5p and MAPK1 was confirmed. Furthermore, knockdown of MAPK1 partly rescued the miR-766-5p inhibition-mediated promoting effect on proliferation and migration in LUAD cells transfected with PRKCZ-AS1#1. Overall, above results suggested that PRKCZ-AS1 promotes the occurrence of LUAD by sponging miR-766-5p to upregulate MAPK1 expression, which may provide new insights into LUAD treatment.
Collapse
Affiliation(s)
- Ming Wang
- Department of Thoracic Surgery, Shulan (Hangzhou) Hospital, Hangzhou, Zhejiang Province, China
| | - Qin Liao
- Department of Oncology, Shulan (Hangzhou) Hospital, Hangzhou, Zhejiang Province, China
| | - Pengfei Zou
- Department of Infectious Diseases, Shulan (Hangzhou) Hospital, Hangzhou, Zhejiang Province, China
| |
Collapse
|
28
|
Zhao X, Zhong Q, Cheng X, Wang S, Wu R, Leng X, Shao L. miR-449c-5p availability is antagonized by circ-NOTCH1 for MYC-induced NOTCH1 upregulation as well as tumor metastasis and stemness in gastric cancer. J Cell Biochem 2020; 121:4052-4063. [PMID: 31943342 DOI: 10.1002/jcb.29575] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 12/09/2019] [Indexed: 02/06/2023]
Abstract
Gastric cancer (GC), identified as the most common gastrointestinal malignancy, is one of the primary causes of cancer-related mortality in the world. Although surgery and chemotherapy for GC treatment have been improved, the 5-year overall survival rate is still unsatisfactory. Circ-NOTCH1 is a novel circular RNA derived from its host gene NOTCH1, and has not been studied in any cancers. Here we explored the potential role and mediatory mechanism of circ-NOTCH1 in GC. In this study, circ-NOTCH1 exhibited increased expression in GC tissues and cells. Suppression of circ-NOTCH1 inhibited cell migration, invasion, tumor spheroids number, and side population ratio. Circ-NOTCH1 also promoted GC growth and metastasis in vivo. Additionally, it was found that circ-NOTCH1 could bind to miR-449c-5p. Circ-NOTCH1 promoted metastasis and stemness in GC through sponging miR-449c-5p. Subsequently, MYC was identified as a downstream gene of miR-449c-5p. MYC could bind to the promoter of NOTCH1 to regulate GC progression. Furthermore, rescue assays demonstrated that NOTCH1 knockdown reversed the effects of overexpression of MYC in metastasis and stemness in AGS cells/sh-circNOTCH1. Above findings explained that circ-NOTCH1 promoted metastasis and stemness in GC by targeting miR-449c-5p/MYC/NOTCH1 axis, suggesting the possibility of circ-NOTCH1 as a therapeutic marker for GC.
Collapse
Affiliation(s)
- Xiaoyong Zhao
- The Fourth General Surgery Department, Jiaozuo People's Hospital, Jiaozuo, Henan, China
| | - Qinggui Zhong
- Urological Surgery Department, Jiaozuo People's Hospital, Jiaozuo, Henan, China
| | - Xueyan Cheng
- Surgical Outpatient Clinic, Jiaozuo People's Hospital, Jiaozuo, Henan, China
| | - Shang Wang
- The Fourth General Surgery Department, Jiaozuo People's Hospital, Jiaozuo, Henan, China
| | - Ruiqing Wu
- The Fourth General Surgery Department, Jiaozuo People's Hospital, Jiaozuo, Henan, China
| | - Xinling Leng
- The Fourth General Surgery Department, Jiaozuo People's Hospital, Jiaozuo, Henan, China
| | - Lei Shao
- The Fourth General Surgery Department, Jiaozuo People's Hospital, Jiaozuo, Henan, China
| |
Collapse
|
29
|
Hu Z, Li L, Cheng P, Liu Q, Zheng X, Peng F, Zhang Q. lncRNA MSC-AS1 activates Wnt/β-catenin signaling pathway to modulate cell proliferation and migration in kidney renal clear cell carcinoma via miR-3924/WNT5A. J Cell Biochem 2020; 121:4085-4093. [PMID: 31916281 DOI: 10.1002/jcb.29594] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 12/09/2019] [Indexed: 12/12/2022]
Abstract
Kidney renal clear cell carcinoma (KIRC) is the most general subtype of renal cell carcinoma, which composes about 1/20 of adult malignancies. The anomaly of long noncoding RNAs (lncRNAs) expression is proved to mediate cancer progression of various types. The function and mediation mechanism of MSC-AS1 has rarely been detected in KIRC before. This study started with the mediation of MSC-AS1 on cell function. In this study, MSC-AS1 was dramatically upregulated in KIRC and correlated with dismal prognosis of KIRC patients. Knockdown of MSC-AS1 would suppress the proliferative and migratory properties of KIRC cells. MSC-AS1 was found to directly downregulate miR-3924 expression while miR-3924 directly downregulated WNT5A expression. Meanwhile, MSC-AS1 could promote the expression of WNT5A, indicating the existence of MSC-AS1/miR-3924/WNT5A. Further assays indicated that MSC-AS1 could enhance Wnt/β-catenin pathway. By means of rescue assays, the mediation of MSC-AS1/miR-3924/WNT5A/β-catenin axis on KIRC cell proliferation, migration and migration was verified. This study revealed that MSC-AS1 regulates KIRC cell proliferation and migration via miR-3924/WNT5A/β-catenin axis. MSC-AS1 might contribute to new strategies for KIRC treatment.
Collapse
Affiliation(s)
- Zhaoxiong Hu
- Department of Nephrology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Linhong Li
- Department of Geriatric Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Peng Cheng
- Department of Nephrology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Qin Liu
- Department of Neurology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xuan Zheng
- Department of Nephrology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Feng Peng
- Department of Oncology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Qinghong Zhang
- Department of Nephrology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| |
Collapse
|
30
|
Hu Y, Wang L, Li Z, Wan Z, Shao M, Wu S, Wang G. Potential Prognostic and Diagnostic Values of CDC6, CDC45, ORC6 and SNHG7 in Colorectal Cancer. Onco Targets Ther 2019; 12:11609-11621. [PMID: 32021241 PMCID: PMC6942537 DOI: 10.2147/ott.s231941] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 11/18/2019] [Indexed: 01/20/2023] Open
Abstract
Background Colorectal cancer (CRC) is a common human malignancy. The aims of this study are to investigate the gene expression profile of CRC and to explore potential strategy for CRC diagnosis, therapy and prognosis. Methods We use affy and Limma package of Bioconductor R to do differential expression genes (DEGs) and differential expression lncRNAs (DELs) analysis from the gene datasets (GSE8671, GSE21510, GSE32323, GSE39582 and TCGA) respectively. Then, DEGs were analyzed by GO and KEGG pathway and Kaplan-Meier survival curve and Cox regression analyses were used to find aberrantly expressed genes associated with survival outcome of CRC patients. Real-time PCR assay was used to verify the aberrantly expressed genes expression in CRC samples. Results 306 up-regulation and 213 down-regulation common DEGs were found. A total of 485 DELs were identified, of which 241 up-regulated and 244 down-regulated. Then, GO and KEGG pathway analyses showed that DEGs were involved in cell cycle, mineral absorption, DNA replication, and Nitrogen metabolism. Among them, Kaplan-Meier survival curve and Cox regression analyses revealed that CDC6, CDC45, ORC6 and SNHG7 levels were significantly associated with survival outcome of CRC patients. Finally, real-time PCR assay was used to verify that the CDC6, CDC45, ORC6 and SNHG7 expression were up-regulated in 198 CRC samples compared with the expression levels in individual-matched adjacent mucosa samples. Conclusion CDC6, CDC45, ORC6 and SNHG7 are implicated in CRC initiation and progression and could be explored as potential diagnosis, therapy and prognosis targets for CRC.
Collapse
Affiliation(s)
- Yang Hu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, People's Republic of China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, People's Republic of China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, People's Republic of China
| | - Liping Wang
- Department of Clinical Oncology, The First People's Hospital of Chenzhou, Chenzhou 432000, Hunan, People's Republic of China
| | - Zhixing Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, People's Republic of China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, People's Republic of China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, People's Republic of China
| | - Zirui Wan
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, People's Republic of China
| | - Mingjie Shao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, People's Republic of China.,Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha 410008, People's Republic of China
| | - Shaobin Wu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, People's Republic of China.,Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha 410008, People's Republic of China
| | - Guo Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, People's Republic of China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, People's Republic of China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, People's Republic of China
| |
Collapse
|