1
|
Li Q, Li C, Zhang Y, Zheng Z, Wang Y, Yang Y, Zhu Q, Wang R, Xu W, Zhu C, Tian Q, Wang M, Ye L. The tumor suppressor SALL2 opposes chemotherapeutic resistance in breast cancer. Mol Cell Biochem 2025; 480:2971-2983. [PMID: 39572504 DOI: 10.1007/s11010-024-05155-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 11/02/2024] [Indexed: 05/03/2025]
Abstract
Chemotherapy continues to be the primary treatment for certain types of breast cancer. However, despite an initial positive response to chemotherapeutic agents, the development of resistance is inevitable. The exact molecular mechanisms underlying this phenomenon remain unclear. In this research, a significant downregulation of SALL2 expression was observed in chemo-resistant breast cancer, which was attributed to promoter methylation. Decreased SALL2 expression correlated significantly with poorer relapse-free survival in chemotherapy-treated patients with breast cancer. Functionally, SALL2 silencing induced a stem cell-like phenotype in breast cancer cells, fostering resistance to cisplatin both in vitro and in vivo. This resistance was mediated, at least in part, through the transcriptional regulation of BTG2, a negative regulator of stemness, achieved by direct binding to its promoter regions. These findings underscore the critical role of SALL2 in modulating cisplatin response and propose SALL2 as a potential prognostic biomarker for chemotherapy response in breast cancer.
Collapse
Affiliation(s)
- Qiji Li
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Chenxin Li
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Yuhao Zhang
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Zihan Zheng
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Yun Wang
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Yingqian Yang
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Qingqing Zhu
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Rui Wang
- School of Medicine, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Wanhui Xu
- School of Medicine, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Chengming Zhu
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Qin Tian
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.
| | - Meng Wang
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors & Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Liping Ye
- The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|
2
|
Liang M, Sheng L, Ke Y, Wu Z. The research progress on radiation resistance of cervical cancer. Front Oncol 2024; 14:1380448. [PMID: 38651153 PMCID: PMC11033433 DOI: 10.3389/fonc.2024.1380448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024] Open
Abstract
Cervical carcinoma is the most prevalent gynecology malignant tumor and ranks as the fourth most common cancer worldwide, thus posing a significant threat to the lives and health of women. Advanced and early-stage cervical carcinoma patients with high-risk factors require adjuvant treatment following surgery, with radiotherapy being the primary approach. However, the tolerance of cervical cancer to radiotherapy has become a major obstacle in its treatment. Recent studies have demonstrated that radiation resistance in cervical cancer is closely associated with DNA damage repair pathways, the tumor microenvironment, tumor stem cells, hypoxia, cell cycle arrest, and epigenetic mechanisms, among other factors. The development of tumor radiation resistance involves complex interactions between multiple genes, pathways, and mechanisms, wherein each factor interacts through one or more signaling pathways. This paper provides an overview of research progress on an understanding of the mechanism underlying radiation resistance in cervical cancer.
Collapse
Affiliation(s)
| | | | - Yumin Ke
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Zhuna Wu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
3
|
Hermosilla VE, Gyenis L, Rabalski AJ, Armijo ME, Sepúlveda P, Duprat F, Benítez-Riquelme D, Fuentes-Villalobos F, Quiroz A, Hepp MI, Farkas C, Mastel M, González-Chavarría I, Jackstadt R, Litchfield DW, Castro AF, Pincheira R. Casein kinase 2 phosphorylates and induces the SALL2 tumor suppressor degradation in colon cancer cells. Cell Death Dis 2024; 15:223. [PMID: 38493149 PMCID: PMC10944491 DOI: 10.1038/s41419-024-06591-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 02/29/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
Spalt-like proteins are Zinc finger transcription factors from Caenorhabditis elegans to vertebrates, with critical roles in development. In vertebrates, four paralogues have been identified (SALL1-4), and SALL2 is the family's most dissimilar member. SALL2 is required during brain and eye development. It is downregulated in cancer and acts as a tumor suppressor, promoting cell cycle arrest and cell death. Despite its critical functions, information about SALL2 regulation is scarce. Public data indicate that SALL2 is ubiquitinated and phosphorylated in several residues along the protein, but the mechanisms, biological consequences, and enzymes responsible for these modifications remain unknown. Bioinformatic analyses identified several putative phosphorylation sites for Casein Kinase II (CK2) located within a highly conserved C-terminal PEST degradation motif of SALL2. CK2 is a serine/threonine kinase that promotes cell proliferation and survival and is often hyperactivated in cancer. We demonstrated that CK2 phosphorylates SALL2 residues S763, T778, S802, and S806 and promotes SALL2 degradation by the proteasome. Accordingly, pharmacological inhibition of CK2 with Silmitasertib (CX-4945) restored endogenous SALL2 protein levels in SALL2-deficient breast MDA-MB-231, lung H1299, and colon SW480 cancer cells. Silmitasertib induced a methuosis-like phenotype and cell death in SW480 cells. However, the phenotype was significantly attenuated in CRISPr/Cas9-mediated SALL2 knockout SW480 cells. Similarly, Sall2-deficient tumor organoids were more resistant to Silmitasertib-induced cell death, confirming that SALL2 sensitizes cancer cells to CK2 inhibition. We identified a novel CK2-dependent mechanism for SALL2 regulation and provided new insights into the interplay between these two proteins and their role in cell survival and proliferation.
Collapse
Affiliation(s)
- V E Hermosilla
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Laboratorio de Transducción de Señales y Cáncer, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Dept of Orofacial Sciences and Dept of Anatomy, University of California-San Francisco, San Francisco, CA, USA
| | - L Gyenis
- Department of Biochemistry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - A J Rabalski
- Department of Biochemistry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
- Odyssey Therapeutics, Boston, MA, USA
| | - M E Armijo
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Laboratorio de Transducción de Señales y Cáncer, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - P Sepúlveda
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Laboratorio de Transducción de Señales y Cáncer, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - F Duprat
- Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - D Benítez-Riquelme
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Laboratorio de Transducción de Señales y Cáncer, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - F Fuentes-Villalobos
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Laboratorio de Transducción de Señales y Cáncer, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Laboratorio de Inmunovirología. Departamento de Microbiologia. Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - A Quiroz
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Laboratorio de Transducción de Señales y Cáncer, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - M I Hepp
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Laboratorio de Transducción de Señales y Cáncer, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Laboratorio de Investigación en Ciencias Biomédicas, Departamento de Ciencias Básicas y Morfología, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - C Farkas
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Laboratorio de Transducción de Señales y Cáncer, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- Laboratorio de Investigación en Ciencias Biomédicas, Departamento de Ciencias Básicas y Morfología, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - M Mastel
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg. Cancer Progression and Metastasis Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - I González-Chavarría
- Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - R Jackstadt
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg. Cancer Progression and Metastasis Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - D W Litchfield
- Department of Biochemistry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - A F Castro
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.
- Laboratorio de Transducción de Señales y Cáncer, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.
| | - R Pincheira
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.
- Laboratorio de Transducción de Señales y Cáncer, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.
| |
Collapse
|
4
|
Chai T, Liu J, Liu Z, Fan R. Exosome-transported of circ_0081069 induces SPIN1 production by binding to miR-195-5p to inhibit radiosensitivity in esophageal squamous cell carcinoma. J Biochem Mol Toxicol 2024; 38:e23659. [PMID: 38348706 DOI: 10.1002/jbt.23659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 01/10/2024] [Accepted: 01/18/2024] [Indexed: 02/15/2024]
Abstract
Circ_0081069 plays a key role in tumor growth; however, its effect on radiosensitivity in esophageal squamous cell carcinoma (ESCC) remains unknown. The study is performed to reveal the association of circ_0081069 expression and radiosensitivity in ESCC and the underlying mechanism. Circ_0081069, miR-195-5p, and spindlin 1 (SPIN1) RNA expression were detected by quantitative real-time polymerase chain reaction. Protein expression was checked by Western blot analysis or immunohistochemistry assay. Cell viability, proliferation, cell apoptosis, migration, and invasion were investigated by cell counting kit-8, 5-Ethynyl-29-deoxyuridine, flow cytometry analysis, scratch test, and transwell assays, respectively. The sensitivity of ESCC cells to radiation was investigated by cell colony formation assay. The interactions among circ_0081069, miR-195-5p, and SPIN1 were identified by dual-luciferase reporter assay and RNA Immunoprecipitation assay. Xenograft mouse model assay was performed to determine the effect of circ_0007841 on radiosensitivity in vivo. Circ_0081069 and SPIN1 expression were upregulated, whereas miR-195-5p was downregulated in ESCC tissues, ESCC cells, and radiation-stimulated ESCC cells. Circ_0081069 silencing inhibited ESCC cell proliferation, invasion, and migration but improved cell apoptosis. In addition, circ_0081069 knockdown enhanced ESCC cell radiosensitivity in vitro and in vivo. Circ_0081069 bound to miR-195-5p and regulated radiosensitivity by binding to miR-195-5p in ESCC cells. Moreover, SPIN1, a target of miR-195-5p, rescued miR-195-5p-mediated effects in ESCC cells. Circ_0081069 was secreted from ESCC cells by being packaged into exosomes. Further, circ_0081069-Exo inhibited radiosensitivity in ESCC cells. Exosome-mediated transfer of circ_0081069 induced SPIN1 production by binding to miR-195-5p, further inhibiting radiosensitivity in ESCC.
Collapse
Affiliation(s)
- Ting Chai
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Radiotherapy, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Junqi Liu
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zongwen Liu
- Department of Radiotherapy, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ruitai Fan
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
5
|
Zhao Y, Ma Y, Bai Z, Wang T, Song D, Li T. Comparison of central venous catheter thoracic drainage and traditional closed thoracic drainage following minimally invasive surgery for esophageal carcinoma: a retrospective analysis. J Cardiothorac Surg 2023; 18:267. [PMID: 37794478 PMCID: PMC10552284 DOI: 10.1186/s13019-023-02373-4] [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: 03/02/2023] [Accepted: 09/26/2023] [Indexed: 10/06/2023] Open
Abstract
OBJECTIVE To compare the effectiveness and safety of central venous catheter thoracic drainage (CVCTD) with traditional closed thoracic drainage (TCTD) after minimally invasive surgery for esophageal cancer. METHODS We conducted a retrospective investigation of 103 patients who underwent minimally invasive esophageal cancer surgery at our institution between January 2017 and December 2019. Among them, 44 patients underwent CVCTD, while 59 received TCTD. We compared the following outcomes between the two cohorts: drainage volume, duration of drainage, postoperative complications (including pleural effusion, pulmonary infection, atelectasis, anastomotic leakage, etc.), length of hospital stay, and postoperative pain assessment. RESULTS No significant differences were observed between the experimental and control groups regarding postoperative thoracic drainage, the timing of postoperative tube removal, or postoperative complications. However, significant disparities were noted in the duration of postoperative hospitalization, drainage tube healing time, and pain threshold among the esophageal cancer patients in both cohorts (p < 0.05). CONCLUSION CVCTD is a secure and potent alternative to TCTD following minimally invasive surgery for esophageal carcinoma. It potentially contributes to reducing the incidence of postoperative complications while curtailing the duration of hospitalization. Additional research is warranted to substantiate these findings.
Collapse
Affiliation(s)
- Yang Zhao
- Clinical Medical College, Ningxia Medical University, Yinchuan, Ningxia, 750004, China
- Department of Surgical Oncology II, General Hospital of Ningxia Medical University, No.804 Shengli Road, Xingqing District, Yinchuan, Ningxia, Ningxia, 750004, China
| | - Yue Ma
- Clinical Medical College, Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Zhixia Bai
- Department of Anesthesiology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Tao Wang
- Department of Surgical Oncology II, General Hospital of Ningxia Medical University, No.804 Shengli Road, Xingqing District, Yinchuan, Ningxia, Ningxia, 750004, China
| | - Dong Song
- Department of Surgical Oncology II, General Hospital of Ningxia Medical University, No.804 Shengli Road, Xingqing District, Yinchuan, Ningxia, Ningxia, 750004, China
| | - Tao Li
- Clinical Medical College, Ningxia Medical University, Yinchuan, Ningxia, 750004, China.
- Department of Surgical Oncology II, General Hospital of Ningxia Medical University, No.804 Shengli Road, Xingqing District, Yinchuan, Ningxia, Ningxia, 750004, China.
| |
Collapse
|
6
|
Khojasteh-Leylakoohi F, Mohit R, Khalili-Tanha N, Asadnia A, Naderi H, Pourali G, Yousefli Z, Khalili-Tanha G, Khazaei M, Maftooh M, Nassiri M, Hassanian SM, Ghayour-Mobarhan M, Ferns GA, Shahidsales S, Lam AKY, Giovannetti E, Nazari E, Batra J, Avan A. Down regulation of Cathepsin W is associated with poor prognosis in pancreatic cancer. Sci Rep 2023; 13:16678. [PMID: 37794108 PMCID: PMC10551021 DOI: 10.1038/s41598-023-42928-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 09/16/2023] [Indexed: 10/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with a very poor prognosis. Therefore, there has been a focus on identifying new biomarkers for its early diagnosis and the prediction of patient survival. Genome-wide RNA and microRNA sequencing, bioinformatics and Machine Learning approaches to identify differentially expressed genes (DEGs), followed by validation in an additional cohort of PDAC patients has been undertaken. To identify DEGs, genome RNA sequencing and clinical data from pancreatic cancer patients were extracted from The Cancer Genome Atlas Database (TCGA). We used Kaplan-Meier analysis of survival curves was used to assess prognostic biomarkers. Ensemble learning, Random Forest (RF), Max Voting, Adaboost, Gradient boosting machines (GBM), and Extreme Gradient Boosting (XGB) techniques were used, and Gradient boosting machines (GBM) were selected with 100% accuracy for analysis. Moreover, protein-protein interaction (PPI), molecular pathways, concomitant expression of DEGs, and correlations between DEGs and clinical data were analyzed. We have evaluated candidate genes, miRNAs, and a combination of these obtained from machine learning algorithms and survival analysis. The results of Machine learning identified 23 genes with negative regulation, five genes with positive regulation, seven microRNAs with negative regulation, and 20 microRNAs with positive regulation in PDAC. Key genes BMF, FRMD4A, ADAP2, PPP1R17, and CACNG3 had the highest coefficient in the advanced stages of the disease. In addition, the survival analysis showed decreased expression of hsa.miR.642a, hsa.mir.363, CD22, BTNL9, and CTSW and overexpression of hsa.miR.153.1, hsa.miR.539, hsa.miR.412 reduced survival rate. CTSW was identified as a novel genetic marker and this was validated using RT-PCR. Machine learning algorithms may be used to Identify key dysregulated genes/miRNAs involved in the disease pathogenesis can be used to detect patients in earlier stages. Our data also demonstrated the prognostic and diagnostic value of CTSW in PDAC.
Collapse
Affiliation(s)
- Fatemeh Khojasteh-Leylakoohi
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Mohit
- Department of Anesthesia, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Nima Khalili-Tanha
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Asadnia
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Naderi
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ghazaleh Pourali
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Yousefli
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ghazaleh Khalili-Tanha
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mina Maftooh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammadreza Nassiri
- Recombinant Proteins Research Group, The Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Ghayour-Mobarhan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton and Sussex Medical School, Division of Medical Education, Falmer, Brighton, BN1 9PH, Sussex, UK
| | | | - Alfred King-Yin Lam
- Pathology, School of Medicine and Dentistry, Griffith University, Gold Coast Campus, Gold Coast, QLD, 4222, Australia
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam U.M.C., VU. University Medical Center (VUMC), Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC Start up Unit, Fondazione Pisana Per La Scienza, Pisa, Italy
| | - Elham Nazari
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Health Information, Technology and Management, School of Allied Medical Sciences, Shahid BeheshtiUniversity of Medical Science, Tehran, Iran.
| | - Jyotsna Batra
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, 4000, Australia
- Translational Research Institute, Queensland University of Technology, Brisbane, 4102, Australia
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- College of Medicine, University of Warith Al-Anbiyaa, Karbala, Iraq.
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, 4000, Australia.
| |
Collapse
|
7
|
Lin Y, Ye Y, Huang Q, Zheng B, Yang Y, Chen Y, Li W, Ke H, Lin C, Zhang Y, Wang L, Chen J, Xu Y. Influence of age as a continuous variable on survival outcomes and treatment options in patients with upper thoracic esophageal carcinoma. J Cancer 2023; 14:1039-1048. [PMID: 37151386 PMCID: PMC10158516 DOI: 10.7150/jca.83490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 03/30/2023] [Indexed: 05/09/2023] Open
Abstract
Background: This retrospective review of patients with upper thoracic esophageal squamous cell carcinoma (ESCC) analyzed the prognostic value of age, as a continuous variable, and offered insight into treatment options. Methods: 568 upper ESCC patients underwent radical therapy between 2004 and 2016. Age as a continuous variable was entered into the Cox regression model with penalized spline (P-spline) analysis to investigate a correlation between age and survival outcomes. Results: Before adjustment, P-spline regression revealed U-shaped survival curves. Sixty years was the optimal cut-off age for differences in overall and progression-free survival (OS, PFS). The cohort was divided into age groups ≤ 50, 51-69, and ≥ 70 years. Multivariate analyses showed no significant differences in either PFS or OS for patients aged ≤ 50 and 51-69 years. After adjusting for covariates, P-spline regression showed that the risk of mortality and disease progression increased with age, and ≥ 70 years was an unfavorable independent prognostic factor. For age ≥ 70 years, the OS and PFS associated with non-surgery was comparable to that of surgery. For patients younger, the OS and PFS of patients given surgery was significantly better than that of patients given non-surgery. Conclusion: Age was an independent prognostic factor for upper ESCC. Patients ≥ 70 years achieved no significant survival benefit from surgery, but for those younger than 70 years surgery was the preferred treatment option.
Collapse
Affiliation(s)
- Yu Lin
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Yuling Ye
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Qiuyuan Huang
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Binglin Zheng
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Yong Yang
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Yuanmei Chen
- Department of Thoracic Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hosptial, Fuzhou 350014, China
| | - Weiguang Li
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Hongqian Ke
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Chuyan Lin
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Yiping Zhang
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Liyan Wang
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Junqiang Chen
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Yuanji Xu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
| |
Collapse
|
8
|
Zapata-García JA, Riveros-Magaña AR, Ortiz-Lazareno PC, Hernández-Flores G, Jave-Suárez LF, Aguilar-Lemarroy A. Comparative Genomic Hybridization and Transcriptome Sequencing Reveal Genes with Gain in Acute Lymphoblastic Leukemia: JUP Expression Emerges as a Survival-Related Gene. Diagnostics (Basel) 2022; 12:diagnostics12112788. [PMID: 36428851 PMCID: PMC9689318 DOI: 10.3390/diagnostics12112788] [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: 09/29/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) in children or adults is characterized by structural and numeric aberrations in chromosomes; these anomalies strongly correlate with prognosis and clinical outcome. Therefore, this work aimed to identify the genes present in chromosomal gain regions found more frequently in patients with acute lymphoblastic leukemia (ALL) and ALL-derived cell lines using comparative genomic hybridization (CGH). In addition, validation of the genes found in these regions was performed utilizing RNAseq from JURKAT, CEM, and SUP-B15 cell lines, as well as expression microarrays derived from a MILE study. Chromosomes with common gain zones that were maintained in six or more samples were 14, 17, and 22, in which a total of 22 genes were identified. From them, NT5C3B, CNP, ACLY, and GNB1L maintained overexpression at the mRNA level in the cell lines and in patients with ALL. It is noteworthy that SALL2 showed very high expression in T-ALL, while JUP was highly expressed in B-ALL lineages. Interestingly, the latter correlated with worse survival in patients. This provided evidence that the measurement of these genes has high potential for clinical utility; however, their expressions should first be evaluated with a sensitive test in a more significant number of patients.
Collapse
Affiliation(s)
- Jessica Alejandra Zapata-García
- Programa de Doctorado en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara C.P. 44340, Mexico
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara C.P. 44340, Mexico
| | - Alma Rocío Riveros-Magaña
- Centro Universitario del Sur, Universidad de Guadalajara, Ciudad Guzmán C.P. 49000, Mexico
- Hospital General Zona 9, Ciudad Guzmán C.P. 49000, Mexico
| | - Pablo Cesar Ortiz-Lazareno
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara C.P. 44340, Mexico
| | - Georgina Hernández-Flores
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara C.P. 44340, Mexico
| | - Luis Felipe Jave-Suárez
- Programa de Doctorado en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara C.P. 44340, Mexico
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara C.P. 44340, Mexico
| | - Adriana Aguilar-Lemarroy
- Programa de Doctorado en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara C.P. 44340, Mexico
- División de Inmunología, Centro de Investigación Biomédica de Occidente (CIBO), Instituto Mexicano del Seguro Social (IMSS), Guadalajara C.P. 44340, Mexico
- Correspondence: ; Tel.: +52-331-520-7625
| |
Collapse
|
9
|
Riffo E, Palma M, Hepp MI, Benítez-Riquelme D, Torres VA, Castro AF, Pincheira R. The Sall2 transcription factor promotes cell migration regulating focal adhesion turnover and integrin β1 expression. Front Cell Dev Biol 2022; 10:1031262. [PMID: 36438565 PMCID: PMC9682130 DOI: 10.3389/fcell.2022.1031262] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/25/2022] [Indexed: 01/02/2025] Open
Abstract
SALL2/Sall2 is a transcription factor associated with development, neuronal differentiation, and cancer. Interestingly, SALL2/Sall2 deficiency leads to failure of the optic fissure closure and neurite outgrowth, suggesting a positive role for SALL2/Sall2 in cell migration. However, in some cancer cells, SALL2 deficiency is associated with increased cell migration. To further investigate the role of Sall2 in the cell migration process, we used immortalized Sall2 knockout (Sall2 -/- ) and Sall2 wild-type (Sall2 +/+ ) mouse embryonic fibroblasts (iMEFs). Our results indicated that Sall2 positively regulates cell migration, promoting cell detachment and focal adhesions turnover. Sall2 deficiency decreased cell motility and altered focal adhesion dynamics. Accordingly, restoring Sall2 expression in the Sall2 -/- iMEFs by using a doxycycline-inducible Tet-On system recovered cell migratory capabilities and focal adhesion dynamics. In addition, Sall2 promoted the autophosphorylation of Focal Adhesion Kinase (FAK) at Y397 and increased integrin β1 mRNA and its protein expression at the cell surface. We demonstrated that SALL2 increases ITGB1 promoter activity and binds to conserved SALL2-binding sites at the proximal region of the ITGB1 promoter, validated by ChIP experiments. Furthermore, the overexpression of integrin β1 or its blockade generates a cell migration phenotype similar to that of Sall2 +/+ or Sall2 -/- cells, respectively. Altogether, our data showed that Sall2 promotes cell migration by modulating focal adhesion dynamics, and this phenotype is associated with SALL2/Sall2-transcriptional regulation of integrin β1 expression and FAK autophosphorylation. Since deregulation of cell migration promotes congenital abnormalities, tumor formation, and spread to other tissues, our findings suggest that the SALL2/Sall2-integrin β1 axis could be relevant for those processes.
Collapse
Affiliation(s)
- Elizabeth Riffo
- Laboratorio de Transducción de Señales y Cáncer, Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Mario Palma
- Laboratorio de Transducción de Señales y Cáncer, Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Matías I. Hepp
- Laboratorio de Investigación en Ciencias Biomédicas, Departamento de Ciencias Básicas y Morfología, Facultad de Medicina, Universidad Católica de la Santísima Concepción, Concepción, Chile
| | - Diego Benítez-Riquelme
- Laboratorio de Transducción de Señales y Cáncer, Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Vicente A. Torres
- Millennium Institute on Immunology and Immunotherapy, ICOD, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Ariel F. Castro
- Laboratorio de Transducción de Señales y Cáncer, Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Roxana Pincheira
- Laboratorio de Transducción de Señales y Cáncer, Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| |
Collapse
|
10
|
Gallerani G, Rossi T, Valgiusti M, Angeli D, Fici P, De Fanti S, Bandini E, Cocchi C, Frassineti GL, Bonafè M, Fabbri F. CNA Profiling of Single CTCs in Locally Advanced Esophageal Cancer Patients during Therapy Highlights Unexplored Molecular Pathways. Cancers (Basel) 2021; 13:6369. [PMID: 34944989 PMCID: PMC8699413 DOI: 10.3390/cancers13246369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Here, we monitored the evolution of CTCs spread in 11 patients affected by locally advanced EC who were undergoing therapy. METHODS In this perspective study, we designed multiple blood biopsies from individual patients: before and after neoadjuvant chemo-radio therapy and after surgery. We developed a multi-target array, named Grab-all assay, to estimate CTCs for their epithelial (EpCAM/E-Cadherin/Cytokeratins) and mesenchymal/stem (N-Cadherin/CD44v6/ABCG2) phenotypes. Identified CTCs were isolated as single cells by DEPArray, subjected to whole genome amplification, and copy number aberration (CNA) profiles were determined. Through bioinformatic analysis, we assessed the genomic imbalance of single CTCs, investigated specific focal copy number changes previously reported in EC and aberrant pathways using enrichment analysis. RESULTS Longitudinal monitoring allowed the identification of CTCs in at least one time-point per patient. Through single cell CNA analysis, we revealed that CTCs showed significantly dynamic genomic imbalance during treatment. Individual CTCs from relapsed patients displayed a higher degree of genomic imbalance relative to disease-free patients' groups. Genomic aberrations previously reported in EC occurred mostly in post-neoadjuvant therapy CTCs. In-depth analysis showed that networks enrichment in all time-point CTCs were inherent to innate immune system. Transcription/gene regulation, post-transcriptional and epigenetic modifications were uniquely affected in CTCs of relapsed patients. CONCLUSIONS Our data add clues to the comprehension of the role of CTCs in EC aggressiveness: chromosomal aberrations on genes related to innate immune system behave as relevant to the onset of CTC-status, whilst pathways of transcription/gene regulation, post-transcriptional and epigenetic modifications seem linked to patients' outcome.
Collapse
Affiliation(s)
- Giulia Gallerani
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (T.R.); (P.F.); (E.B.); (C.C.); (F.F.)
| | - Tania Rossi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (T.R.); (P.F.); (E.B.); (C.C.); (F.F.)
| | - Martina Valgiusti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.V.); (G.L.F.)
| | - Davide Angeli
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Pietro Fici
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (T.R.); (P.F.); (E.B.); (C.C.); (F.F.)
| | - Sara De Fanti
- Interdepartmental Centre “Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate)”, University of Bologna, 40126 Bologna, Italy;
| | - Erika Bandini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (T.R.); (P.F.); (E.B.); (C.C.); (F.F.)
| | - Claudia Cocchi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (T.R.); (P.F.); (E.B.); (C.C.); (F.F.)
| | - Giovanni Luca Frassineti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.V.); (G.L.F.)
| | - Massimiliano Bonafè
- Department of Experimental and Diagnostic Medicine, University of Bologna, 40126 Bologna, Italy;
| | - Francesco Fabbri
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (T.R.); (P.F.); (E.B.); (C.C.); (F.F.)
| |
Collapse
|
11
|
SALL Proteins; Common and Antagonistic Roles in Cancer. Cancers (Basel) 2021; 13:cancers13246292. [PMID: 34944911 PMCID: PMC8699250 DOI: 10.3390/cancers13246292] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/06/2021] [Accepted: 12/09/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary Transcription factors play essential roles in regulating gene expression, impacting the cell phenotype and function, and in the response of cells to environmental conditions. Alterations in transcription factors, including gene amplification or deletion, point mutations, and expression changes, are implicated in carcinogenesis, cancer progression, metastases, and resistance to cancer treatments. Not surprisingly, transcription factor activity is altered in numerous cancers, representing a unique class of cancer drug targets. This review updates and integrates information on the SALL family of transcription factors, highlighting the synergistic and/or antagonistic functions they perform in various cancer types. Abstract SALL proteins are a family of four conserved C2H2 zinc finger transcription factors that play critical roles in organogenesis during embryonic development. They regulate cell proliferation, survival, migration, and stemness; consequently, they are involved in various human genetic disorders and cancer. SALL4 is a well-recognized oncogene; however, SALL1–3 play dual roles depending on the cancer context and stage of the disease. Current reviews of SALLs have focused only on SALL2 or SALL4, lacking an integrated view of the SALL family members in cancer. Here, we update the recent advances of the SALL members in tumor development, cancer progression, and therapy, highlighting the synergistic and/or antagonistic functions they perform in similar cancer contexts. We identified common regulatory mechanisms, targets, and signaling pathways in breast, brain, liver, colon, blood, and HPV-related cancers. In addition, we discuss the potential of the SALL family members as cancer biomarkers and in the cancer cells’ response to therapies. Understanding SALL proteins’ function and relationship will open new cancer biology, clinical research, and therapy perspectives.
Collapse
|
12
|
Blomain ES, Moding EJ. Liquid Biopsies for Molecular Biology-Based Radiotherapy. Int J Mol Sci 2021; 22:11267. [PMID: 34681925 PMCID: PMC8538046 DOI: 10.3390/ijms222011267] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/13/2021] [Accepted: 10/15/2021] [Indexed: 11/29/2022] Open
Abstract
Molecular alterations drive cancer initiation and evolution during development and in response to therapy. Radiotherapy is one of the most commonly employed cancer treatment modalities, but radiobiologic approaches for personalizing therapy based on tumor biology and individual risks remain to be defined. In recent years, analysis of circulating nucleic acids has emerged as a non-invasive approach to leverage tumor molecular abnormalities as biomarkers of prognosis and treatment response. Here, we evaluate the roles of circulating tumor DNA and related analyses as powerful tools for precision radiotherapy. We highlight emerging work advancing liquid biopsies beyond biomarker studies into translational research investigating tumor clonal evolution and acquired resistance.
Collapse
Affiliation(s)
- Erik S. Blomain
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA;
| | - Everett J. Moding
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA;
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
13
|
Integration of machine learning and genome-scale metabolic modeling identifies multi-omics biomarkers for radiation resistance. Nat Commun 2021; 12:2700. [PMID: 33976213 PMCID: PMC8113601 DOI: 10.1038/s41467-021-22989-1] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 04/09/2021] [Indexed: 02/07/2023] Open
Abstract
Resistance to ionizing radiation, a first-line therapy for many cancers, is a major clinical challenge. Personalized prediction of tumor radiosensitivity is not currently implemented clinically due to insufficient accuracy of existing machine learning classifiers. Despite the acknowledged role of tumor metabolism in radiation response, metabolomics data is rarely collected in large multi-omics initiatives such as The Cancer Genome Atlas (TCGA) and consequently omitted from algorithm development. In this study, we circumvent the paucity of personalized metabolomics information by characterizing 915 TCGA patient tumors with genome-scale metabolic Flux Balance Analysis models generated from transcriptomic and genomic datasets. Metabolic biomarkers differentiating radiation-sensitive and -resistant tumors are predicted and experimentally validated, enabling integration of metabolic features with other multi-omics datasets into ensemble-based machine learning classifiers for radiation response. These multi-omics classifiers show improved classification accuracy, identify clinical patient subgroups, and demonstrate the utility of personalized blood-based metabolic biomarkers for radiation sensitivity. The integration of machine learning with genome-scale metabolic modeling represents a significant methodological advancement for identifying prognostic metabolite biomarkers and predicting radiosensitivity for individual patients.
Collapse
|
14
|
Yu X, Huang M, Yang G. Long non‑coding RNA BANCR promotes proliferation, invasion and migration in esophageal squamous cell carcinoma cells via the Raf/MEK/ERK signaling pathway. Mol Med Rep 2021; 23:465. [PMID: 33880577 PMCID: PMC8097753 DOI: 10.3892/mmr.2021.12104] [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: 07/29/2020] [Accepted: 03/01/2021] [Indexed: 12/24/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a major histological type of esophageal cancer, identified as a leading cause of tumor-associated death worldwide. In addition, long non-coding RNA (lncRNA) BRAF-activated non-coding RNA (BANCR) expression is increased in the plasma of patients with ESCC, which can be reversed by tumor resection. Thus, the aim of the present study was to investigate the underlying mechanism of BANCR in ESCC progression. The relative mRNA expression of BANCR was determined via reverse transcription-quantitative PCR. The cell behaviors of Eca-109 cells were detected using Cell Counting Kit-8, colony formation, wound healing and Transwell chamber assays. Finally, the expression levels of proteins involved in the Raf/MEK/ERK signaling pathway and cell metastasis were analyzed with western blotting. The results revealed that lncRNA BANCR was highly expressed in ESCC cells compared with in normal esophageal cells. BANCR overexpression enhanced proliferation, migration and invasion of ESCC cells, and BANCR silencing exerted opposite effects. Moreover, BANCR overexpression induced activation of the Raf/MEK/ERK signaling pathway in ESCC cells. Notably, U0126, a specific MEK inhibitor, decreased MEK and ERK expression, and blocked the promotive effects of BANCR overexpression on the proliferation, migration and invasion of ESCC cells. Overall, lncRNA BANCR facilitated the proliferation, migration and invasion of ESCC cells via the Raf/MEK/ERK signaling pathway. Thus, lncRNA BANCR may be a promising target for inhibiting ESCC growth and metastasis.
Collapse
Affiliation(s)
- Xiaogang Yu
- Department of Gastroenterology and Hepatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Meng Huang
- Department of Radiology, Suining Municipal Hospital of TCM, Suining, Sichuan 629000, P.R. China
| | - Guodong Yang
- Department of Gastroenterology and Hepatology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
15
|
Cabrera-Licona A, Pérez-Añorve IX, Flores-Fortis M, Moral-Hernández OD, González-de la Rosa CH, Suárez-Sánchez R, Chávez-Saldaña M, Aréchaga-Ocampo E. Deciphering the epigenetic network in cancer radioresistance. Radiother Oncol 2021; 159:48-59. [PMID: 33741468 DOI: 10.1016/j.radonc.2021.03.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 02/15/2021] [Accepted: 03/09/2021] [Indexed: 12/16/2022]
Abstract
Radiotherapy, in addition to surgery and systemic chemotherapy, remains the core of the current clinical management of cancer. Radioresistance is one of the major causes of disease progression and mortality in cancer; therefore, it is a significant challenge in the treatment of locally advanced, recurrent and metastatic cancer. Epigenetic mechanisms that control hallmarks of cancer have a key role in the development of radiation resistance of cancer cells. Recent advances in DNA methylation, histone modification, chromatin remodeling and non-coding RNAs identified in the control of signal transduction pathways in cancer and cancer stem cells have provided even greater promise in the improvement of understanding cancer radioresistance. Many epigenetic drugs that target epigenetic enzymes revert the radioresistant phenotypes decreasing the possibility that resistant cancer cells will develop refractory tumors to radiotherapy. Epigenetic profiles identified as regulators of DNA damage repair, hypoxia, cell survival, apoptosis and invasion are determinants in the development of tumor radioresistance; hence, they also are promising in personalized medicine to develop novel targeted therapies or biomarkers to follow-up the effectiveness of radiotherapy. Now, it is clear that radiotherapy can influence a complex epigenetic network for transcriptional reprogramming, enabling the cells to adapt and avoid the effect of radiotherapy. This review aims to highlight the epigenetic modifications identified in cancer radioresistance and to discuss approaches to disable epigenetic networks to increase the sensitivity and specificity of radiotherapy.
Collapse
Affiliation(s)
- Ariana Cabrera-Licona
- Departamento de Ciencias Naturales, Unidad Cuajimalpa, Universidad Autonoma Metropolitana, Ciudad de Mexico, Mexico; Posgrado en Ciencias Naturales e Ingenieria, Unidad Cuajimalpa, Universidad Autonoma Metropolitana, Ciudad de Mexico, Mexico
| | - Isidro X Pérez-Añorve
- Departamento de Ciencias Naturales, Unidad Cuajimalpa, Universidad Autonoma Metropolitana, Ciudad de Mexico, Mexico
| | - Mauricio Flores-Fortis
- Departamento de Ciencias Naturales, Unidad Cuajimalpa, Universidad Autonoma Metropolitana, Ciudad de Mexico, Mexico; Posgrado en Ciencias Naturales e Ingenieria, Unidad Cuajimalpa, Universidad Autonoma Metropolitana, Ciudad de Mexico, Mexico
| | - Oscar Del Moral-Hernández
- Laboratorio de Virologia y Epigenetica del Cancer, Facultad de Ciencias Quimico Biologicas, Universidad Autonoma de Guerrero, Chilpancingo, Mexico
| | | | - Rocio Suárez-Sánchez
- Laboratorio de Medicina Genomica, Departamento de Genetica, Instituto Nacional de Rehabilitacion LGII, Ciudad de Mexico, Mexico
| | - Margarita Chávez-Saldaña
- Laboratorio de Biologia de la Reproduccion, Instituto Nacional de Pediatria, Ciudad de Mexico, Mexico
| | - Elena Aréchaga-Ocampo
- Departamento de Ciencias Naturales, Unidad Cuajimalpa, Universidad Autonoma Metropolitana, Ciudad de Mexico, Mexico.
| |
Collapse
|
16
|
Liu WH, Qiao HY, Xu J, Wang WQ, Wu YL, Wu X. LINC00473 contributes to the radioresistance of esophageal squamous cell carcinoma by regulating microRNA‑497‑5p and cell division cycle 25A. Int J Mol Med 2020; 46:571-582. [PMID: 32468021 PMCID: PMC7307861 DOI: 10.3892/ijmm.2020.4616] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 03/31/2020] [Indexed: 12/20/2022] Open
Abstract
Long non-coding RNA (lncRNA) LINC00473 plays a carcinogenic role in a variety of different tumor types. Nevertheless, the mechanisms through which LINC00473 regulates the radiosensitivity of esophageal squamous cell carcinoma (ESCC) cells remains elusive. In the present study, reverse transcription-quantitative PCR was used to quantify the expression of LINC00473, microRNA (miRNA/miR)-497-5p and cell division cycle 25A (CDC25A) in ESCC tissues. The association between LINC00473 expression and the clinicopathological characteristics of patients with ESCC was also assessed. Furthermore, Cell Counting kit-8 and colony formation assays were carried out to monitor the proliferation of ESCC cells exposed to X-ray radiation. A dual-luciferase reporter assay was also conducted to analyze the interaction between LINC00473 and miR-497-5p, as well as the interaction between CDC25A and miR-497-5p. The findings of the present study demonstrated that in ESCC tissues and cells, the expression levels of LINC00473 and CDC25A were significantly upregulated, while the expression of miR-497-5p was downregulated. The high expression level of LINC00473 was associated with a higher T stage, lymph node metastasis stage and a lower tumor differentiation grade in patients with ESCC. Following irradiation, transfection with miR-497-5p mimics reduced the promoting effect of LINC00473 overexpression on ESCC cell proliferation, and partially impeded the resistance of ESCC cells to X-ray radiation induced by LINC00473 overexpression. Moreover, transfection with miR-497-5p inhibitors partially alleviated the inhibitory effects of LINC00473 knockdown on cellular proliferation, and partly reversed the sensitivity of cells to X-ray irradiation induced by LINC00473 knockdown. Furthermore, it was confirmed that miR-497-5p was able to bind LINC00473 and the 3′-untranslated region of CDC25A. On the whole, the findings of the present study demonstrate that LINC00473 reduces the radiosensitivity of ESCC cells by modulating the miR-497-5p/CDC25A axis.
Collapse
Affiliation(s)
- Wei-Hua Liu
- Department of Radiology, The Third People's Hospital of Linyi, Linyi, Shandong 276023, P.R. China
| | - Han-Yong Qiao
- Department of Special Inspection, The Third People's Hospital of Linyi, Linyi, Shandong 276023, P.R. China
| | - Jian Xu
- Department of Radiology, The Third People's Hospital of Linyi, Linyi, Shandong 276023, P.R. China
| | - Wei-Qing Wang
- Department of Radiology, The Third People's Hospital of Linyi, Linyi, Shandong 276023, P.R. China
| | - Yi-Lei Wu
- Department of Oncology, The Third People's Hospital of Linyi, Linyi, Shandong 276023, P.R. China
| | - Xia Wu
- Department of Oncology, The Third People's Hospital of Linyi, Linyi, Shandong 276023, P.R. China
| |
Collapse
|
17
|
Oike T, Ohno T. Molecular mechanisms underlying radioresistance: data compiled from isogenic cell experiments. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:273. [PMID: 32355717 PMCID: PMC7186667 DOI: 10.21037/atm.2020.02.90] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Takahiro Oike
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Gunma, Japan.,Gunma University Heavy Ion Medical Center, Gunma, Japan
| | - Tatsuya Ohno
- Department of Radiation Oncology, Gunma University Graduate School of Medicine, Gunma, Japan.,Gunma University Heavy Ion Medical Center, Gunma, Japan
| |
Collapse
|
18
|
Liu J, Xue N, Guo Y, Niu K, Gao L, Zhang S, Gu H, Wang X, Zhao D, Fan R. CircRNA_100367 regulated the radiation sensitivity of esophageal squamous cell carcinomas through miR-217/Wnt3 pathway. Aging (Albany NY) 2019; 11:12412-12427. [PMID: 31851619 PMCID: PMC6949088 DOI: 10.18632/aging.102580] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 11/26/2019] [Indexed: 12/13/2022]
Abstract
Background: Circular RNAs (circRNAs) play important roles in regulating the radioresistance of esophageal squamous cell carcinoma (ESCC). This study aimed to determine the role of hsa_circRNA_100367 in regulating radioresistance of ESCC. Results: Higher expression and potency of endothelial to mesenchymal transformation (EMT) was found in radioresistant ESCC cells (KYSE-150R) than in ESCC cells (KYSE-150). Silencing circRNA_100367 inhibited the proliferation and migration of KYSE-150R cells, and decreased the expression of β-catenin (an important molecule in Wnt pathway) in KYSE-150R cells. Additionally, circRNA_100367 bound to miR-217, and miR-217 targeted Wnt3. Low Wnt3 expression was associated with the short survival time in patients with ESCC and Wnt3 knockdown inhibited the proliferation and migration of KYSE-150R cells. CircRNA_100367 enhanced the radioresistance of KYSE-150R cells through miR-217/Wnt3 pathway. In vivo, circRNA_100367 silence reduced the growth of KYSE-150R cells under radiation. Conclusion: Our results revealed that circRNA_100367 attenuated radioresistance of ESCC through miR-217/Wnt3 pathway. Methods: CircRNAs related with the radioresistance of ESCC were analyzed by hierarchical cluster analysis. The relationship between circRNA_100367 and miR-217, Wnt3 was detected by RNA immunoprecipitation (RIP), RNA pull-down and luciferase reporte assays. The proliferation and migration ESCC cells were detected by MTT, Transwell and colony formation assays.
Collapse
Affiliation(s)
- Junqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Erqi, Zhengzhou 450000, China
| | - Nannan Xue
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Erqi, Zhengzhou 450000, China
| | - Yuexin Guo
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Erqi, Zhengzhou 450000, China
| | - Kerun Niu
- Department of Molecular and Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Liang Gao
- Center of Experimental Orthopaedics, Saarland University Medical Center, Kirrberger Strasse, Homburg 66421, Germany
| | - Song Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Erqi, Zhengzhou 450000, China
| | - Hao Gu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Erqi, Zhengzhou 450000, China
| | - Xin Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Erqi, Zhengzhou 450000, China
| | - Di Zhao
- Endocrinology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Ruitai Fan
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Erqi, Zhengzhou 450000, China
| |
Collapse
|
19
|
Ye L, Lin C, Wang X, Li Q, Li Y, Wang M, Zhao Z, Wu X, Shi D, Xiao Y, Ren L, Jian Y, Yang M, Ou R, Deng G, Ouyang Y, Chen X, Li J, Song L. Epigenetic silencing of SALL2 confers tamoxifen resistance in breast cancer. EMBO Mol Med 2019; 11:e10638. [PMID: 31657150 PMCID: PMC6895605 DOI: 10.15252/emmm.201910638] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 09/19/2019] [Accepted: 09/24/2019] [Indexed: 12/21/2022] Open
Abstract
Resistance to tamoxifen is a clinically major challenge in breast cancer treatment. Although downregulation of estrogen receptor-alpha (ERα) is the dominant mechanism of tamoxifen resistance, the reason for ERα decrease during tamoxifen therapy remains elusive. Herein, we reported that Spalt-like transcription factor 2 (SALL2) expression was significantly reduced during tamoxifen therapy through transcription profiling analysis of 9 paired primary pre-tamoxifen-treated and relapsed tamoxifen-resistant breast cancer tissues. SALL2 transcriptionally upregulated ESR1 and PTEN through directly binding to the DNA promoters. By contrast, silencing SALL2 induced downregulation of ERα and PTEN and activated the Akt/mTOR signaling, resulting in estrogen-independent growth and tamoxifen resistance in ERα-positive breast cancer. Furthermore, hypermethylation of SALL2 promoter was found in tamoxifen-resistant breast cancer. Importantly, in vivo experiments showed that DNA methyltransferase inhibitor-mediated SALL2 restoration resensitized tamoxifen-resistant breast cancer to tamoxifen therapy. These findings shed light on the mechanism of SALL2 in regulation of ER and represent a potential clinical signature that can be used to categorize breast cancer patients who may benefit from co-therapy with tamoxifen and DNMT inhibitor.
Collapse
Affiliation(s)
- Liping Ye
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Chuyong Lin
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Xi Wang
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Qiji Li
- Department of Orthopaedic SurgeryThe Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenChina
| | - Yue Li
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Meng Wang
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Zekun Zhao
- Division of BiosciencesUniversity College LondonLondonUK
| | - Xianqiu Wu
- Clinical Experimental CenterDepartment of Pathology (Clinical Biobanks)Jiangmen Central HospitalAffiliated Jiangmen Hospital of Sun Yat‐sen UniversityJiangmenGuangdongChina
| | - Dongni Shi
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Yunyun Xiao
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Liangliang Ren
- Department of BiochemistryZhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| | - Yunting Jian
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Meisongzhu Yang
- Department of BiochemistryZhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory of Protein Modification and DegradationSchool of Basic Medical SciencesAffiliated Cancer Hospital & Institute of Guangzhou Medical UniversityGuangzhouChina
| | - Ruizhang Ou
- Department of PathologySchool of Basic Medical ScienceSouthern Medical UniversityGuangzhouChina
| | - Guangzheng Deng
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Ying Ouyang
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Xiangfu Chen
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Jun Li
- Department of BiochemistryZhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| | - Libing Song
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| |
Collapse
|
20
|
Sato K, Shimokawa T, Imai T. Difference in Acquired Radioresistance Induction Between Repeated Photon and Particle Irradiation. Front Oncol 2019; 9:1213. [PMID: 31799186 PMCID: PMC6863406 DOI: 10.3389/fonc.2019.01213] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/23/2019] [Indexed: 12/21/2022] Open
Abstract
In recent years, advanced radiation therapy techniques, including stereotactic body radiotherapy and carbon–ion radiotherapy, have progressed to such an extent that certain types of cancer can be treated with radiotherapy alone. The therapeutic outcomes are particularly promising for early stage lung cancer, with results matching those of surgical resection. Nevertheless, patients may still experience local tumor recurrence, which might be exacerbated by the acquisition of radioresistance after primary radiotherapy. Notwithstanding the risk of tumors acquiring radioresistance, secondary radiotherapy is increasingly used to treat recurrent tumors. In this context, it appears essential to comprehend the radiobiological effects of repeated photon and particle irradiation and their underlying cellular and molecular mechanisms in order to achieve the most favorable therapeutic outcome. However, to date, the mechanisms of acquisition of radioresistance in cancer cells have mainly been studied after repeated in vitro X-ray irradiation. By contrast, other critical aspects of radioresistance remain mostly unexplored, including the response to carbon-ion irradiation of X-ray radioresistant cancer cells, the mechanisms of acquisition of carbon-ion resistance, and the consequences of repeated in vivo X-ray or carbon-ion irradiation. In this review, we discuss the underlying mechanisms of acquisition of X-ray and carbon-ion resistance in cancer cells, as well as the phenotypic differences between X-ray and carbon-ion-resistant cancer cells, the biological implications of repeated in vivo X-ray or carbon-ion irradiation, and the main open questions in the field.
Collapse
Affiliation(s)
- Katsutoshi Sato
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, The Tisch Cancer Institute, New York, NY, United States
| | - Takashi Shimokawa
- Department of Accelerator and Medical Physics, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Sciences and Technology, Chiba, Japan
| | - Takashi Imai
- Medical Databank, Department of Radiation Medicine, QST Hospital, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| |
Collapse
|
21
|
Imai A, Mochizuki D, Misawa Y, Nakagawa T, Endo S, Mima M, Yamada S, Kawasaki H, Kanazawa T, Misawa K. SALL2 Is a Novel Prognostic Methylation Marker in Patients with Oral Squamous Carcinomas: Associations with SALL1 and SALL3 Methylation Status. DNA Cell Biol 2019; 38:678-687. [PMID: 31188017 DOI: 10.1089/dna.2018.4597] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Staging and pathological grading systems are convenient, but imperfect predictors of recurrence of head and neck squamous cell carcinoma. Therefore, to identify potential alternative prognostic markers, we investigated the methylation status of the promoter of Sal-like protein 2 (SALL2). SALL2 mRNA expression was absent in 8/9 (88.9%) University of Michigan squamous cell carcinoma cell lines, whereas two nonmalignant cell lines had stable expression. The normalized methylation value of SALL2 in cancer cell lines was significantly higher than in normal cell lines. SALL2 methylation found in 74 of 233 (31.8%) tumor specimens was correlated with the methylation status of both SALL1 and SALL3. SALL2 methylation was not associated with any difference in disease-free survival (DFS). Therefore, the presence of SALL2 methylation was statistically correlated with a decrease in DFS in patients with oral cancer (log-rank test, p = 0.032). Furthermore, it was associated with disease recurrence in 36.2% of oral cancer cases, with an odds ratio of 2.922 (95% confidence interval = 1.198-7.130; p = 0.018) by multivariate Cox proportional hazard regression analysis. This study suggests that cytosine-phosphate- guanosine (CpG) hypermethylation is a likely mechanism of SALL2 inactivation and supports the hypothesis that SALL2 could serve as an important clinical risk assessment.
Collapse
Affiliation(s)
- Atsushi Imai
- 1 Department of Otolaryngology/Head and Neck Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Daiki Mochizuki
- 1 Department of Otolaryngology/Head and Neck Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Yuki Misawa
- 1 Department of Otolaryngology/Head and Neck Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takuya Nakagawa
- 2 Department of Otorhinolaryngology/Head and Neck Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shiori Endo
- 1 Department of Otolaryngology/Head and Neck Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Masato Mima
- 1 Department of Otolaryngology/Head and Neck Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Satoshi Yamada
- 1 Department of Otolaryngology/Head and Neck Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hideya Kawasaki
- 3 Preeminent Medical Photonics Education and Research Center Institute for NanoSuit Research, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takeharu Kanazawa
- 4 Department of Otolaryngology, Tokyo Voice Center, International University of Health and Welfare, Tokyo, Japan
| | - Kiyoshi Misawa
- 1 Department of Otolaryngology/Head and Neck Surgery, Hamamatsu University School of Medicine, Hamamatsu, Japan
| |
Collapse
|
22
|
He W, Yan Q, Fu L, Han Y. A five-gene signature to predict the overall survival time of patients with esophageal squamous cell carcinoma. Oncol Lett 2019; 18:1381-1387. [PMID: 31423201 PMCID: PMC6607091 DOI: 10.3892/ol.2019.10449] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 05/15/2019] [Indexed: 01/06/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the six most commonly diagnosed tumor types in the Chinese population. Gene expression profiles help to predict the prognosis of patients with ESCC. Disease recurrence as the survival endpoint has been analyzed in the majority of previous studies; therefore, the aim of the present study was to construct a robust gene signature in order to determine the overall survival (OS) of patients with ESCC. The gene expression and clinical data of patients with ESCC were downloaded from The Cancer Genome Atlas (TCGA) database. Of the selected data (172 samples from surviving patients), 72 samples were randomly selected as modeling data, and verification was conducted using the entire dataset. Data from the Gene Expression Omnibus was analyzed simultaneously, and a venn diagram was constructed to determine the intersection between these two sets of results; a total of 97 genes were found to be associated with OS. Kyoto Encyclopedia of Genes and Genomes analysis demonstrated that these genes were primarily associated with specific pathways (Homo sapiens), including DNA replication, protein processing in endoplasmic reticulum and influenza A. A five-gene signature was identified with a robust likelihood-based survival modeling approach. Using regression coefficient modeling, a prognostic model consisting of the C-X-C motif chemokine ligand 8, DNA damage inducible transcript 3, RAB27A, member RAS oncogene family, replication factor C subunit 2 and elongation factor for RNA polymerase II 2 genes was constructed and validated. Based on these results, patients were subdivided into high and low-risk groups. Compared with the high-risk group, the OS time of patients in the low-risk group was significantly increased. Furthermore, it was determined that the five genes were all differentially expressed in ESCC tissues compared with normal tissues, indicating the potential role of these genes in ESCC initiation and progression. In another independent cohort, this five-gene signature was further confirmed and was considered as an independent prognostic biomarker for OS prediction in patients with ESCC. In conclusion, the OS of patients with ESCC may be predicted using this five-gene signature, which may be useful in identifying patients with high-risk ESCC.
Collapse
Affiliation(s)
- Wenwu He
- Department of Thoracic Surgery, Sichuan Cancer Hospital and Research Institute, Chengdu, Sichuan 610041, P.R. China
| | - Qunlun Yan
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan 637007, P.R. China
| | - Liangmin Fu
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan 637007, P.R. China
| | - Yongtao Han
- Department of Thoracic Surgery, Sichuan Cancer Hospital and Research Institute, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
23
|
Sadeghpour S, Ghorbian S. Evaluation of the potential clinical prognostic value of lncRNA-BANCR gene in esophageal squamous cell carcinoma. Mol Biol Rep 2019; 46:991-995. [PMID: 30552615 DOI: 10.1007/s11033-018-4556-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 12/04/2018] [Indexed: 01/28/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is the seventh most common cause of cancer death in worldwide. LncRNA-BANCR is a long non-coding RNA (lncRNA), which has made new windows in cancer investigations. The aim of this survey was to determine the lncRNA-BANCR gene expression changes in patients with ESCC. In case-control investigation was performed on 150 formalin fixed-paraffin embedded tissues (75 cancerous and 75 non-cancerous tissues) of ESCC patients. The lncRNA-BANCR gene expression alteration was assessed by Real-Time PCR technique. Our findings revealed that lncRNA-BANCR gene expression was increased significantly in tumor tissues compared with the non-cancerous tissues (p = 0.0025). In addition, lncRNA-BANCR gene expression changes was positively associated with the lymph node metastasis (p = 0.013), tumor differentiation (p = 0.019) and tumor stage (p = 0.017). Our results suggest a possible role of lncRNA-BANCR in proliferation of esophageal tissues and may be considered as a potential prognostic value for ESCC metastasis.
Collapse
Affiliation(s)
- Shiva Sadeghpour
- Department of Molecular Genetics, Ahar Branch, Islamic Azad University, Ahar, Iran
| | - Saeid Ghorbian
- Department of Molecular Genetics, Ahar Branch, Islamic Azad University, Ahar, Iran.
| |
Collapse
|
24
|
Gao P, Wang X, Jin Y, Hu W, Duan Y, Shi A, Du Y, Song D, Yang M, Li S, Han B, Zhao G, Zhang H, Fan Z, Miao QR. Nogo-B receptor increases the resistance to tamoxifen in estrogen receptor-positive breast cancer cells. Breast Cancer Res 2018; 20:112. [PMID: 30208932 PMCID: PMC6134690 DOI: 10.1186/s13058-018-1028-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 07/19/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUNDS Tamoxifen is typically used to treat patients with estrogen receptor alpha (ERα)-positive breast cancer. However, 30% of these patients gain acquired resistance to tamoxifen during or after tamoxifen treatment. As a Ras modulator, Nogo-B receptor (NgBR) is required for tumorigenesis through the signaling crosstalk with epidermal growth factor (EGF) receptor (EGFR)-mediated pathways. NgBR is highly expressed in many types of cancer cells and regulates the sensitivity of hepatocellular carcinoma to chemotherapy. In this study, we found the expression of NgBR is increased in tamoxifen-resistant ERα-positive breast cancer cells. METHODS Tamoxifen-resistant ERα-positive MCF-7 and T47D breast cancer cell lines were established by culturing with gradually increased concentration of 4-hydroxytamoxifen (4-OHT). The effects of NgBR on tamoxifen resistance was determined by depleting NgBR in these cell lines using previously validated small interfering RNA (siRNA). The effects of 4-OHT on cell viability and apoptosis were determined using well-accepted methods such as clonogenic survival assay and Annexin V/propidium iodide staining. The alteration of EGF-stimulated signaling and gene expression was determined by western blot analysis and real-time PCR, respectively. RESULTS NgBR knockdown with siRNA attenuates EGF-induced phosphorylation of ERα and restores the sensitivity to tamoxifen in ERα-positive breast cancer cells. Mechanistically, our data demonstrated that NgBR knockdown increases the protein levels of p53 and decreases survivin, which is an apoptosis inhibitor. CONCLUSIONS These results suggested that NgBR is a potential therapeutic target for increasing the sensitivity of ERα-positive breast cancer to tamoxifen.
Collapse
Affiliation(s)
- Pin Gao
- Department of Breast Surgery, The First Hospital of Jilin University, 71 Xinmin street, Changchun, 130021 Jilin Province China
- Division of Pediatric Surgery, Department of Surgery, Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226 USA
- Division of Pediatric Pathology, Department of Pathology, Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226 USA
| | - Xiang Wang
- Division of Pediatric Surgery, Department of Surgery, Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226 USA
- Division of Pediatric Pathology, Department of Pathology, Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226 USA
- Department of Human Anatomy, Histology, and Embryology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, 100191 China
| | - Ying Jin
- Department of Breast Surgery, The First Hospital of Jilin University, 71 Xinmin street, Changchun, 130021 Jilin Province China
- Division of Pediatric Surgery, Department of Surgery, Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226 USA
- Division of Pediatric Pathology, Department of Pathology, Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226 USA
| | - Wenquan Hu
- Division of Pediatric Surgery, Department of Surgery, Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226 USA
- Division of Pediatric Pathology, Department of Pathology, Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226 USA
| | - Yajun Duan
- Division of Pediatric Surgery, Department of Surgery, Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226 USA
- Division of Pediatric Pathology, Department of Pathology, Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226 USA
- Department of Human Anatomy, Histology, and Embryology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, 100191 China
| | - Aiping Shi
- Department of Breast Surgery, The First Hospital of Jilin University, 71 Xinmin street, Changchun, 130021 Jilin Province China
| | - Ye Du
- Department of Breast Surgery, The First Hospital of Jilin University, 71 Xinmin street, Changchun, 130021 Jilin Province China
| | - Dong Song
- Department of Breast Surgery, The First Hospital of Jilin University, 71 Xinmin street, Changchun, 130021 Jilin Province China
| | - Ming Yang
- Department of Breast Surgery, The First Hospital of Jilin University, 71 Xinmin street, Changchun, 130021 Jilin Province China
| | - Sijie Li
- Department of Breast Surgery, The First Hospital of Jilin University, 71 Xinmin street, Changchun, 130021 Jilin Province China
| | - Bing Han
- Department of Breast Surgery, The First Hospital of Jilin University, 71 Xinmin street, Changchun, 130021 Jilin Province China
| | - Gang Zhao
- Department of Breast Surgery, The First Hospital of Jilin University, 71 Xinmin street, Changchun, 130021 Jilin Province China
| | - Hongquan Zhang
- Department of Human Anatomy, Histology, and Embryology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) and State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing, 100191 China
- College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071 China
| | - Zhimin Fan
- Department of Breast Surgery, The First Hospital of Jilin University, 71 Xinmin street, Changchun, 130021 Jilin Province China
- College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071 China
| | - Qing Robert Miao
- Division of Pediatric Surgery, Department of Surgery, Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226 USA
- Division of Pediatric Pathology, Department of Pathology, Children’s Research Institute, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Milwaukee, WI 53226 USA
- College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071 China
| |
Collapse
|
25
|
Hepp MI, Escobar D, Farkas C, Hermosilla VE, Álvarez C, Amigo R, Gutiérrez JL, Castro AF, Pincheira R. A Trichostatin A (TSA)/Sp1-mediated mechanism for the regulation of SALL2 tumor suppressor in Jurkat T cells. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2018; 1861:S1874-9399(18)30028-2. [PMID: 29778644 DOI: 10.1016/j.bbagrm.2018.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 05/03/2018] [Accepted: 05/16/2018] [Indexed: 11/26/2022]
Abstract
SALL2 is a transcription factor involved in development and disease. Deregulation of SALL2 has been associated with cancer, suggesting that it plays a role in the disease. However, how SALL2 is regulated and why is deregulated in cancer remain poorly understood. We previously showed that the p53 tumor suppressor represses SALL2 under acute genotoxic stress. Here, we investigated the effect of Histone Deacetylase Inhibitor (HDACi) Trichostatin A (TSA), and involvement of Sp1 on expression and function of SALL2 in Jurkat T cells. We show that SALL2 mRNA and protein levels were enhanced under TSA treatment. Both, TSA and ectopic expression of Sp1 transactivated the SALL2 P2 promoter. This transactivation effect was blocked by the Sp1-binding inhibitor mithramycin A. Sp1 bound in vitro and in vivo to the proximal region of the P2 promoter. TSA induced Sp1 binding to the P2 promoter, which correlated with dynamic changes on H4 acetylation and concomitant recruitment of p300 or HDAC1 in a mutually exclusive manner. Our results suggest that TSA-induced Sp1-Lys703 acetylation contributes to the transcriptional activation of the P2 promoter. Finally, using a CRISPR/Cas9 SALL2-KO Jurkat-T cell model and gain of function experiments, we demonstrated that SALL2 upregulation is required for TSA-mediated cell death. Thus, our study identified Sp1 as a novel transcriptional regulator of SALL2, and proposes a novel epigenetic mechanism for SALL2 regulation in Jurkat-T cells. Altogether, our data support SALL2 function as a tumor suppressor, and SALL2 involvement in cell death response to HDACi.
Collapse
Affiliation(s)
- Matías I Hepp
- Departamento de Bioquímica y Biología Molecular, Facultad Cs. Biológicas, Universidad de Concepción, Chile.
| | - David Escobar
- Departamento de Bioquímica y Biología Molecular, Facultad Cs. Biológicas, Universidad de Concepción, Chile
| | - Carlos Farkas
- Departamento de Bioquímica y Biología Molecular, Facultad Cs. Biológicas, Universidad de Concepción, Chile
| | - Viviana E Hermosilla
- Departamento de Bioquímica y Biología Molecular, Facultad Cs. Biológicas, Universidad de Concepción, Chile
| | - Claudia Álvarez
- Departamento de Bioquímica y Biología Molecular, Facultad Cs. Biológicas, Universidad de Concepción, Chile
| | - Roberto Amigo
- Departamento de Bioquímica y Biología Molecular, Facultad Cs. Biológicas, Universidad de Concepción, Chile
| | - José L Gutiérrez
- Departamento de Bioquímica y Biología Molecular, Facultad Cs. Biológicas, Universidad de Concepción, Chile
| | - Ariel F Castro
- Departamento de Bioquímica y Biología Molecular, Facultad Cs. Biológicas, Universidad de Concepción, Chile
| | - Roxana Pincheira
- Departamento de Bioquímica y Biología Molecular, Facultad Cs. Biológicas, Universidad de Concepción, Chile.
| |
Collapse
|
26
|
Misawa K, Misawa Y, Imai A, Mochizuki D, Endo S, Mima M, Ishikawa R, Kawasaki H, Yamatodani T, Kanazawa T. Epigenetic modification of SALL1 as a novel biomarker for the prognosis of early stage head and neck cancer. J Cancer 2018; 9:941-949. [PMID: 29581773 PMCID: PMC5868161 DOI: 10.7150/jca.23527] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 01/31/2018] [Indexed: 12/12/2022] Open
Abstract
This study examined Sal-like protein (SALL)1 methylation profiles in head and neck squamous-cell carcinoma (HNSCC) patients at diagnosis and follow-up, and evaluated their prognostic significance and value as a biomarker. SALL1 expression was examined in a panel of cell lines by quantitative reverse transcription PCR (qRT-PCR). Promoter methylation was determined by quantitative methylation-specific polymerase chain reaction (qMSP) and was compared to the clinical characteristics of 205 samples. SALL1 promoter methylation was associated with transcriptional inhibition and was correlated with disease recurrence in 31.7% of cases, with an odds ratio of 1.694 (95% confidence interval: 1.093-2.626; P = 0.018) by multivariate Cox proportional hazard regression analysis. SALL1 promoter hypermethylation showed highly discriminatory receiver operator characteristic curve profiles that clearly distinguished HNSCC from adjacent normal mucosal tissue, and was correlated with reduced disease-free survival in early stage T1 and T2 patients (log-rank test, P < 0.001). SALL1 methylation was significantly correlated with the methylation status of both SALL3 and CDH1. This study suggests that CpG hypermethylation is a likely mechanism of SALL1 gene inactivation, supporting the hypothesis that SALL1 might play a role in HNSCC tumorigenesis and could serve as an important biomarker.
Collapse
Affiliation(s)
- Kiyoshi Misawa
- Department of Otolaryngology/Head and Neck Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Yuki Misawa
- Department of Otolaryngology/Head and Neck Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Atsushi Imai
- Department of Otolaryngology/Head and Neck Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Daiki Mochizuki
- Department of Otolaryngology/Head and Neck Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Shiori Endo
- Department of Otolaryngology/Head and Neck Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Masato Mima
- Department of Otolaryngology/Head and Neck Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Ryuji Ishikawa
- Department of Otolaryngology/Head and Neck Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Hideya Kawasaki
- Department of Regenerative & Infectious Pathology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takashi Yamatodani
- Department of Otolaryngology/Head and Neck Surgery, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Takeharu Kanazawa
- Department of Otolaryngology/Head and Neck Surgery, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
27
|
Ou Y, Zhang Q, Tang Y, Lu Z, Lu X, Zhou X, Liu C. DNA methylation enzyme inhibitor RG108 suppresses the radioresistance of esophageal cancer. Oncol Rep 2018; 39:993-1002. [PMID: 29328411 PMCID: PMC5802040 DOI: 10.3892/or.2018.6210] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/15/2017] [Indexed: 01/28/2023] Open
Abstract
Esophageal cancer (EC) is the eighth most common highly aggressive cancer worldwide. The purpose of this study was to investigate the effect of the DNA methyltransferase inhibitor RG108 on the radiosensitivity of EC cells. MTT and clonogenic assays were performed to assess the effect of RG108 on the proliferation and radiosensitivity of Eca-109 and TE-1 human EC cells. The cell cycle progression and alterations in apoptosis were analyzed by flow cytometry. For the in vivo analysis, the Eca-109 cells were inoculated into nude mice to establish tumors. Tissues from xenografts were obtained to detect changes to microvessels and tumor growth by immunohistochemistry (IHC). RNA-seq was used to identify differentially expressed genes. We found that RG108 increased the radiosensitivity of EC cells. Apoptosis and G2/M-phase arrest were induced by X-ray irradiation and were significantly enhanced by RG108. In addition, growth of tumor xenografts from the Eca-109 cells was significantly inhibited by irradiation in combination with RG108. The RNA-seq analysis revealed that, compared with radiation alone, X-ray irradiation in combination with RG108 altered the expression of 121 genes in multiple pathways, including the TGF-β signaling pathway and the Epstein-Barr virus infection pathway. In conclusion, RG108 induced radiosensitivity in EC cells both in vitro and in vivo.
Collapse
Affiliation(s)
- Yao Ou
- Department of Radiotherapy, Changzhou Tumor Hospital, Soochow University, Changzhou, Jiangsu 213001, P.R. China
| | - Quan Zhang
- Department of Radiotherapy, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Yiting Tang
- Department of Radiotherapy, Changzhou Tumor Hospital, Soochow University, Changzhou, Jiangsu 213001, P.R. China
| | - Zhonghua Lu
- Department of Radiotherapy, Changzhou Tumor Hospital, Soochow University, Changzhou, Jiangsu 213001, P.R. China
| | - Xujing Lu
- Department of Radiotherapy, Changzhou Tumor Hospital, Soochow University, Changzhou, Jiangsu 213001, P.R. China
| | - Xifa Zhou
- Department of Radiotherapy, Changzhou Tumor Hospital, Soochow University, Changzhou, Jiangsu 213001, P.R. China
| | - Changmin Liu
- Department of Oncology, The Affiliated Hospital of Binzhou Medical College, Binzhou, Shandong 256603, P.R. China
| |
Collapse
|
28
|
Microarray analyses reveal genes related to progression and prognosis of esophageal squamous cell carcinoma. Oncotarget 2017; 8:78838-78850. [PMID: 29108269 PMCID: PMC5668002 DOI: 10.18632/oncotarget.20232] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 07/13/2017] [Indexed: 01/08/2023] Open
Abstract
Esophageal squamous cell carcinoma is a high morbidity and mortality cancer in China. Here are few biomarkers and therapeutic targets. Our study was aimed to identify candidate genes correlated to ESCC. Oncomine, The Cancer Genome Atlas, Gene Expression Omnibus were retrieved for eligible ESCC data. Deregulated genes were identified by meta-analysis and validated by an independent dataset. Survival analyses and bioinformatics analyses were used to explore potential mechanisms. Copy number variant analyses identified upstream mechanisms of candidate genes. In our study, top 200 up/down-regulated genes were identified across two microarrays. A total of 139 different expression genes were validated in GSE53625. Survival analysis found that nine genes were closely related to prognosis. Furthermore, Gene Ontology analyses and Kyoto Encyclopedia of Genes and Genomes analyses showed that different expression genes were mainly enriched in cell division, cell cycle and cell-cell adhesion pathways. Copy number variant analyses indicated that overexpression of ECT2 and other five genes were correlated with copy number amplification. The current study demonstrated that ECT2 and other eight candidate genes were correlated to progression and prognosis of esophageal squamous cell carcinoma, which might provide novel insights to the mechanisms.
Collapse
|