1
|
Yarza R, Bover M, Agulló-Ortuño MT, Iglesias-Docampo LC. Current approach and novel perspectives in nasopharyngeal carcinoma: the role of targeting proteasome dysregulation as a molecular landmark in nasopharyngeal cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:202. [PMID: 34154654 PMCID: PMC8215824 DOI: 10.1186/s13046-021-02010-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 06/07/2021] [Indexed: 12/15/2022]
Abstract
Nasopharyngeal carcinoma (NPC) represents a molecularly paradigmatic tumor given the complex diversity of environmental as well as host dependent factors that are closely implicated in tissue transformation and carcinogenesis. Epstein Barr Virus (EBV) plays a key role in tissue invasion, hyperplasia and malignant transformation. Therefore, EBV related oncoviral proteins such as Latent Membrane Protein family (LMP1, LMP2), Epstein Barr Nuclear Antigen 1 (EBNA1) and EBV related glycoprotein B (gB) are responsible for inducing intracellular signalling aberrations leading to sustained proliferation and further acquisition of NPC related invasive nature and metastatic potential.Dysregulation of proteasome signaling seems to be centrally implicated in oncoviral protein stabilization as well as in modulating tumor microenvironment. Different studies in vitro and in vivo suggest a potential role of proteasome inhibitors in the therapeutic setting of NPC. Furthermore, alterations affecting proteasome signalling in NPC have been associated to tumor growth and invasion, distant metastasis, immune exclusion and resistance as well as to clinical poor prognosis. So on, recent studies have shown the efficacy of immunotherapy as a suitable therapeutic approach to NPC. Nevertheless, novel strategies seem to look for combinatorial regimens aiming to potentiate immune recognition as well as to restore both primary and acquired immune resistance.In this work, our goal is to thoroughly review the molecular implications of proteasome dysregulation in the molecular pathogenesis of NPC, together with their direct relationship with EBV related oncoviral proteins and their role in promoting immune evasion and resistance. We also aim to hypothesize about the feasibility of the use of proteasome inhibitors as part of immunotherapy-including combinatorial regimens for their potential role in reversing immune resistance and favouring tumor recognition and eventual tumor death.
Collapse
Affiliation(s)
- Ramon Yarza
- Medical Oncology Division, Hospital Universitarioss 12 de Octubre, Avda. Córdoba s/n, E-28041, Madrid, Spain. .,Clinical and Translational Laboratory, Instituto de Investigación Hospital 12 de Octubre (I+12), Madrid, Spain.
| | - Mateo Bover
- Medical Oncology Division, Hospital Universitarioss 12 de Octubre, Avda. Córdoba s/n, E-28041, Madrid, Spain.,Clinical and Translational Laboratory, Instituto de Investigación Hospital 12 de Octubre (I+12), Madrid, Spain
| | - Maria Teresa Agulló-Ortuño
- Clinical and Translational Laboratory, Instituto de Investigación Hospital 12 de Octubre (I+12), Madrid, Spain. .,Lung Cancer Group, Clinical Research Program (H12O-CNIO), Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain. .,Biomedical Research Networking Centre: Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain. .,Facultad de Fisioterapia y Enfermería, Universidad de Castilla La Mancha (UCLM), Toledo, Spain.
| | - Lara Carmen Iglesias-Docampo
- Medical Oncology Division, Hospital Universitarioss 12 de Octubre, Avda. Córdoba s/n, E-28041, Madrid, Spain.,Clinical and Translational Laboratory, Instituto de Investigación Hospital 12 de Octubre (I+12), Madrid, Spain.,Lung Cancer Group, Clinical Research Program (H12O-CNIO), Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| |
Collapse
|
2
|
Ning Y, Zeng Z, Deng Y, Feng W, Huang L, Liu H, Lin J, Zhang C, Fan Y, Liu L. VPS33B interacts with NESG1 to suppress cell growth and cisplatin chemoresistance in ovarian cancer. Cancer Sci 2021; 112:1785-1797. [PMID: 33788346 PMCID: PMC8088924 DOI: 10.1111/cas.14864] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/12/2021] [Accepted: 02/17/2021] [Indexed: 12/12/2022] Open
Abstract
The pathogenesis and cisplatin chemoresistance of ovarian cancer (OC) are still unclear. Vacuolar protein sorting‐associated 33B (VPS33B) has not been reported in OC to date. In this study, immunohistochemistry was used to detect VPS33B protein expression between OC and ovarian tissues. MTT, EdU, colony formation, cell cycle, in vivo tumorigenesis, western blot, ChIP, EMSA, co‐immunoprecipitation (CoIP), qRT‐PCR, and microconfocal microscopy were used to explore the function and molecular mechanisms of VPS33B in OC cells. The results of the present study demonstrated that VPS33B protein expression was obviously reduced in OC compared with that in ovarian tissues. Overexpressed VPS33B suppressed cell cycle transition, cell growth, and chemoresistance to cisplatin in vitro and in vivo. Analysis of the mechanism indicated that overexpressed VPS33B regulated the epidermal growth factor receptor (EGFR)/PI3K/AKT/c‐Myc/p53/miR‐133a‐3p feedback loop and reduced the expression of the cell cycle factor CDK4. Nasopharyngeal epithelium‐specific protein 1 (NESG1) as a tumor suppressor not only interacted with VPS33B, but was also induced by VPS33B by the attenuation of PI3K/AKT/c‐Jun‐mediated transcription inhibition. Overexpressed NESG1 further suppressed cell growth by mediating VPS33B‐modulated signals in VPS33B‐overexpressing OC cells. Finally, NESG1 induced VPS33B expression by reducing the inhibition of PI3K/AKT/c‐Jun‐mediated transcription. Our study is the first to demonstrate that VPS33B serves as a tumor suppressor, and VPS33B can interact with NESG1 to suppress cell growth and promote cisplatin sensitivity by regulating the EGFR/PI3K/AKT/c‐Myc/p53/miR‐133a‐3p feedback loop in OC cells.
Collapse
Affiliation(s)
- Yingxia Ning
- Department of Gynecology, the Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.,Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhaoyang Zeng
- Department of Gynecology, the Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yuao Deng
- Department of Gynecology and Obstetrics, Shenzhen People's Hospital, Shenzhen, China
| | - Weifeng Feng
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Lun Huang
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Huiling Liu
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiazhi Lin
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chen Zhang
- Department of Clinical Pharmacy, Guangzhou First People's Hospital, Guangzhou, China
| | - Yue Fan
- Cancer Research Institute, Southern Medical University, Guangzhou, China
| | - Longyang Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
3
|
Chen Y, Liu Z, Wang H, Tang Z, Liu Y, Liang Z, Deng X, Zhao M, Fu Q, Li L, Cai H, Xie W, Fang W. VPS33B negatively modulated by nicotine functions as a tumor suppressor in colorectal cancer. Int J Cancer 2019; 146:496-509. [PMID: 31125123 DOI: 10.1002/ijc.32429] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 04/12/2019] [Accepted: 05/13/2019] [Indexed: 12/19/2022]
Abstract
The biological role of vacuolar protein sorting 33B (VPS33B) has not been examined in colorectal cancer (CRC). We report that VPS33B was downregulated in dextran sulfate sodium/azoxymethane (DSS/AOM) -induced CRC mice models and nicotine-treated CRC cells via the PI3K/AKT/c-Jun pathway. Reduced VPS33B is an unfavorable factor promoting poor prognosis in human CRC patients. VPS33B overexpression suppressed CRC proliferation, intrahepatic metastasis and chemoresistance of cisplatin (DDP) in vivo and in vitro through modulating the epidermal growth factor receptor (EGFR)/RAS/ERK/c-Myc/p53/miR-133a-3p feedback loop and the downstream cell cycle or EMT-related factors. Furthermore, NESG1 as a newly identified tumor suppressor interacted with VPS33B via colocalization in the cytoplasm, and it was stimulated by VPS33B through the downregulation of RAS/ERK/c-Jun-mediated transcription. NESG1 also activated VPS33B expression via the RAS/ERK/c-Jun pathway. Suppression of NESG1 increased cell growth, migration and invasion via the reversion of the VPS33B-modulating signal in VPS33B-overexpressed cells. Taken together, VPS33B as a tumor suppressor is easily dysregulated by chemical carcinogens and it interacts with NESG1 to modulate the EGFR/RAS/ERK/c-Myc/p53/miR-133a-3p feedback loop and thus suppress the malignant phenotype of CRC.
Collapse
Affiliation(s)
- Yiyu Chen
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.,School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhen Liu
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Huijun Wang
- School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Zibo Tang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yiyi Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Zixi Liang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaojie Deng
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Mengyang Zhao
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Qiaofen Fu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Libo Li
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongbing Cai
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Weibing Xie
- School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Weiyi Fang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
4
|
Chen Y, Jiang J, Zhao M, Luo X, Liang Z, Zhen Y, Fu Q, Deng X, Lin X, Li L, Luo R, Liu Z, Fang W. microRNA-374a suppresses colon cancer progression by directly reducing CCND1 to inactivate the PI3K/AKT pathway. Oncotarget 2018; 7:41306-41319. [PMID: 27191497 PMCID: PMC5173061 DOI: 10.18632/oncotarget.9320] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Accepted: 04/24/2016] [Indexed: 01/07/2023] Open
Abstract
microRNA-374a (miR-374a) exhibits oncogenic functions in various tumor types. Here we report that miR-374a suppresses proliferation, invasion, migration and intrahepatic metastasis in colon adenocarcinoma cell lines HCT116 and SW620. Notably, we detected that PI3K/AKT signaling and its downstream cell cycle factors including c-Myc, cyclin D1 (CCND1), CDK4 and epithelial-mesenchymal transition (EMT)-related genes including ZEB1, N-cadherin, Vimentin, Slug, and Snail were all significantly downregulated after miR-374a overexpression. Conversely, cell cycle inhibitors p21 and p27 were upregulated. Expression of E-cadherin was only decreased in HCT116, without any obvious differences observed in SW620 cells. Furthermore, luciferase reporter assays confirmed that miR-374a could directly reduce CCND1. Interestingly, when CCND1 was silenced or overexpressed, levels of pPI3K, pAkt as well as cell cycle and EMT genes were respectively downregulated or upregulated. We examined miR-374a levels by in situ hybridization and its correlation with CCND1 expression in CRC tumor tissues. High miR-374a expression with low level of CCND1 was protective factor in CRC. Together these findings indicate that miR-374a inactivates the PI3K/AKT axis by inhibiting CCND1, suppressing of colon cancer progression.
Collapse
Affiliation(s)
- Yiyu Chen
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, PR China.,Cancer Research Institute, Southern Medical University, Guangzhou, PR China
| | - Jingwen Jiang
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, PR China
| | - Mengyang Zhao
- Cancer Research Institute, Southern Medical University, Guangzhou, PR China
| | - Xiaojun Luo
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, PR China
| | - Zixi Liang
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, PR China.,Cancer Research Institute, Southern Medical University, Guangzhou, PR China
| | - Yan Zhen
- Cancer Research Institute, Southern Medical University, Guangzhou, PR China
| | - Qiaofen Fu
- Cancer Research Institute, Southern Medical University, Guangzhou, PR China
| | - Xiaojie Deng
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, PR China.,Cancer Research Institute, Southern Medical University, Guangzhou, PR China
| | - Xian Lin
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, PR China
| | - Libo Li
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, PR China
| | - Rongcheng Luo
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, PR China
| | - Zhen Liu
- Cancer Research Institute, Southern Medical University, Guangzhou, PR China.,Department of Pathology, School of Basic Medicine, Guangzhou Medical College, Guangzhou, PR China
| | - Weiyi Fang
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, PR China.,Cancer Research Institute, Southern Medical University, Guangzhou, PR China
| |
Collapse
|
5
|
Qi W, Shao F, Huang Q. Expression of Coiled-Coil Domain Containing 34 (CCDC34) and its Prognostic Significance in Pancreatic Adenocarcinoma. Med Sci Monit 2017; 23:6012-6018. [PMID: 29257799 PMCID: PMC5745713 DOI: 10.12659/msm.907951] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 11/27/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Coiled-coil domain containing 34 (CCDC34) promotes cell proliferation and invasive properties in human cancer. The aim of this study was to compare the expression of CCDC34 in pancreatic adenocarcinoma with normal pancreatic tissue, and to evaluate the prognostic significance of CCDC34 expression in patients with pancreatic adenocarcinoma, using bioinformatics. MATERIAL AND METHODS The expression and prognostic value of CCDC34 were initially predicted using Oncomine and The Cancer Genome Atlas (TCGA) databases. Pancreatic adenocarcinoma tissue samples (N=90) and matched normal pancreatic tissues (N=90) were studied using immunohistochemistry to measure CCDC34 protein expression levels. Univariate Kaplan-Meier, and multivariate Cox analysis were used to determine the prognostic role of CCDC34 expression. RESULTS Oncomine and TCGA databases predicted that CCDC34 mRNA expression levels were significantly increased in pancreatic adenocarcinoma compared with normal pancreatic tissues (P<0.05), and that patients with increased CCDC34 mRNA expression levels had significantly lower overall survival (OS) (P=0.031). Immunohistochemistry showed that expression levels of CCDC34 protein in pancreatic adenocarcinoma were significantly increased, compared with normal pancreas (P=0.000). Patients with pancreatic adenocarcinoma with increased expression of tissue CCDC34 had significantly reduced OS compared with patients with low expression (P=0.000). Univariate and multivariate survival analysis showed that increased expression of CCDC34 was an independent predictor of poor prognosis in patients with pancreatic adenocarcinoma (all, P=0.000). CONCLUSIONS Compared with normal pancreas, CCDC34 expression was significantly increased in pancreatic adenocarcinoma, and increased CCDC34 expression was an independent predictor of poor patient prognosis.
Collapse
Affiliation(s)
- Wei Qi
- Medical College of Shandong University, Jinan, Shandong, P.R. China
- Department of General Surgery, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui, P.R. China
| | - Feng Shao
- Department of General Surgery, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui, P.R. China
| | - Qiang Huang
- Department of General Surgery, Anhui Provincial Hospital, Anhui Medical University, Hefei, Anhui, P.R. China
| |
Collapse
|
6
|
Yin DT, Xu J, Lei M, Li H, Wang Y, Liu Z, Zhou Y, Xing M. Characterization of the novel tumor-suppressor gene CCDC67 in papillary thyroid carcinoma. Oncotarget 2016; 7:5830-41. [PMID: 26716505 PMCID: PMC4868724 DOI: 10.18632/oncotarget.6709] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 11/25/2015] [Indexed: 02/07/2023] Open
Abstract
Background Some studies showed an association of coiled-coil domain-containing (CCDC) genes with cancers. Our previous limited data specifically suggested a possible pathogenic role of CCDC67 in papillary thyroid cancer (PTC), but this has not been firmly established. The present study was to further investigate and establish this role of CCDC67 in PTC. Results The expression of CCDC67, both at mRNA and protein levels, was sharply down-regulated in PTC compared with normal thyroid tissues. Lower CCDC67 expression was significantly associated with aggressive tumor behaviors, such as advanced tumor stages and lymph node metastasis, as well as BRAF mutation. Introduced expression of CCDC67 in TPC-1 cells robustly inhibited cell proliferation, colony formation and migration, induced G1 phase cell cycle arrest, and increased cell apoptosis. Methods Primary PTC tumors and matched normal thyroid tissues were obtained from 200 unselected patients at the initial surgery for detection of CCDC67 mRNA and protein by RT-PCR and Western blotting analyses, respectively. Genomic DNA sequencing was performed to detect BRAF mutation in PTC tumors. Clinicopathological data were retrospectively reviewed for correlation analyses. PTC cell line TPC-1 with stable transfection of CCDC67 was used to investigate the functions of CCDC67. Conclusions This large study demonstrates down-regulation of CCDC67 in PTC, an inverse relationship between CCDC67 expression and PTC aggressiveness and BRAF mutation, and a robust inhibitory effect of CCDC67 on PTC cellular activities. These results are consistent with CCDC67 being a novel and impaired tumor suppressor gene in PTC, providing important prognostic and therapeutic implications for this cancer.
Collapse
Affiliation(s)
- De Tao Yin
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.,Key Discipline Laboratory of Clinical Medicine Henan, Zhengzhou 450052, P. R. China
| | - Jianhui Xu
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.,Key Discipline Laboratory of Clinical Medicine Henan, Zhengzhou 450052, P. R. China
| | - Mengyuan Lei
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.,Key Discipline Laboratory of Clinical Medicine Henan, Zhengzhou 450052, P. R. China
| | - Hongqiang Li
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.,Key Discipline Laboratory of Clinical Medicine Henan, Zhengzhou 450052, P. R. China
| | - Yongfei Wang
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.,Key Discipline Laboratory of Clinical Medicine Henan, Zhengzhou 450052, P. R. China
| | - Zhen Liu
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.,Key Discipline Laboratory of Clinical Medicine Henan, Zhengzhou 450052, P. R. China
| | - Yubing Zhou
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
| | - Mingzhao Xing
- Division of Endocrinology and Metabolism, the Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
7
|
A Review: Proteomics in Nasopharyngeal Carcinoma. Int J Mol Sci 2015; 16:15497-530. [PMID: 26184160 PMCID: PMC4519910 DOI: 10.3390/ijms160715497] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Revised: 06/08/2015] [Accepted: 07/01/2015] [Indexed: 12/24/2022] Open
Abstract
Although radiotherapy is generally effective in the treatment of major nasopharyngeal carcinoma (NPC), this treatment still makes approximately 20% of patients radioresistant. Therefore, the identification of blood or biopsy biomarkers that can predict the treatment response to radioresistance and that can diagnosis early stages of NPC would be highly useful to improve this situation. Proteomics is widely used in NPC for searching biomarkers and comparing differentially expressed proteins. In this review, an overview of proteomics with different samples related to NPC and common proteomics methods was made. In conclusion, identical proteins are sorted as follows: Keratin is ranked the highest followed by such proteins as annexin, heat shock protein, 14-3-3σ, nm-23 protein, cathepsin, heterogeneous nuclear ribonucleoproteins, enolase, triosephosphate isomerase, stathmin, prohibitin, and vimentin. This ranking indicates that these proteins may be NPC-related proteins and have potential value for further studies.
Collapse
|
8
|
ENO1 promotes tumor proliferation and cell adhesion mediated drug resistance (CAM-DR) in Non-Hodgkin's Lymphomas. Exp Cell Res 2015; 335:216-23. [PMID: 26024773 DOI: 10.1016/j.yexcr.2015.05.020] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Revised: 05/02/2015] [Accepted: 05/23/2015] [Indexed: 01/07/2023]
Abstract
Enolases are glycolytic enzymes responsible for the ATP-generated conversion of 2-phosphoglycerate to phosphoenolpyruvate. In addition to the glycolytic function, Enolase 1 (ENO1) has been reported up-regulation in several tumor tissues. In this study, we investigated the expression and biologic function of ENO1 in Non-Hodgkin's Lymphomas (NHLs). Clinically, by western blot analysis we observed that ENO1 expression was apparently higher in diffuse large B-cell lymphoma than in the reactive lymphoid tissues. Subsequently, immunohistochemical staining of 144 NHLs suggested that the expression of ENO1 was significantly lower in the indolent lymphomas compared with the progressive lymphomas. Further, we identified ENO1 as an independent prognostic factor, and it was significantly correlated with overall survival of NHL patients. In addition, we found that ENO1 could promote cell proliferation, regulate cell cycle associated gene and PI3K/AKT signaling pathway in NHLs. Finally, we verified that ENO1 participated in the process of lymphoma cell adhesion mediated drug resistance (CAM-DR). Adhesion to FN or HS5 cells significantly protected OCI-Ly8 and Daudi cells from cytotoxicity compared with those cultured in suspension, and these effects were attenuated when transfected with ENO1-siRNA. Based on the study, we propose that inhibition of ENO1 expression may be a novel strategy for therapy for NHLs patients, and it may be a target for drug resistance.
Collapse
|
9
|
Fu QF, Liu Y, Fan Y, Hua SN, Qu HY, Dong SW, Li RL, Zhao MY, Zhen Y, Yu XL, Chen YY, Luo RC, Li R, Li LB, Deng XJ, Fang WY, Liu Z, Song X. Alpha-enolase promotes cell glycolysis, growth, migration, and invasion in non-small cell lung cancer through FAK-mediated PI3K/AKT pathway. J Hematol Oncol 2015; 8:22. [PMID: 25887760 PMCID: PMC4359783 DOI: 10.1186/s13045-015-0117-5] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 02/09/2015] [Indexed: 02/04/2023] Open
Abstract
Background During tumor formation and expansion, increasing glucose metabolism is necessary for unrestricted growth of tumor cells. Expression of key glycolytic enzyme alpha-enolase (ENO1) is controversial and its modulatory mechanisms are still unclear in non-small cell lung cancer (NSCLC). Methods The expression of ENO1 was examined in NSCLC and non-cancerous lung tissues, NSCLC cell lines, and immortalized human bronchial epithelial cell (HBE) by quantitative real-time reverse transcription PCR (qRT-PCR), immunohistochemistry, and Western blot, respectively. The effects and modulatory mechanisms of ENO1 on cell glycolysis, growth, migration, invasion, and in vivo tumorigenesis and metastasis in nude mice were also analyzed. Results ENO1 expression was increased in NSCLC tissues in comparison to non-cancerous lung tissues. Similarly, NSCLC cell lines A549 and SPCA-1 also express higher ENO1 than HBE cell line in both mRNA and protein levels. Overexpressed ENO1 significantly elevated NSCLC cell glycolysis, proliferation, clone formation, migration, and invasion in vitro, as well as tumorigenesis and metastasis in vivo by regulating the expression of glycolysis, cell cycle, and epithelial-mesenchymal transition (EMT)-associated genes. Conversely, ENO1 knockdown reversed these effects. More importantly, our further study revealed that stably upregulated ENO1 activated FAK/PI3K/AKT and its downstream signals to regulate the glycolysis, cell cycle, and EMT-associated genes. Conclusion This study showed that ENO1 is responsible for NSCLC proliferation and metastasis; thus, ENO1 might serve as a potential molecular therapeutic target for NSCLC treatment. Electronic supplementary material The online version of this article (doi:10.1186/s13045-015-0117-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qiao-Fen Fu
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic China. .,Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Yan Liu
- Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Yue Fan
- Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Sheng-Ni Hua
- Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Hong-Ying Qu
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Su-Wei Dong
- Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Rui-Lei Li
- Department of Cancer Biotherapy Center, Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, People's Republic China.
| | - Meng-Yang Zhao
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic China. .,Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Yan Zhen
- Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Xiao-Li Yu
- Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Yi-Yu Chen
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic China. .,Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Rong-Cheng Luo
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Rong Li
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Li-Bo Li
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Xiao-Jie Deng
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic China. .,Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Wei-Yi Fang
- Cancer Center, Traditional Chinese Medicine-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, People's Republic China. .,Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Zhen Liu
- Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China. .,Department of Pathology, Basic School of Guangzhou Medical University, Guangzhou, Guangdong, People's Republic China.
| | - Xin Song
- Cancer Research Institute of Southern Medical University, Guangzhou, Guangdong, People's Republic China. .,Department of Cancer Biotherapy Center, Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, Yunnan, People's Republic China.
| |
Collapse
|