1
|
Li Z, Chen X, Li Y, Xu Y, Zhou Y. Aberrant DNA Methylation in Esophageal Squamous Cell Carcinoma and its Clinical Implications in Systemic Chemotherapy. Int J Med Sci 2025; 22:1002-1014. [PMID: 39991775 PMCID: PMC11843145 DOI: 10.7150/ijms.109161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 01/25/2025] [Indexed: 02/25/2025] Open
Abstract
Esophageal cancer is a significant global health concern, with esophageal squamous cell carcinoma being the predominant subtype in high-incidence regions like China. Despite advances in multidisciplinary treatments, the prognosis for ESCC remains poor, with systemic chemotherapy facing the challenge of drug resistance. Epigenetic alterations, particularly DNA methylation, play a crucial role in ESCC carcinogenesis and therapeutic response. Aberrant DNA methylations, including global hypomethylation and promoter-specific hyper-methylation, disrupt critical pathways such as cell cycle regulation, apoptosis, and DNA repair, contributing to chemoresistance. Several studies have identified methylation markers that predict treatment response, particularly for chemotherapy, targeted therapy and immunotherapy, such as p16 and GPX3 for cisplatin, MTHFR for 5-FU, CHFR for paclitaxel. DNA methyltransferase inhibitors and other epigenetic therapies are being explored to reverse these methylation changes and enhance therapeutic efficacy. However, the clinical utility of these markers remains limited due to the lack of large-scale validation and concerns over off-target effects. This review aims to summarize all aberrant methylation alterations in ESCC and the clinical implications of aberrantly methylated candidate genes identified in ESCC systemic chemotherapy, with the goal of further understanding the underlying molecular mechanisms, refining methylation-targeting therapies, and integrating them with conventional treatments to improve patient outcomes.
Collapse
Affiliation(s)
- Zhao Li
- Department of Health Prevention and Care, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 Dahua Road, Beijing 100730, China
| | - Xi Chen
- Department of Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 Dahua Road, Beijing, 100730, China
| | - Yongkang Li
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, No. 5 Dong Dan San Tiao, Beijing, 100005, China
| | - Yan Xu
- Department of Health Prevention and Care, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 Dahua Road, Beijing 100730, China
| | - Yang Zhou
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Panjiayuan Nanli No.17, Beijing 100021, China
| |
Collapse
|
2
|
Wang H, Jin H, Zhao S. Expression and clinical significance of RHCG in endometrial cancer. Histol Histopathol 2024; 39:611-621. [PMID: 37732703 DOI: 10.14670/hh-18-663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Endometrial cancer (EC) is the most common gynecological cancer. Rhesus family, C glycoprotein (RHCG) has been evidenced to be involved in the occurrence and development of various tumors. This study aimed to investigate the expression and clinical significance of RHCG in EC. Bioinformatics analysis was based on the RNAseq counts data from TCGA database, and the prognosis analysis was performed using the Kaplan-Meier method; 4 cases of endometrioid adenocarcinomas samples and 4 cases of normal proliferative endometrium were collected for qPCR and western blot; immunohistochemistry analysis was employed to assess the expression of RHCG in a tissue microarray; the correlation between RHCG and clinicopathological factors was analyzed through Mann-Whitney U test. The lentiviral interference vector was further constructed. The results demonstrated that RHCG was highly expressed in EC tissues, and RHCG was an independent factor affecting the overall survival of patients. Additionally, the expression of RHCG was related to FIGO stage and tumor infiltrate. After interfering with shRHCG, the proliferation activity of EC cells decreased, the migration ability of cells decreased, the apoptosis of cells increased, and the tumor outgrowth was arrested. In summary, RHCG promotes the malignant proliferation and migration of EC, and makes the cells have anti-apoptotic activity. Our study provides a theoretical basis for RHCG to become a potential therapeutic target for EC in the future.
Collapse
Affiliation(s)
- Huifang Wang
- Department of Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, PR China
- Department of Gynecology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei Province, PR China
| | - Haihong Jin
- Department of Gynecology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei Province, PR China
| | - Sufen Zhao
- Department of Gynecology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, PR China.
| |
Collapse
|
3
|
Liu W, Wang Y, Zhang Y, Zhou M, Gu H, Lu M, Xia Y. Rh family C glycoprotein contributes to psoriatic inflammation through regulating the dysdifferentiation and cytokine secretion of keratinocytes. J Dermatol Sci 2024; 114:2-12. [PMID: 38514279 DOI: 10.1016/j.jdermsci.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 02/16/2024] [Accepted: 02/27/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND Keratinocyte dysdifferentiation and proinflammatory cytokine production play a central role in psoriatic inflammation. According to recent studies, the Rh family C glycoprotein (RHCG) enhances cell proliferation and disrupts cell differentiation. However, the specific role of RHCG psoriasis development remains unclear. OBJECTIVE We here explored the effect of RHCG on keratinocytes under psoriatic inflammation. METHODS The cell counting kit‑8 assay was conducted to assess proliferation. RHCG protein expression was assessed through western blotting and enzyme-linked immunosorbent assays. The expression of proinflammatory cytokines and differentiation markers was analyzed through a quantitative reverse-transcription polymerase chain reaction. RESULTS Both RHCG mRNA and protein levels increased in psoriatic skin. Notably, cultured keratinocytes treated with an M5 cocktail, which mimics psoriatic inflammation, exhibited higher RHCG expression. Furthermore, RHCG overexpression promoted keratinocyte proliferation, accompanied by an increase in the production of interleukin (IL)-1β, IL-6, and IL-8, and tumor necrosis factor-α. RHCG overexpression also resulted in higher expression of keratin 17, a differentiation marker. Conversely, RHCG gene knockdown reduced keratinocyte proliferation and cytokine secretion. RHCG inhibition in cells recovered both keratin 1 and loricrin expression. Additionally, RHCG overexpression facilitated the phosphorylation of nuclear factor-kappa B and extracellular signal-regulated protein kinase signaling pathways. Importantly, when these signaling pathways were inhibited, the effect of RHCG on keratinocytes was attenuated. CONCLUSION These findings support the substantial role of RHCG in psoriatic inflammation development and suggest that RHCG serves as a potential target for psoriasis treatment.
Collapse
Affiliation(s)
- Wei Liu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yaqi Wang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yitian Zhang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mingzhu Zhou
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hanjiang Gu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mei Lu
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
4
|
Yue Z, Wang D, Li X. A promising anoikis-related prognostic signature predicts prognosis of skin cutaneous melanoma. J Cancer Res Clin Oncol 2023; 149:17757-17770. [PMID: 37930439 DOI: 10.1007/s00432-023-05468-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND Skin cutaneous melanoma (SKCM) is a highly aggressive disease with a poor prognosis for advanced tumors. Anoikis is a caspase-dependent cell death process triggered by extracellular matrix (ECM) detachment, rectifies detachment-induced metabolic defects that compromise cell survival, recent study revealed the crucial role of anoikis for cancer cells to survive during metastasis. However, limited research focused on the role of anoikis in SKCM. METHODS Our study utilized the 27 anoikis-related genes (ARGs) to divide SKCM patients into two clusters, and obtain differentially expressed genes (DEGs) for each cluster. These DEGs were used in stepwise Cox regression analysis to develop a prediction model for SKCM patients consisting of nine ARGs, called the anoikis-related signature (ARS). Subsequently, we used the risk scores calculated from the ARS to divide SKCM patients into two groups and explored differences in immune microenvironment, immune checkpoint reactivity, and drug sensitivity between the groups. RESULTS Nine ARGs were identified to stratify SKCM patients into two risk groups, patients in the high-risk group had a poor prognosis and suppressed immune cell infiltration. Moreover, higher expression of immune checkpoint molecules and a greater sensitivity to immunotherapy and chemotherapy drugs were observed in the low-risk group. Finally, all of the ARS hub genes were found to be upregulated in SKCM tissues and cell lines. CONCLUSION A novel ARGs signature was identified for predicting the prognosis of SKCM. Based on the immune landscape associated with ARS discovered in our study, targeting ARS hub genes may be a promising treatment for SKCM.
Collapse
Affiliation(s)
- Zhanghui Yue
- Department of Dermatology, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Yuelu District, Changsha, 410000, Hunan Province, People's Republic of China
| | - Dan Wang
- Department of Dermatology, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Yuelu District, Changsha, 410000, Hunan Province, People's Republic of China.
| | - Xuemei Li
- Department of Dermatology, The Third Xiangya Hospital, Central South University, No. 138 Tongzipo Road, Yuelu District, Changsha, 410000, Hunan Province, People's Republic of China
| |
Collapse
|
5
|
Li X, Dong H, Zheng Y, Ding S, Li Y, Li H, Huang H, Zhong C, Xie T, Xu Y. AKAP12 inhibits esophageal squamous carcinoma cell proliferation, migration, and cell cycle via the PI3K/AKT signaling pathway. Mol Cell Probes 2023; 72:101939. [PMID: 37879503 DOI: 10.1016/j.mcp.2023.101939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 10/12/2023] [Accepted: 10/17/2023] [Indexed: 10/27/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) consistently ranks as one of the most challenging variants of squamous cell carcinomas, primarily due to the lack of effective early detection strategies. We herein aimed to elucidate the underlying mechanisms and biological role associated with A-kinase anchoring protein 12 (AKAP12) in the context of ESCC. Bioinformatic analysis had revealed significantly lower expression level of AKAP12 in ESCC tissue samples than in their non-cancerous counterparts. To gain deeper insights into the potential role of AKAP12 in the progression of ESCC, we conducted a single-gene set enrichment analysis of AKAP12 on ESCC datasets. Our findings suggested that AKAP12 exhibits functions inhibiting cell cycle progression, tumor proliferation, and epithelial-mesenchymal transition. To further validate our findings, we subjected ESCC cell lines to AKAP12 overexpression using CRISPR/Cas9-SAM. In vitro analyses demonstrated that increased expression of AKAP12 significantly reduced cell proliferation, migration, and cell cycle progression. Simultaneously, genes associated with this biological role undergo corresponding regulatory shifts. These observations provided valuable insights into the biological role played by AKAP12 in ESCC progression. In summary, AKAP12 shows promise as a new potential biomarker for early ESCC diagnosis, offering potential advantages for subsequent therapeutic intervention and disease management.
Collapse
Affiliation(s)
- Xingyi Li
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Nantong University, The First People's Hospital of Nantong, 226001, Nantong, China; Department of Thoracic and Cardiovascular Surgery, First Affiliated Hospital of Huzhou Teachers College, The First Hospital of Huzhou, 313000, Huzhou, China
| | - Hao Dong
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Nantong University, The First People's Hospital of Nantong, 226001, Nantong, China
| | - Yifan Zheng
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Nantong University, The First People's Hospital of Nantong, 226001, Nantong, China
| | - Shengguang Ding
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Nantong University, The First People's Hospital of Nantong, 226001, Nantong, China
| | - Yan Li
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Nantong University, The First People's Hospital of Nantong, 226001, Nantong, China
| | - Hefei Li
- Department of Thoracic and Cardiovascular Surgery, The Third Affiliated Hospital of Nantong University, The Third People's Hospital of Nantong, 226001, Nantong, China
| | - HaiTao Huang
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Nantong University, The First People's Hospital of Nantong, 226001, Nantong, China
| | - Congjun Zhong
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Nantong University, The First People's Hospital of Nantong, 226001, Nantong, China
| | - Tian Xie
- Department of Cardiothoracic Surgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210002, China.
| | - Yiming Xu
- Department of Thoracic and Cardiovascular Surgery, The Second Affiliated Hospital of Nantong University, The First People's Hospital of Nantong, 226001, Nantong, China.
| |
Collapse
|
6
|
Luo D, Wang X, Feng W. Comprehensive analysis of cuproptosis and copper homeostasis genotyping and related immune land scape in lung adenocarcinoma. Sci Rep 2023; 13:16554. [PMID: 37783723 PMCID: PMC10545825 DOI: 10.1038/s41598-023-43795-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/28/2023] [Indexed: 10/04/2023] Open
Abstract
Cuproptosis is a manner of cell death which is related to the homeostasis of copper ions in the cellular environment and is expected to open a new direction of anti-tumor therapy. However, the studies on cuproptosis and copper homeostasis in lung adenocarcinoma (LUAD) are still limited. In this study, we identified new cuproptosis and copper homeostasis related genes (CHRGs) which were effective in stratifying genotyping clusters with survival differences based on transcriptomic data obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). Weighted Gene Co-expression Network Analysis (WGCNA) further expands the screening boundary of CHRGs, and finally we established a 10-CHRGs-based prognostic signature using lasso-penalized cox regression method, which were validated in GSE30219. Comprehensive bioinformatics analysis revealed these genes are potential regulators of modulating immunotherapy efficacy, drug resistance, tumor microenvironment infiltration, and tumor mutation patterns. Lastly, the scRNA-seq datasets GSE183219 and GSE203360 offers the evidences that CHRGs signature are mainly distributed in cancer epithelial cells, real time quantitative polymerase chain reaction (RT-qPCR) also confirmed the differential expression of these genes between normal lung cell line and lung adenocarcinoma cell lines. Collectively, our findings revealed new cuproptosis and copper homeostasis related genotyping clusters and genes which may play important roles in predicting prognosis, influencing tumor microenvironment and drug efficacy in LUAD patients.
Collapse
Affiliation(s)
- Dayuan Luo
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Xiang Wang
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Wei Feng
- Department of Cardiothoracic Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
7
|
Sun G, Ye H, Yang Q, Zhu J, Qiu C, Shi J, Dai L, Wang K, Zhang J, Wang P. Using Proteome Microarray and Gene Expression Omnibus Database to Screen Tumour-Associated Antigens to Construct the Optimal Diagnostic Model of Oesophageal Squamous Cell Carcinoma. Clin Oncol (R Coll Radiol) 2023; 35:e582-e592. [PMID: 37433700 DOI: 10.1016/j.clon.2023.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 05/09/2023] [Accepted: 06/30/2023] [Indexed: 07/13/2023]
Abstract
AIMS Autoantibodies against tumour-associated antigens (TAAs) are promising biomarkers for early immunodiagnosis of cancers. This study was designed to screen and verify autoantibodies against TAAs in sera as diagnostic biomarkers for oesophageal squamous cell carcinoma (ESCC). MATERIALS AND METHODS The customised proteome microarray based on cancer driver genes and the Gene Expression Omnibus database were used to identify potential TAAs. The expression levels of the corresponding autoantibodies in serum samples obtained from 243 ESCC patients and 243 healthy controls were investigated by enzyme-linked immunosorbent assay (ELISA). In total, 486 serum samples were randomly divided into the training set and the validation set in the ratio of 2:1. Logistic regression analysis, recursive partition analysis and support vector machine were performed to establish different diagnostic models. RESULTS Five and nine candidate TAAs were screened out by proteome microarray and bioinformatics analysis, respectively. Among these 14 anti-TAAs autoantibodies, the expression level of nine (p53, PTEN, GNA11, SRSF2, CXCL8, MMP1, MSH6, LAMC2 and SLC2A1) anti-TAAs autoantibodies in the cancer patient group was higher than that in the healthy control group based on the results from ELISA. In the three constructed models, a logistic regression model including four anti-TAA autoantibodies (p53, SLC2A1, GNA11 and MMP1) was considered to be the optimal diagnosis model. The sensitivity and specificity of the model in the training set and the validation set were 70.4%, 72.8% and 67.9%, 67.9%, respectively. The area under the receiver operating characteristic curve for detecting early patients in the training set and the validation set were 0.84 and 0.85, respectively. CONCLUSIONS This approach to screen novel TAAs is feasible, and the model including four autoantibodies could pave the way for the diagnosis of ESCC.
Collapse
Affiliation(s)
- G Sun
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan Province, China; Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, China
| | - H Ye
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan Province, China; Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Q Yang
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan Province, China; Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, China
| | - J Zhu
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan Province, China; Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, China
| | - C Qiu
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, China
| | - J Shi
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, China; Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - L Dai
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, China; Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - K Wang
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, China; Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - J Zhang
- Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, China; Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - P Wang
- Department of Epidemiology and Statistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan Province, China; Henan Key Laboratory of Tumor Epidemiology and State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan Province, China.
| |
Collapse
|
8
|
Zhao H, Wei Y, Zhang J, Zhang K, Tian L, Liu Y, Zhang S, Zhou Y, Wang Z, Shi S, Fu Z, Fu J, Zhao J, Li X, Zhang L, Zhao L, Liu K. HPV16 infection promotes the malignant transformation of the esophagus and progression of esophageal squamous cell carcinoma. J Med Virol 2023; 95:e29132. [PMID: 37792307 DOI: 10.1002/jmv.29132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/27/2023] [Accepted: 09/05/2023] [Indexed: 10/05/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) may be correlated with HPV infection, and the mechanism underlying the ESCC formation induced by HPV16 infection remains elusive. Here, we overexpressed HPV16 E6 and E7 and coordinated the overexpression of these two genes in EPC2 and ESCC cells. We found that E7 and coordinated expression of E6 and E7 promoted the proliferation of EPC2 cells, and upregulation of shh was responsible for cell proliferation since the use of vismodegib led to the failure of organoid formation. Meanwhile, overexpression of E6 and E7 in ESCC cells promoted cell proliferation, migration, and invasion in vitro. Importantly, E6 and E7 coordinately increased the capability of tumor growth in nude mice, while vismodegib slowed the growth of tumors in NCG mice. Moreover, a series of genes and proteins changed in cell lines after overexpression of the E6 and E7 genes, the potential biological processes and pathways were systematically analyzed using a bioinformatics assay. Together, these findings suggest that the activation of the hedgehog pathway induced by HPV16 infection may initially transform basal cells in the esophagus and promote following malignant processes in ESCC cells. The application of hedgehog inhibitors may represent a therapeutic avenue for ESCC treatment.
Collapse
Affiliation(s)
- Hongzhou Zhao
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
- School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Yuxuan Wei
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
- School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Jiaying Zhang
- School of Life Science, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Kun Zhang
- Department of General Surgery, The First Hospital of Fuzhou, Fuzhou, Fujian, People's Republic of China
| | - Liming Tian
- Department of Gynecology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Yongpan Liu
- School of Life Science, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Shihui Zhang
- Centre for Translational Stem Cell Biology, School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, People's Republic of China
| | - Yijian Zhou
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
- School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Zhuo Wang
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
- School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Songlin Shi
- School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Zhichao Fu
- Department of Radiotherapy, 900 Hospital of the Joint Logistics Team (Dongfang Hospital, Xiamen University), Fuzhou, Fujian, People's Republic of China
| | - Jianqian Fu
- Department of Medical Oncology, The Fifth Hospital of Xiamen, Xiamen, Fujian, People's Republic of China
| | - Jing Zhao
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
- School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Xinxin Li
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
- School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Lijia Zhang
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Liran Zhao
- School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
| | - Kuancan Liu
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
- School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China
- School of Life Science, Nanchang Normal University, Nanchang, Jiangxi, People's Republic of China
| |
Collapse
|
9
|
Yu N, Zhang Y, Gao R. A Tensor Method Based on Enhanced Tensor Nuclear Norm and Hypergraph Laplacian Regularization for Pan-Cancer Omics Data Analysis. IEEE J Biomed Health Inform 2023; 27:1225-1236. [PMID: 37015665 DOI: 10.1109/jbhi.2022.3231908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
As a powerful data representation technique, tensor robust principal component analysis (TRPCA) has been widely used for clustering and feature selection tasks. However, it ignores the significant difference in singular values of tensor data and the manifold information contained in different views, thereby causing serious degradation of conventional TRPCA performance. In this paper, a novel tensor method based on enhanced tensor nuclear norm and hypergraph Laplacian regularization (ETHLR) is developed to address the above problem. ETHLR can jointly learn the prior knowledge of singular values and high-order manifold structures in the unified tensor space and the view-specific feature spaces, respectively. Specifically, the enhanced tensor nuclear norm, namely, the weighted tensor Schatten p-norm, is used to shrink the singular values by fully considering the salient difference information of singular values and the complementary information embedded in the tensor space; the hypergraph Laplacian constraint helps encode high-order geometric structures among multiple samples in the nonlinear view-specific feature space. Furthermore, we employ inexact augmented Lagrange multipliers (ALM) to optimize the ETHLR method. Numerous experiments on pan-cancer omics data show that the superiority of ETHLR over several state-of-the-art competitors.
Collapse
|
10
|
Wang D, Dai J, Suo C, Wang S, Zhang Y, Chen X. Molecular subtyping of esophageal squamous cell carcinoma by large-scale transcriptional profiling: Characterization, therapeutic targets, and prognostic value. Front Genet 2022; 13:1033214. [DOI: 10.3389/fgene.2022.1033214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/28/2022] [Indexed: 11/11/2022] Open
Abstract
The tumor heterogeneity of the transcriptional profiles is independent of genetic variation. Several studies have successfully identified esophageal squamous cell carcinoma (ESCC) subtypes based on the somatic mutation profile and copy number variations on the genome. However, transcriptome-based classification is limited. In this study, we classified 141 patients with ESCC into three subtypes (Subtype 1, Subtype 2, and Subtype 3) via tumor sample gene expression profiling. Differential gene expression (DGE) analysis of paired tumor and normal samples for each subtype revealed significant difference among subtypes. Moreover, the degree of change in the expression levels of most genes gradually increased from Subtype 1 to Subtype 3. Gene set enrichment analysis (GSEA) identified the representative pathways in each subtype: Subtype 1, abnormal Wnt signaling pathway activation; Subtype 2, inhibition of glycogen metabolism; and Subtype 3, downregulation of neutrophil degranulation process. Weighted gene co-expression network analysis (WGCNA) was used to elucidate the finer regulation of biological pathways and discover hub genes. Subsequently, nine hub genes (CORO1A, CD180, SASH3, CD52, CD300A, CD14, DUSP1, KIF14, and MCM2) were validated to be associated with survival in ESCC based on the RNA sequencing (RNA-seq) data from The Cancer Genome Atlas (TCGA) database. The clustering analysis of ESCC granted better understanding of the molecular characteristics of ESCC and led to the discover of new potential therapeutic targets that may contribute to the clinical treatment of ESCC.
Collapse
|
11
|
Wang Y, Song T, Li K, Liu H, Han Y, Xu T, Cao F, Li Y, Yu Y. Heparanase is a prognostic biomarker independent of tumor purity and hypoxia based on bioinformatics and immunohistochemistry analysis of esophageal squamous cell carcinoma. World J Surg Oncol 2022; 20:236. [PMID: 35840985 PMCID: PMC9288057 DOI: 10.1186/s12957-022-02698-9] [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: 01/01/2022] [Accepted: 07/02/2022] [Indexed: 11/10/2022] Open
Abstract
Background Esophageal squamous cell carcinoma (ESCC) is a common malignant tumor of the digestive tract with a poor prognosis. The tumor microenvironment (TME) is mainly composed of tumor cells, stromal cells, and immune cells and plays an important role in ESCC development. There are substantial differences in tumor purity among different parts of ESCC tissues, consisting of distinct immune and stromal cells and variations in the status of hypoxia. Thus, prognostic models of ESCC based on bioinformatic analysis of tumor tissues are unreliable. Method Differentially expressed genes (DEGs) independent of tumor purity and hypoxia were screened by Spearman correlation analysis of public ESCC cohorts. Subsequently, the DEGs were subjected to Cox regression analysis. Then, we constructed a protein–protein interaction (PPI) network of the DEGs using Cytoscape. Intersection analysis of the univariate Cox and PPI results indicated that heparanase (HPSE), an endo-β-D-glucuronidase capable of cleaving heparan sulfate side chains, was a predictive factor. Gene set enrichment analysis (GSEA) was used to reveal the potential function of HPSE, and single-cell sequencing data were analyzed to evaluate the distribution of HPSE in immune cells. Furthermore, a human ESCC tissue microarray was used to validate the expression and prognostic value of HPSE. Result We found that HPSE was downregulated in ESCC tissues and was not correlated with tumor purity or hypoxia status. HPSE is involved in multiple biological processes. ESCC patients with low HPSE expression in cancerous tissues exhibited poor prognosis. Conclusions These results indicate that low HPSE expression in cancerous tissues correlates with poor prognosis in patients with ESCC. HPSE is a novel prognostic biomarker independent of tumor purity and hypoxia status in ESCC. Supplementary Information The online version contains supplementary material available at 10.1186/s12957-022-02698-9.
Collapse
Affiliation(s)
- Yu Wang
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, People's Republic of China
| | - Tongjun Song
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, People's Republic of China
| | - Kai Li
- Department of Pathology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, People's Republic of China
| | - Hao Liu
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, People's Republic of China
| | - Yan Han
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, People's Republic of China
| | - Tao Xu
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, People's Republic of China
| | - Fengjun Cao
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, People's Republic of China
| | - Yong Li
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, People's Republic of China.
| | - Yuandong Yu
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, People's Republic of China.
| |
Collapse
|
12
|
Wang Z, Wang J, Zhao H, Zhao T, Chen Y, Jiang M, Zhang S, Wei Y, Zhang J, Zhou Y, Shi S, Fu Z, Yang Y, Zhang Y, Yang L, Que J, Liu K. Targeting the SOX2/PARP1 complex to intervene in the growth of esophageal squamous cell carcinoma. Biomed Pharmacother 2022; 153:113309. [PMID: 35738180 DOI: 10.1016/j.biopha.2022.113309] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 11/30/2022] Open
Abstract
Elevated SOX2 protein levels are closely correlated with the increased incidence of esophageal squamous cell carcinoma (ESCC). However, establishing effective target measures for ESCC treatments continue to be researched. It has been previously proposed that SOX2 represents a potential therapeutic target for ESCC. Here, we found that the enzyme Poly(ADP-Ribose) polymerase 1 (PARP1) enriched in ESCCs interact with SOX2. Inhibition of PARP1 with 3-aminobenzamide (3-ABA) or shRNA knockdown reduced the proliferation of ESCCs, accompanied by decreased protein levels of SOX2. RNA sequencing demonstrated that PARP1 inhibition affected multiple signaling pathways involved in cancer cell proliferation. Additionally, 3-ABA synergistically suppressed the growth of ESCC cells when combined with cisplatin, and metformin potentiated the suppressive effect of 3-ABA on ESCC cell growth. Together these findings suggest that targeting SOX2 binding partner PARP1 provides a possible avenue to treat patients with high levels of SOX2.
Collapse
Affiliation(s)
- Zhuo Wang
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Junkai Wang
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Hongzhou Zhao
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Tingting Zhao
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Yunyun Chen
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Ming Jiang
- Department of Gastroenterology of The Children's Hospital, Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310058, China
| | - Shihui Zhang
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Yuxuan Wei
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Jiaying Zhang
- School of Life Science, Xiamen University, Xiamen, Fujian 361102, China
| | - Yijian Zhou
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Songlin Shi
- School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhichao Fu
- Department of radiotherapy, 900 Hospital of the Joint Logistics Team (Dongfang Hospital, Xiamen University), Fuzhou, Fujian 350025, China
| | - Yaxin Yang
- Department of Biology, University of Rochester, NY 14627, USA
| | - Yujun Zhang
- School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Ling Yang
- School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Jianwen Que
- Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA.
| | - Kuancan Liu
- Central Laboratory, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China; School of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
13
|
Zhang K, Fan R, Zhao D, Liu P, Yang Z, Liu J, Zhang S, Rao S, Wang Y, Wan L. CircATIC
inhibits esophageal carcinoma progression and promotes radiosensitivity by elevating
RHCG
through sponging
miR‐10‐3p. Thorac Cancer 2022; 13:934-946. [PMID: 35307984 PMCID: PMC8977172 DOI: 10.1111/1759-7714.14326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 01/05/2022] [Accepted: 01/10/2022] [Indexed: 11/29/2022] Open
Abstract
Background Methods Results Conclusions
Collapse
Affiliation(s)
- Kai Zhang
- Department of Radiation Oncology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan China
| | - Ruitai Fan
- Department of Radiation Oncology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan China
| | - Deyao Zhao
- Department of Radiation Oncology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan China
| | - Peipei Liu
- Department of Respiratory and Critical Care Medicine Nanyang Central Hospital Nanyang Henan China
| | - Zheng Yang
- Department of Radiotherapy Nanyang Central Hospital Nanyang Henan China
| | - Junqi Liu
- Department of Radiation Oncology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan China
| | - Song Zhang
- Department of Radiation Oncology The First Affiliated Hospital of Zhengzhou University Zhengzhou Henan China
| | - Shilei Rao
- Department of Radiotherapy Nanyang Central Hospital Nanyang Henan China
| | - Yang Wang
- Department of Radiotherapy Nanyang Central Hospital Nanyang Henan China
| | - Lixin Wan
- Department of Oncology Nanyang Central Hospital Nanyang Henan China
| |
Collapse
|
14
|
Wang L, Liu G, Bolor-Erdene E, Li Q, Mei Y, Zhou L. Identification of KIF4A as a prognostic biomarker for esophageal squamous cell carcinoma. Aging (Albany NY) 2021; 13:24050-24070. [PMID: 34775374 PMCID: PMC8610135 DOI: 10.18632/aging.203585] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 09/03/2021] [Indexed: 12/24/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is the most common and aggressive tumor worldwide, and the long-term survival of these patients remains poor. Three databases (GSE17351, GSE20347, and GSE100942) were obtained from Gene Expression Omnibus, and 193 differentially expressed genes including 56 upregulated and 137 downregulated genes were identified by paired test using limma R package. Then, functional enrichments by gene ontology and Kyoto Encyclopedia of Genes and Genomes analyses showed these genes were mainly related protein digestion and absorption, and IL-17 signaling pathway. We then constructed a protein-protein interaction network and cytoHubba module to determine the six hub genes and overall survival analysis of the six hub genes were evaluated by UALCAN and GEPIA2 analysis. Ultimately, the experimental results confirmed the KIF4A was overexpressed in the ESCC tissues and cell lines compared with the normal esophageal mucosal tissues and was linked to poor prognosis. Moreover, we also revealed that KIF4A facilitates proliferation, cell cycle, migration, and invasion of ESCC in vivo and in vitro. Overall, these findings demonstrated that KIF4A could serve as diagnostic and prognostic biomarkers and may help facilitate therapeutic targets in ESCC patients.
Collapse
Affiliation(s)
- Lingwei Wang
- Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China.,East Hospital of Tongji University School of Medicine, Shanghai 200120, China
| | - Gang Liu
- East Hospital of Tongji University School of Medicine, Shanghai 200120, China
| | - Enkhbat Bolor-Erdene
- Department of Cardiothoracic Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Qinchuan Li
- East Hospital of Tongji University School of Medicine, Shanghai 200120, China
| | - Yunqing Mei
- Department of Cardiothoracic Surgery, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China.,Department of Cardiothoracic Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Lei Zhou
- Department of Cardiothoracic Surgery, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| |
Collapse
|
15
|
Miao L, Feng G, Yuan H. CircRNAs: a family number of miRNA regulatory transcriptome in laryngeal carcinoma. J Clin Lab Anal 2021; 35:e24038. [PMID: 34617636 PMCID: PMC8605118 DOI: 10.1002/jcla.24038] [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: 08/28/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 01/22/2023] Open
Abstract
Laryngeal carcinoma (LC) is a common head and neck cancer, which is the result of mutational changes due to gene dysregulation and etiological factors such as tobacco and smoking. A large number of patients received a poor prognosis due to diagnosis at an advanced stage. This highlights the need for definitive, early, and efficient diagnoses. With rapid development of high‐throughput sequencing, circular RNA (circRNA) has been reported to play a pivotal role in cancer. CircRNA functions as a microRNA (miRNA) sponge in the regulation of mRNA expression, forming circRNA‐miRNA regulatory axis. In this review, we described the axis in LC. The result indicated that CDR1as, hsa_circ_0042823, hsa_circ_0023028, circPARD3, hsa_circ_103862, hsa_circ_0000218, circMYLK, circCORO1C, hsa_circ_100290, circ‐CCND1, hsa_circ_0057481, circFLAN, and circRASSF2 expressed higher in LC, whereas, hsa_circ_0036722 and hsa_circ_0042666 expressed lower. The circRNAs regulated the target genes by sponging miRNAs and contributed to the pathogenesis of LC.
Collapse
Affiliation(s)
- Limin Miao
- Department of Geriatric Dentistry, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.,Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Guanying Feng
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Oral and Maxillofacial, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Hua Yuan
- Jiangsu Province Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China.,Department of Oral and Maxillofacial, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
16
|
Zhao X, Li H, Lyu S, Zhai J, Ji Z, Zhang Z, Zhang X, Liu Z, Wang H, Xu J, Fan H, Kou J, Li L, Lang R, He Q. Single-cell transcriptomics reveals heterogeneous progression and EGFR activation in pancreatic adenosquamous carcinoma. Int J Biol Sci 2021; 17:2590-2605. [PMID: 34326696 PMCID: PMC8315026 DOI: 10.7150/ijbs.58886] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 06/08/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic adenosquamous carcinoma (PASC) — a rare pathological pancreatic cancer (PC) type — has a poor prognosis due to high malignancy. To examine the heterogeneity of PASC, we performed single-cell RNA sequencing (scRNA-seq) profiling with sample tissues from a healthy donor pancreas, an intraductal papillary mucinous neoplasm, and a patient with PASC. Of 9,887 individual cells, ten cell subpopulations were identified, including myeloid, immune, ductal, fibroblast, acinar, stellate, endothelial, and cancer cells. Cancer cells were divided into five clusters. Notably, cluster 1 exhibited stem-like phenotypes expressing UBE2C, ASPM, and TOP2A. We found that S100A2 is a potential biomarker for cancer cells. LGALS1, NPM1, RACK1, and PERP were upregulated from ductal to cancer cells. Furthermore, the copy number variations in ductal and cancer cells were greater than in the reference cells. The expression of EREG, FCGR2A, CCL4L2, and CTSC increased in myeloid cells from the normal pancreas to PASC. The gene sets expressed by cancer-associated fibroblasts were enriched in the immunosuppressive pathways. We demonstrate that EGFR-associated ligand-receptor pairs are activated in ductal-stromal cell communications. Hence, this study revealed the heterogeneous variations of ductal and stromal cells, defined cancer-associated signaling pathways, and deciphered intercellular interactions following PASC progression.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital affiliated to Capital Medical University, Beijing 100020, China
| | - Han Li
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shaocheng Lyu
- Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital affiliated to Capital Medical University, Beijing 100020, China
| | - Jialei Zhai
- Department of Pathology, Beijing Chaoyang Hospital affiliated to Capital Medical University, Beijing 100020, China
| | - Zhiwei Ji
- College of Artificial Intelligence, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Zhigang Zhang
- School of Information Management and Statistics, Hubei University of Economics, Wuhan 430205, Hubei, China
| | - Xinxue Zhang
- Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital affiliated to Capital Medical University, Beijing 100020, China
| | - Zhe Liu
- Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital affiliated to Capital Medical University, Beijing 100020, China
| | - Huaguang Wang
- Department of Pharmacology, Beijing Chaoyang Hospital affiliated to Capital Medical University, Beijing 100020, China
| | - Junming Xu
- Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital affiliated to Capital Medical University, Beijing 100020, China
| | - Hua Fan
- Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital affiliated to Capital Medical University, Beijing 100020, China
| | - Jiantao Kou
- Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital affiliated to Capital Medical University, Beijing 100020, China
| | - Lixin Li
- Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital affiliated to Capital Medical University, Beijing 100020, China
| | - Ren Lang
- Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital affiliated to Capital Medical University, Beijing 100020, China
| | - Qiang He
- Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital affiliated to Capital Medical University, Beijing 100020, China
| |
Collapse
|
17
|
Cai Z, Wei Y, Chen S, Gong Y, Fu Y, Dai X, Zhou Y, Yang H, Tang L, Liu H. Screening and identification of key biomarkers in alimentary tract cancers: A bioinformatic analysis. Cancer Biomark 2021; 29:221-233. [PMID: 32623389 DOI: 10.3233/cbm-201580] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Alimentary tract cancers (ATCs) are the most malignant cancers in the world. Numerous studies have revealed the tumorigenesis, diagnosis and treatment of ATCs, but many mechanisms remain to be explored. METHODS To identify the key genes of ATCs, microarray datasets of oesophageal cancer, gastric cancer and colorectal cancer were obtained from the Gene Expression Omnibus (GEO) database. In total, 207 differentially expressed genes (DEGs) were screened. KEGG and GO function enrichment analyses were conducted, and a protein-protein interaction (PPI) network was generated and gene modules analysis was performed using STRING and Cytoscape. RESULTS Five hub genes were screened, and the associated biological processes indicated that these genes were mainly enriched in cellular processes, protein binding and metabolic processes. Clinical survival analysis showed that COL10A1 and KIF14 may be significantly associated with the tumorigenesis or pathology grade of ATCs. In addition, relative human ATC cell lines along with blood samples and tumour tissues of ATC patients were obtained. The data proved that high expression of COL10A1 and KIF14 was associated with tumorigenesis and could be detected in blood. CONCLUSION In conclusion, the identification of hub genes in the present study helped us to elucidate the molecular mechanisms of tumorigenesis and identify potential diagnostic indicators and targeted treatment for ATCs.
Collapse
Affiliation(s)
- Zeling Cai
- Genter of General Surgery, The Affiliated Haimen People's Hospital of Nantong University, Nantong, Jiangsu, China.,Genter of General Surgery, The Affiliated Haimen People's Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yi Wei
- Center of Gastrointestinal Disease, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China.,Genter of General Surgery, The Affiliated Haimen People's Hospital of Nantong University, Nantong, Jiangsu, China
| | - Shuai Chen
- Center of Gastrointestinal Disease, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China.,Genter of General Surgery, The Affiliated Haimen People's Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yu Gong
- Center of Gastrointestinal Disease, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Yue Fu
- Center of Gastrointestinal Disease, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Xianghua Dai
- Genter of General Surgery, The Affiliated Haimen People's Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yan Zhou
- Center of Gastrointestinal Disease, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Haojun Yang
- Center of Gastrointestinal Disease, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Liming Tang
- Center of Gastrointestinal Disease, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Hanyang Liu
- Center of Gastrointestinal Disease, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| |
Collapse
|
18
|
Zhou YM, Yao YL, Liu W, Shen XM, Shi LJ, Wu L. MicroRNA-134 inhibits tumor stem cell migration and invasion in oral squamous cell carcinomas via downregulation of PI3K-Akt signaling pathway by inhibiting LAMC2 expression. Cancer Biomark 2021; 29:51-67. [PMID: 32568182 DOI: 10.3233/cbm-191362] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is the most common malignant neoplasm of the mouth. Some studies have found that multiple microRNAs (miRs) participate in OSCC physiological and pathological processes. METHODS We explored the mechanism of action of miR-134 in OSCC involving the PI3K-Akt signaling pathway. Different bioinformatics methods were used to analyze the potential genes and their related miRs in OSCC. Tumor stem cells were separated from OSCCs through magnetic cell sorting. Regulatory pattern between miR-134 and LAMC2 in OSCC was evaluated by ectopic expression, knockdown and reporter assay experiments. The expression of miR-134, LAMC2, genes in PI3K-Akt signaling pathway, and apoptosis-related genes was detected. Cell proliferation was assessed by MTT assay, cell invasion by scratch test, cell migration by Transwell assay, cell cycle and apoptosis by flow cytometry, and cell growth and migration by xenograft tumor in nude mice. LAMC2 was predicted as the crucial factor related to OSCC using different chip data, and miR-134 was predicted to specifically bind LAMC2 in all five databases. RESULTS Overexpressed miR-134 or silenced LAMC2 was observed to inhibit cell proliferation, migration, invasion of OSCC cells, growth of subcutaneous xenograft in nude mice, as well as promote OSCC cell apoptosis. LAMC2, a target gene of miR-134, decreased following miR-134 promotion, while the PI3K-Akt signaling pathway was inactivated following LAMC2 knockdown. Furthermore, we also observed that the effect of overexpressed miR-134 was enhanced when LAMC2 was knocked down. CONCLUSIONS Taken together, these findings suggest that miR-134-mediated direct downregulation of LAMC2 inhibits migration and invasion of tumor stem cells in OSCC by suppressing the PI3K-Akt signaling pathway.
Collapse
Affiliation(s)
- Yong-Mei Zhou
- Department of Stomatology, Hainan West Central Hospital (Shanghai Ninth People's Hospital, Hainan Branch), Danzhou, Hainan, China.,Department of Oral Mucosal Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China.,Department of Stomatology, Hainan West Central Hospital (Shanghai Ninth People's Hospital, Hainan Branch), Danzhou, Hainan, China
| | - Yi-Lin Yao
- Department of Oral Mucosal Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China.,Department of Stomatology, Hainan West Central Hospital (Shanghai Ninth People's Hospital, Hainan Branch), Danzhou, Hainan, China
| | - Wei Liu
- National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China.,Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xue-Min Shen
- Department of Oral Mucosal Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| | - Lin-Jun Shi
- Department of Oral Mucosal Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| | - Lan Wu
- Department of Oral Mucosal Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
19
|
Shui JE, Wang W, Liu H, Stepanova A, Liao G, Qian J, Ai X, Ten V, Lu J, Cardoso WV. Prematurity alters the progenitor cell program of the upper respiratory tract of neonates. Sci Rep 2021; 11:10799. [PMID: 34031475 PMCID: PMC8144386 DOI: 10.1038/s41598-021-90093-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/29/2021] [Indexed: 11/09/2022] Open
Abstract
The impact of prematurity on human development and neonatal diseases, such as bronchopulmonary dysplasia, has been widely reported. However, little is known about the effects of prematurity on the programs of stem cell self-renewal and differentiation of the upper respiratory epithelium, which is key for adaptation to neonatal life. We developed a minimally invasive methodology for isolation of neonatal basal cells from nasopharyngeal (NP) aspirates and performed functional analysis in organotypic cultures to address this issue. We show that preterm NP progenitors have a markedly distinct molecular signature of abnormal proliferation and mitochondria quality control compared to term progenitors. Preterm progenitors had lower oxygen consumption at baseline and were unable to ramp up consumption to the levels of term cells when challenged. Although they formed a mucociliary epithelium, ciliary function tended to decline in premature cells as they differentiated, compared to term cells. Together, these differences suggested increased sensitivity of preterm progenitors to environmental stressors under non-homeostatic conditions.
Collapse
Affiliation(s)
- Jessica E Shui
- Division of Neonatology and Newborn Medicine, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Wei Wang
- Division of Neonatology and Newborn Medicine, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Helu Liu
- Columbia Center for Human Development, Pulmonary Allergy & Critical Care Medicine, Department of Medicine, Columbia University Irving Medical Center, 650 West 168th Street, BB 8-812, New York, NY, 10032, USA
| | - Anna Stepanova
- Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Grace Liao
- Columbia Center for Human Development, Pulmonary Allergy & Critical Care Medicine, Department of Medicine, Columbia University Irving Medical Center, 650 West 168th Street, BB 8-812, New York, NY, 10032, USA
- Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Jun Qian
- Columbia Center for Human Development, Pulmonary Allergy & Critical Care Medicine, Department of Medicine, Columbia University Irving Medical Center, 650 West 168th Street, BB 8-812, New York, NY, 10032, USA
| | - Xingbin Ai
- Division of Neonatology and Newborn Medicine, Department of Pediatrics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Vadim Ten
- Division of Neonatology, Department of Pediatrics, Columbia University Irving Medical Center, New York, NY, USA
| | - Jining Lu
- Division of Lung Diseases, NHLBI, NIH, Bethesda, MD, USA
| | - Wellington V Cardoso
- Columbia Center for Human Development, Pulmonary Allergy & Critical Care Medicine, Department of Medicine, Columbia University Irving Medical Center, 650 West 168th Street, BB 8-812, New York, NY, 10032, USA.
| |
Collapse
|
20
|
Zhang H, Weström S, Kappelin P, Virtanen M, Vahlquist A, Törmä H. Exploration of novel candidate genes involved in epidermal keratinocyte differentiation and skin barrier repair in man. Differentiation 2021; 119:19-27. [PMID: 34029921 DOI: 10.1016/j.diff.2021.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/11/2021] [Accepted: 04/27/2021] [Indexed: 12/26/2022]
Abstract
A proper skin barrier function requires constant formation of stratum corneum, i.e. the outermost layer of epidermis composed of terminally differentiated keratinocytes. The complex process of converting proliferative basal keratinocytes into corneocytes relies on programmed changes in the activity of many well-established genes. Much remains however to be investigated about this process, e.g. in conjunction with epidermal barrier defects due to genetic errors as in ichthyosis. To this end, we re-analyzed two sets of microarray-data comparing altered gene expression in differentiated vs. proliferating keratinocytes and in the skin of patients with autosomal recessive congenital ichthyosis (ARCI) vs. healthy controls, respectively. We thus identified 24 genes to be upregulated in both sets of array and not previously associated with keratinocyte differentiation. For 10 of these genes (AKR1B10, BLNK, ENDOU, GCNT4, GLTP, RHCG, SLC15A1, TMEM45B, TMEM86A and VSNL1), qPCR analysis confirmed the array results and subsequent immunostainings of normal epidermis showed superficial expression of several of the proteins. Furthermore, induction of keratinocyte differentiation using phorbol esters (PMA) resulted in increased expression of eight of the genes, whereas siRNA silencing of PPARδ, a transcription factor supporting differentiation, had the opposite effect. In summary, our results identify ten new candidate genes seemingly involved in human epidermal keratinocyte differentiation and possibly important for epidermal repair in a genetic skin disease characterized by barrier failure.
Collapse
Affiliation(s)
- Hanqian Zhang
- Department of Medical Sciences/Dermatology, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Simone Weström
- Department of Medical Sciences/Dermatology, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Per Kappelin
- Department of Medical Sciences/Dermatology, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Marie Virtanen
- Department of Medical Sciences/Dermatology, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Anders Vahlquist
- Department of Medical Sciences/Dermatology, Uppsala University, SE-751 85, Uppsala, Sweden
| | - Hans Törmä
- Department of Medical Sciences/Dermatology, Uppsala University, SE-751 85, Uppsala, Sweden.
| |
Collapse
|
21
|
Lin L, Cheng X, Yin D. Aberrant DNA Methylation in Esophageal Squamous Cell Carcinoma: Biological and Clinical Implications. Front Oncol 2020; 10:549850. [PMID: 33194605 PMCID: PMC7645039 DOI: 10.3389/fonc.2020.549850] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 09/18/2020] [Indexed: 12/20/2022] Open
Abstract
Almost all cancer cells possess multiple epigenetic abnormalities, which cooperate with genetic alterations to enable the acquisition of cancer hallmarks during tumorigenesis. As the most frequently found epigenetic change in human cancers, aberrant DNA methylation manifests at two major forms: global genomic DNA hypomethylation and locus-specific promoter region hypermethylation. It has been recognized as a critical contributor to esophageal squamous cell carcinoma (ESCC) malignant transformation. In ESCC, DNA methylation alterations affect genes involved in cell cycle regulation, DNA damage repair, and cancer-related signaling pathways. Aberrant DNA methylation patterns occur not only in ESCC tumors but also in precursor lesions. It adds another layer of complexity to the ESCC heterogeneity and may serve as early diagnostic, prognostic, and chemo-sensitive markers. Characterization of the DNA methylome in ESCC could help better understand its pathogenesis and develop improved therapies. We herein summarize the current research and knowledge about DNA methylation in ESCC and its clinical significance in diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
- Lehang Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xu Cheng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dong Yin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
22
|
Grishin D, Kasap E, Izotov A, Lisitsa A. Multifaceted ammonia transporters. ALL LIFE 2020. [DOI: 10.1080/26895293.2020.1812443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Affiliation(s)
- D.V. Grishin
- Institute of Biomedical Chemistry (IBMC), Moscow, Russia
| | - E.Y. Kasap
- Institute of Biomedical Chemistry (IBMC), Moscow, Russia
| | - A.A. Izotov
- Institute of Biomedical Chemistry (IBMC), Moscow, Russia
| | - A.V. Lisitsa
- Institute of Biomedical Chemistry (IBMC), Moscow, Russia
| |
Collapse
|
23
|
Yu-jing T, Wen-jing T, Biao T. Integrated Analysis of Hub Genes and Pathways In Esophageal Carcinoma Based on NCBI's Gene Expression Omnibus (GEO) Database: A Bioinformatics Analysis. Med Sci Monit 2020; 26:e923934. [PMID: 32756534 PMCID: PMC7431388 DOI: 10.12659/msm.923934] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 05/21/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Esophageal carcinoma (ESCA) is a health challenge with poor prognosis and limited treatment options. Our aim is to screen for hub genes and pathways associated with ESCA pathology as diagnostic or therapeutic targets. MATERIAL AND METHODS We downloaded 2 ESCA-related datasets from the Gene Expression Omnibus (GEO) database. Subsequently, differentially expressed genes (DEGs) of ESCA were determined by statistical analysis. Both Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of DEGs were performed using online analytic tools. Network analysis was employed to construct a protein-protein interaction (PPI) network and to filter hub genes. We evaluated the expression level and impact of hub genes on survival of ESCA patients using the OncoLoc webserver. RESULTS A total of 210 DEGs were identified. The GO analysis showed that the DEGs were enriched in cell division. The KEGG pathway analysis showed DEGs that were enriched in cell cycle regulation, known cancer pathways, the PI3K-Akt signaling pathway, and the cGMP-PKG signaling pathway. The top 10 hub genes were markedly upregulated in ESCA tissue compared with normal esophageal tissue. Moreover, the expression level of the hub genes was different at different pathological stages of ESCA. Further prognostic analysis identified that the top 10 hub genes were related to late survival of ESCA patients, while exhibiting few associations with early survival time. CONCLUSIONS The signaling pathways involving the DEGs probably represent the pathological mechanism underlying ESCA. The hub genes were associated with survival of ESCA patients, and as such have the potential to serve as diagnostic indicators and therapeutic targets.
Collapse
|
24
|
Le X, Mu J, Peng W, Tang J, Xiang Q, Tian S, Feng Y, He S, Qiu Z, Ren G, Huang A, Lin Y, Tao Q, Xiang T. DNA methylation downregulated ZDHHC1 suppresses tumor growth by altering cellular metabolism and inducing oxidative/ER stress-mediated apoptosis and pyroptosis. Theranostics 2020; 10:9495-9511. [PMID: 32863941 PMCID: PMC7449911 DOI: 10.7150/thno.45631] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 07/16/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer progression is an intricate biological process profiled by not only unscheduled proliferation, but also altered metabolism mechanisms. In this article, we introduced a novel tumor suppressor gene (TSG), Zinc Finger DHHC-Type Containing 1 (ZDHHC1, also known as ZNF377), frequently silenced due to epigenetic modification among various cancers, which exerts significant anti-tumor effects through metabolic regulation. Methods: Quantitative reversed-transcription PCR (qRT-PCR), reverse transcription PCR (RT-PCR) and Western blot were employed to demonstrate transcriptional and protein levels of targeted regulators. Methylation of ZDHHC1 promoter was detected by bisulfite genomic sequencing (BGS) and methylation specific PCR (MSP). Proteomics were analyzed by isobaric tags for relative and absolute quantitation (iTRAQ) and gas chromatography-mass spectrometry (GC-MS) were utilized for metabolomics analysis. Cellular functions were examined via corresponding approaches. Nude mice were used for xenograft tumor models. Indirect immunofluorescence staining was utilized to obtain precise location and expression of target proteins. Oxidative and ER stress indicators were detected using specific kits. Results: We found that ZDHHC1 expression was frequently silenced in multiple tumor cells and specimens due to methylation. Restoration of ZDHHC1 expression can curb cancer cell progression via stimulating apoptosis and cell cycle arrest, repressing metastasis, and reversing EMT transition and cell stemness. ZDHHC1's salient anti-tumor abilities were recognized in vivo as well. Metabolomic and proteomic analyses predicted inhibitory role of ZDHHC1 in glucose metabolism pathways in a CYGB-dependent manner, and in pentose phosphate pathway (PPP), which was validated by examining altered key factors. Moreover, we unraveled that ZDHHC1 dedicates to the increment of oxidative stress and endoplasmic reticulum (ER) stress to promote pyroptosis for anticancer purposes. Conclusion: Our study for the first time indicates ZDHHC1 is a potential tumor-suppressor frequently silenced due to promoter methylation, capable of negatively regulating metabolisms of tumor cells while stimulating oxidative stress and ER stress to expedite cell death through induction of pyroptosis and apoptosis, which can be exploited for development of new cancer prevention and therapies.
Collapse
Affiliation(s)
- Xin Le
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Junhao Mu
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weiyan Peng
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun Tang
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qin Xiang
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shaorong Tian
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yixiao Feng
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Sanxiu He
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhu Qiu
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guosheng Ren
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ailong Huang
- MOE Key Laboratory of Molecular Biology for Infectious Diseases, Department of Infectious Disease, Chongqing Medical University, China
| | - Yong Lin
- Lovelace Respiratory Research Institute, Albuquerque, New Mexico, USA
| | - Qian Tao
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Cancer Epigenetics Laboratory, Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir YK Pao Center for Cancer and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Tingxiu Xiang
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
25
|
Jin YQ, Miao DL. Multiomic Analysis of Methylation and Transcriptome Reveals a Novel Signature in Esophageal Cancer. Dose Response 2020; 18:1559325820942075. [PMID: 32728353 PMCID: PMC7364835 DOI: 10.1177/1559325820942075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/09/2020] [Accepted: 06/16/2020] [Indexed: 02/06/2023] Open
Abstract
Background: Epigenetic alterations have been shown to lead to human carcinogenesis. The aim of this study was to perform an integrative analysis to develop an epigenetic signature to predict overall survival (OS) of esophageal cancer. Methods: DNA methylation and messenger RNA expression data of esophageal cancer samples were downloaded from The Cancer Genome Atlas database and were incorporated and analyzed using an R package MethylMix. Functional enrichment analysis of the methylation-related differentially expressed genes (DEGs) was performed. Epigenetic signature and nomogram associated with the OS of esophageal cancer were established by the multivariate Cox model. Results: A total of 71 methylation-related DEGs were identified. Kyoto Encyclopedia of Genes and Genomes pathway analysis revealed that these genes were involved in the biological process related to the initiation and progression of esophageal cancer. Two-gene (FAM24B and FAM200A) risk signature for OS was developed by multivariate Cox analysis, of which had high accuracy. The signature is independent of clinicopathological variables and indicated better predictive power than other clinicopathological variables. Moreover, we developed a novel prognostic nomogram based on risk score and 3 clinicopathological factors. Conclusions: Our study indicated possible methylation-related DEGs and established an epigenetic signature, which may provide novel insights for understanding the pathogenesis of esophageal cancer.
Collapse
Affiliation(s)
- Yi-Qi Jin
- Department of Intervention and Vascular Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Dong-Liu Miao
- Department of Intervention and Vascular Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| |
Collapse
|
26
|
Xu W, Zou H, Wei Z, Song C, Tang C, Yin X, Wang Y, Han S, Cai Y, Han W. Rh type C-glycoprotein functions as a novel tumor suppressor gene by inhibiting tumorigenicity and metastasis in head and neck squamous cell carcinoma. Aging (Albany NY) 2020; 11:3601-3623. [PMID: 31170090 PMCID: PMC6594797 DOI: 10.18632/aging.102000] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 05/24/2019] [Indexed: 02/07/2023]
Abstract
Head and neck squamous cell carcinoma (HNSCC), a major histologic subtype of head and neck cancer, presents great mortality and morbidity worldwide. The aim of this study is to discover new potential biomarkers closely correlated with HNSCC progression. In this study, weighted gene co-expression network analysis was applied to construct a co-expression network, and the brown module was identified as the most correlated with HNSCC progression. Hub gene identification combined with survival analyses determined RHCG as a candidate biomarker for cancer progression and prognosis prediction. Further experimental results proved that RHCG was aberrantly downregulated in HNSCC tissues and cell lines. Moreover, decreased RHCG expression was shown to be associated with advanced stage and dismal prognosis in HNSCC patients. Functional assays revealed that RHCG could inhibit cell viability, clonogenicity, cell migration in vitro and suppress tumor formation in vivo. Further bioinformatics study demonstrated that DNA promoter hypermethylation of RHCG could lead to its downregulation and serve as potential prognostic maker in HNSCC. Our study reveals that RHCG acts as a tumor suppressor gene that plays a crucial role in inhibiting tumorigenicity and metastasis in HNSCC, which will shed light on the potential diagnostic and therapeutic strategies for HNSCC.
Collapse
Affiliation(s)
- Wenguang Xu
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Huihui Zou
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zheng Wei
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Chuanhui Song
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Chuanchao Tang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiteng Yin
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yufeng Wang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Shengwei Han
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yu Cai
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wei Han
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
27
|
Guo Y, Huang Q, Zheng J, Hsueh CY, Yuan X, Heng Y, Zhou L. Diagnostic Role of Dysregulated Circular RNA hsa_circ_0036722 in Laryngeal Squamous Cell Carcinoma. Onco Targets Ther 2020; 13:5709-5719. [PMID: 32606783 PMCID: PMC7306465 DOI: 10.2147/ott.s231076] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 05/17/2020] [Indexed: 12/14/2022] Open
Abstract
Purpose Dysregulated circular RNAs (circRNAs) have been shown to play important roles in various cancers, and could serve as diagnostic biomarkers. However, research focusing on the roles of the circRNAs in laryngeal squamous cell carcinoma (LSCC) is limited. This research aimed to explore the expressions of hsa_circ_0036722 in LSCCs and its diagnostic significance. Materials and Methods The expression levels of the circular RNA, hsa_circ_0036722, and its parental gene, RHCG, in 41 pairs of LSCC tissues and paired adjacent normal tissues were validated with quantitative real-time polymerase chain reaction (qRT-PCR). The diagnostic values of hsa_circ_0036722 alone and combined with RHCG in LSCC were evaluated using receiver operating characteristic (ROC) curves. Bioinformatics analysis predicted likely cross-talk between hsa_circ_0036722 and RHCG. Then, qRT-PCR and luciferase reporter assay were used to examine the effect of hsa_circ_0036722 on miR-1248 and miR‑1248 on RHCG expression. CCK-8 assays were conducted to investigate their effects on LSCC cell line. Results Hsa_circ_0036722 and RHCG were downregulated in LSCC tissues (P < 0.0001). The expression level of hsa_circ_0036722 was significantly correlated with the differentiation level of LSCC (P = 0.018). The area under the ROC curve of hsa_circ_0036722 was 0.838, which reached 0.859 when hsa_circ_0036722 was combined with RHCG as a biomarker. Mechanistically, hsa_circ_0036722 could directly sponge miR-1248 to antagonize its inhibitory effect on RHCG. And downregulation of hsa_circ_0036722 could promote the proliferation of LSCC cell line through upregulating miR-1248. Conclusion Our results indicated that hsa_circ_0036722 was downregulated in LSCC, which regulate the function of RHCG in LSCC via inhibiting miR-1248, and it could serve as a potential diagnostic marker for LSCC.
Collapse
Affiliation(s)
- Yang Guo
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Eye & ENT Hospital of Fudan University, Shanghai, People's Republic of China
| | - Qiang Huang
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Eye & ENT Hospital of Fudan University, Shanghai, People's Republic of China
| | - Juan Zheng
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Eye & ENT Hospital of Fudan University, Shanghai, People's Republic of China
| | - Chi-Yao Hsueh
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Eye & ENT Hospital of Fudan University, Shanghai, People's Republic of China
| | - Xiaohui Yuan
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Eye & ENT Hospital of Fudan University, Shanghai, People's Republic of China
| | - Yu Heng
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Eye & ENT Hospital of Fudan University, Shanghai, People's Republic of China
| | - Liang Zhou
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Key Clinical Disciplines of Otorhinolaryngology, Eye & ENT Hospital of Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
28
|
Multiple Functions of Fubp1 in Cell Cycle Progression and Cell Survival. Cells 2020; 9:cells9061347. [PMID: 32481602 PMCID: PMC7349734 DOI: 10.3390/cells9061347] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 12/11/2022] Open
Abstract
The discovery of novel and critical genes implicated in malignant development is a topic of high interest in cancer research. Intriguingly, a group of genes named “double-agent” genes were reported to have both oncogenic and tumor-suppressive functions. To date, less than 100 “double-agent” genes have been documented. Fubp1 is a master transcriptional regulator of a subset of genes by interacting with a far upstream element (FUSE). Mounting evidence has collectively demonstrated both the oncogenic and tumor suppressive roles of Fubp1 and the debate regarding its roles in tumorigenesis has been around for several years. Therefore, the detailed molecular mechanisms of Fubp1 need to be determined in each context. In the present study, we showed that the Fubp1 protein level was enriched in the S phase and we identified that Fubp1 deficiency altered cell cycle progression, especially in the S phase, by downregulating the mRNA expression levels of Ccna genes encoding cyclin A. Although this Fubp1-cyclin A axis appears to exist in several types of tumors, Fubp1 showed heterogeneous expression patterns among various cancer tissues, suggesting it exhibits multiple and complicated functions in cancer development. In addition, we showed that Fubp1 deficiency confers survival advantages to cells against metabolic stress and anti-cancer drugs, suggesting that Fubp1 may play both positive and negative roles in malignant development.
Collapse
|
29
|
Chen P, Liu C, Li P, Wang Q, Gao X, Wu H, Huang J. High RhCG expression predicts poor survival and promotes migration and proliferation of gastric cancer via keeping intracellular alkaline. Exp Cell Res 2020; 386:111740. [PMID: 31756312 DOI: 10.1016/j.yexcr.2019.111740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 11/15/2019] [Accepted: 11/18/2019] [Indexed: 11/25/2022]
Abstract
Advanced gastric cancer (GC) is aggressive with a high mortality rate. Rhesus family, C glycoprotein (RhCG) participates in tumor progression in many cancers, however its function in GC is still unknown. Here, we showed that RhCG was overexpressed in GC tissues at mRNA (P = 0.036) and protein levels (P < 0.05) compared with normal tissues. High RhCG level was correlated with poor differentiation (P = 0.037), TNM stage (P < 0.001), high HER-2 level (P = 0.018) and worse prognosis (P < 0.001). Cox proportional hazard model indicated that RhCG level was an independent prognostic biomarker. RhCG knockdown significantly decreased pHi and impeded tumor cellular proliferation, migration and invasion and repressed β-catenin and c-myc expression in GC cells. Moreover, GC cells with high RhCG level had reduced oxaliplatin efficacy suggesting a role for RhCG as a therapeutic target for GC. Our findings revealed a function of RhCG in cancer pathogenesis, invasion and metastasis in human GC. We suggest that RhCG act may as a novel prognostic indicator and a therapeutic target for gastric adenocarcinoma.
Collapse
Affiliation(s)
- Pei Chen
- Department of Clinical Biobank, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226000, China; Department of Oncology, Medical School of Nantong University, Nantong, Jiangsu, 226000, China
| | - Can Liu
- Department of Oncology, Medical School of Nantong University, Nantong, Jiangsu, 226000, China
| | - Peng Li
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226000, China
| | - Qingqing Wang
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226000, China
| | - Xiaodong Gao
- Department of General Surgery, Medical School of Nantong University, Nantong, Jiangsu, 226000, China
| | - Han Wu
- Department of General Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226000, China.
| | - JianFei Huang
- Department of Clinical Biobank, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226000, China; Institute of Oncology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu, China.
| |
Collapse
|
30
|
younesian O, younesian S, hosseinzadeh S, joshaghani HR. Association of Selenium and Risk of Esophageal Cancer: A Review. MEDICAL LABORATORY JOURNAL 2020. [DOI: 10.29252/mlj.14.1.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
|
31
|
Jin Y, Li Y, Wang X, Yang Y. Secretory leukocyte protease inhibitor suppresses HPV E6-expressing HNSCC progression by mediating NF-κB and Akt pathways. Cancer Cell Int 2019; 19:220. [PMID: 31462893 PMCID: PMC6708138 DOI: 10.1186/s12935-019-0942-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 08/18/2019] [Indexed: 12/24/2022] Open
Abstract
Background Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide and human papillomavirus (HPV) has been increasingly recognized as a pathogenic factor for the initiation and development of HNSCC. E6 oncogene, an essential component of the HPV 16 virus, acts as a leading cause of the malignant transformation of cancer cells. Therefore, investigating the biological effect and potential mechanisms of E6 oncogene on HNSCC cells and exploring potential therapeutic methods is of great value. Methods MTT assay, cell cycle analysis, and apoptosis assay were implemented to detect the biological effect of E6 oncogene on the growth of HNSCC cells. Wound healing assay and transwell assay were used to evaluate the role of E6 in the migration and invasion of HNSCC cells. Western blot and immunofluorescence assay were adopted to explore the regulatory mechanisms underlying E6-induced HNSCC progression. Then, exogenous secretory leukocyte protease inhibitor (SLPI) was added into the cell culture to investigate whether it could maintain its tumor suppressor effect on E6-expressing HNSCC cells. Results HPV E6 oncogene could promote the proliferation, cell cycle period, apoptosis resistance, migration and invasion of HNSCC cells by activating NF-κB and Akt pathways. Immunohistochemical analysis conducted on HNSCC tissues illustrated that SLPI was further downregulated in HPV positive HNSCC compared to HNSCC without HPV infection. Exogenous SLPI significantly inhibited HPV E6-mediated malignant phenotypes in HNSCC cells by inhibiting the activation of NF-κB and Akt and signaling pathways. Conclusions This study demonstrated that E6 oncogene led to the malignant transformation of HNSCC cells by regulating multiple pathways. SLPI could reverse the effect of E6 oncogene on HNSCC, implying that the functional inhibition of E6 by SLPI may be exploited as an attractive therapeutic strategy.
Collapse
Affiliation(s)
- Yu Jin
- 1Department of General Dentistry, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011 P. R. China.,Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200000 P. R. China
| | - Yuexiu Li
- Department of Stomatology, Tai'an Central Hospital, Tai'an, Shandong 271000 P. R. China
| | - Xin Wang
- 1Department of General Dentistry, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011 P. R. China.,Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200000 P. R. China
| | - Ya Yang
- 1Department of General Dentistry, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011 P. R. China.,Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai, 200000 P. R. China
| |
Collapse
|
32
|
Huang H, Huang Q, Tang T, Zhou X, Gu L, Lu X, Liu F. Differentially Expressed Gene Screening, Biological Function Enrichment, and Correlation with Prognosis in Non-Small Cell Lung Cancer. Med Sci Monit 2019; 25:4333-4341. [PMID: 31181055 PMCID: PMC6582684 DOI: 10.12659/msm.916962] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background The aim of this study was to explore the differently expressed genes and pathways in non-small cell lung cancer (NSCLC) and their correlation with the prognosis. Material/Methods Gene expression data series of GSE19804, GSE101929, and GSE33532 were downloaded from the Gene Expression Ominibus (GEO) database. The overlaping differently expressed genes (DEGs) were identified form the above 3 data series. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEEG) were used to analyze the biological functions and signal pathways of DEGs. The protein–protein interaction (PPI) was analyzed thorough Search Tool for the Retrieval of Interacting Gens (STRING). The relationship between the expression of hub genes and the prognosis of patients was analyzed by Kaplan-Meier Plotter online software. Results Twenty-nine DEGs were identified, with 22 upregulated genes and 7 downregulated genes. The enriched biological processes were mainly related to diet-induced thermogenesis and actin filament binding. The KEGG pathways were enriched in calcium signaling, regulation of lipolysis in adipocytes, and PPAR signaling. Two downregulated genes (MMP1 and SPP1) were identified as hub genes by Cytohubba. Twenty-two dysregulated genes were correlated with patient prognosis. Conclusions Differentially expressed genes are common in NSCLC patients and can be used as biomarkers for patient prognosis.
Collapse
Affiliation(s)
- He Huang
- Department of Respiratory, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Qingdong Huang
- Department of Respiratory, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Tingyu Tang
- Department of Respiratory, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Xiaoxi Zhou
- Department of Respiratory, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Liang Gu
- Department of Respiratory, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Xiaoling Lu
- Department of Respiratory, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| | - Fang Liu
- Department of Respiratory, Zhejiang Hospital, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|
33
|
Yang W, Zhao X, Han Y, Duan L, Lu X, Wang X, Zhang Y, Zhou W, Liu J, Zhang H, Zhao Q, Hong L, Fan D. Identification of hub genes and therapeutic drugs in esophageal squamous cell carcinoma based on integrated bioinformatics strategy. Cancer Cell Int 2019; 19:142. [PMID: 31139019 PMCID: PMC6530124 DOI: 10.1186/s12935-019-0854-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 05/10/2019] [Indexed: 12/13/2022] Open
Abstract
Background Esophageal squamous cell carcinoma (ESCC) is one of leading malignant cancers of gastrointestinal tract worldwide. Until now, the involved mechanisms during the development of ESCC are largely unknown. This study aims to explore the driven-genes and biological pathways in ESCC. Methods mRNA expression datasets of GSE29001, GSE20347, GSE100942, and GSE38129, containing 63 pairs of ESCC and non-tumor tissues data, were integrated and deeply analyzed. The bioinformatics approaches include identification of differentially expressed genes (DEGs) and hub genes, gene ontology (GO) terms analysis and biological pathway enrichment analysis, construction and analysis of protein-protein interaction (PPI) network, and miRNA-gene network construction. Subsequently, GEPIA2 database and qPCR assay were utilized to validate the expression of hub genes. DGIdb database was performed to search the candidate drugs for ESCC. Results Finally, 120 upregulated and 26 downregulated DEGs were identified. The functional enrichment of DEGs in ESCC were mainly correlated with cell cycle, DNA replication, deleted in colorectal cancer (DCC) mediated attractive signaling pathway, and Netrin-1 signaling pathway. The PPI network was constructed using STRING software with 146 nodes and 2392 edges. The most significant three modules in PPI were filtered and analyzed. Totally ten genes were selected and considered as the hub genes and nuclear division cycle 80 (NDC80) was closely related to the survival of ESCC patients. DGIdb database predicted 33 small molecules as the possible drugs for treating ESCC. Conclusions In summary, the data may provide new insights into ESCC pathogenesis and treatments. The candidate drugs may improve the efficiency of personalized therapy in future.
Collapse
Affiliation(s)
- Wanli Yang
- 1State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Xinhui Zhao
- 1State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Yu Han
- 2Department of Otolaryngology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lili Duan
- 1State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Xin Lu
- 3The School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xiaoqian Wang
- 1State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Yujie Zhang
- 1State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Wei Zhou
- 1State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Jinqiang Liu
- 1State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Hongwei Zhang
- 1State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Qingchuan Zhao
- 1State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Liu Hong
- 1State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Daiming Fan
- 1State Key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases, and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
34
|
Lu Y, Boswell W, Boswell M, Klotz B, Kneitz S, Regneri J, Savage M, Mendoza C, Postlethwait J, Warren WC, Schartl M, Walter RB. Application of the Transcriptional Disease Signature (TDSs) to Screen Melanoma-Effective Compounds in a Small Fish Model. Sci Rep 2019; 9:530. [PMID: 30679619 PMCID: PMC6345854 DOI: 10.1038/s41598-018-36656-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 11/22/2018] [Indexed: 12/20/2022] Open
Abstract
Cell culture and protein target-based compound screening strategies, though broadly utilized in selecting candidate compounds, often fail to eliminate candidate compounds with non-target effects and/or safety concerns until late in the drug developmental process. Phenotype screening using intact research animals is attractive because it can help identify small molecule candidate compounds that have a high probability of proceeding to clinical use. Most FDA approved, first-in-class small molecules were identified from phenotypic screening. However, phenotypic screening using rodent models is labor intensive, low-throughput, and very expensive. As a novel alternative for small molecule screening, we have been developing gene expression disease profiles, termed the Transcriptional Disease Signature (TDS), as readout of small molecule screens for therapeutic molecules. In this concept, compounds that can reverse, or otherwise affect known disease-associated gene expression patterns in whole animals may be rapidly identified for more detailed downstream direct testing of their efficacy and mode of action. To establish proof of concept for this screening strategy, we employed a transgenic strain of a small aquarium fish, medaka (Oryzias latipes), that overexpresses the malignant melanoma driver gene xmrk, a mutant egfr gene, that is driven by a pigment cell-specific mitf promoter. In this model, melanoma develops with 100% penetrance. Using the transgenic medaka malignant melanoma model, we established a screening system that employs the NanoString nCounter platform to quantify gene expression within custom sets of TDS gene targets that we had previously shown to exhibit differential transcription among xmrk-transgenic and wild-type medaka. Compound-modulated gene expression was identified using an internet-accessible custom-built data processing pipeline. The effect of a given drug on the entire TDS profile was estimated by comparing compound-modulated genes in the TDS using an activation Z-score and Kolmogorov-Smirnov statistics. TDS gene probes were designed that target common signaling pathways that include proliferation, development, toxicity, immune function, metabolism and detoxification. These pathways may be utilized to evaluate candidate compounds for potential favorable, or unfavorable, effects on melanoma-associated gene expression. Here we present the logistics of using medaka to screen compounds, as well as, the development of a user-friendly NanoString data analysis pipeline to support feasibility of this novel TDS drug-screening strategy.
Collapse
Affiliation(s)
- Yuan Lu
- Xiphophorus Genetic Stock Center, Department of Chemistry and Biochemistry, 419 Centennial Hall, Texas State University, San Marcos, TX, USA
| | - William Boswell
- Xiphophorus Genetic Stock Center, Department of Chemistry and Biochemistry, 419 Centennial Hall, Texas State University, San Marcos, TX, USA
| | - Mikki Boswell
- Xiphophorus Genetic Stock Center, Department of Chemistry and Biochemistry, 419 Centennial Hall, Texas State University, San Marcos, TX, USA
| | - Barbara Klotz
- Developmental Biochemistry, Biozentrum, University of Würzburg, Würzburg, Germany.,Comprehensive Cancer Center Mainfranken, University Clinic Würzburg, D-97074, Würzburg, Germany
| | - Susanne Kneitz
- Developmental Biochemistry, Biozentrum, University of Würzburg, Würzburg, Germany.,Comprehensive Cancer Center Mainfranken, University Clinic Würzburg, D-97074, Würzburg, Germany
| | - Janine Regneri
- Developmental Biochemistry, Biozentrum, University of Würzburg, Würzburg, Germany.,Comprehensive Cancer Center Mainfranken, University Clinic Würzburg, D-97074, Würzburg, Germany
| | - Markita Savage
- Xiphophorus Genetic Stock Center, Department of Chemistry and Biochemistry, 419 Centennial Hall, Texas State University, San Marcos, TX, USA
| | - Cristina Mendoza
- Xiphophorus Genetic Stock Center, Department of Chemistry and Biochemistry, 419 Centennial Hall, Texas State University, San Marcos, TX, USA
| | - John Postlethwait
- Institute of Neuroscience, University of Oregon, Eugene, Oregon, USA
| | | | - Manfred Schartl
- Developmental Biochemistry, Biozentrum, University of Würzburg, Würzburg, Germany.,Comprehensive Cancer Center Mainfranken, University Clinic Würzburg, D-97074, Würzburg, Germany.,Hagler Institute for Advanced Studies and Department of Biology, Texas A&M University, College Station, USA
| | - Ronald B Walter
- Xiphophorus Genetic Stock Center, Department of Chemistry and Biochemistry, 419 Centennial Hall, Texas State University, San Marcos, TX, USA.
| |
Collapse
|
35
|
Zhao Q, Liu S. [High expression of MMP20 may be a potential prognostic factor for poor outcomes of patients with endometrial carcinoma]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2018; 38:1239-1244. [PMID: 30377136 DOI: 10.3969/j.issn.1673-4254.2018.10.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To explore the expression of MMP20 in endometrial carcinoma and its clinical significance. METHODS Bioinformatics analysis was used to explore the correlation of MMP20 expression with the prognosis of the patients with endometrial carcinoma. We collected 21 pairs of fresh endometrial carcinoma and adjacent endometrial tissues to detect the expression of MMP20 mRNA using real-time PCR. We also examined MMP20 protein expressions in 134 paraffin-embedded endometrial carcinoma tissues and 34 paraffin-embedded endometrial tissues using immunohistochemistry. The correlation of MMP20 expression with the clinicopathological features and prognosis of the patients were analyzed. RESULTS Based on TCGA database analysis, the patients with endometrial carcinoma showing an increased MMP20 mRNA expression had a significantly poorer prognosis than those with a low MMP20 mRNA expression (P=0.00065). Real-time PCR analysis and immunohistochemistry confirmed the up-regulated expressions of MMP20 at both the mRNA (P=0.0062) and protein (P=0.005) levels in endometrial carcinoma tissues compared to the adjacent endometrial tissues. An elevated MMP20 protein expression was positively associated with FIGO stage of endometrial carcinoma (P=0.014) and inversely correlated with the survival time of the patients (P=0.019). The Cox regression model analysis showed that an increased MMP20 expression was an independent predictor of a poor prognosis of the patients with endometrial carcinoma (P=0.067). CONCLUSIONS High expression of MMP20 is a potential prognostic factor for poor outcomes of patients with endometrial carcinoma.
Collapse
Affiliation(s)
- Qian Zhao
- Department of Infection, Guiyang Maternal and Children's Healthcare Hospital, Guiyang 550003, China
| | - Shu Liu
- Department of Breast, Guiyang Maternal and Children's Healthcare Hospital, Guiyang 550003, China
| |
Collapse
|
36
|
RHCG suppresses cervical cancer progression through inhibiting migration and inducing apoptosis regulated by TGF-β1. Biochem Biophys Res Commun 2018; 503:86-93. [DOI: 10.1016/j.bbrc.2018.05.183] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 05/27/2018] [Indexed: 01/23/2023]
|