1
|
Naik A, Lattab B, Qasem H, Decock J. Cancer testis antigens: Emerging therapeutic targets leveraging genomic instability in cancer. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200768. [PMID: 38596293 PMCID: PMC10876628 DOI: 10.1016/j.omton.2024.200768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/11/2024]
Abstract
Cancer care has witnessed remarkable progress in recent decades, with a wide array of targeted therapies and immune-based interventions being added to the traditional treatment options such as surgery, chemotherapy, and radiotherapy. However, despite these advancements, the challenge of achieving high tumor specificity while minimizing adverse side effects continues to dictate the benefit-risk balance of cancer therapy, guiding clinical decision making. As such, the targeting of cancer testis antigens (CTAs) offers exciting new opportunities for therapeutic intervention of cancer since they display highly tumor specific expression patterns, natural immunogenicity and play pivotal roles in various biological processes that are critical for tumor cellular fitness. In this review, we delve deeper into how CTAs contribute to the regulation and maintenance of genomic integrity in cancer, and how these mechanisms can be exploited to specifically target and eradicate tumor cells. We review the current clinical trials targeting aforementioned CTAs, highlight promising pre-clinical data and discuss current challenges and future perspectives for future development of CTA-based strategies that exploit tumor genomic instability.
Collapse
Affiliation(s)
- Adviti Naik
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Boucif Lattab
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Hanan Qasem
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
- College of Health and Life Sciences (CHLS), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | - Julie Decock
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
- College of Health and Life Sciences (CHLS), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Doha, Qatar
| |
Collapse
|
2
|
Xie X, Lin H, Zhang X, Song P, He X, Zhong J, Shi J. Overexpression of GDP dissociation inhibitor 1 gene associates with the invasiveness and poor outcomes of colorectal cancer. Bioengineered 2021; 12:5595-5606. [PMID: 34515625 PMCID: PMC8806759 DOI: 10.1080/21655979.2021.1967031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 12/24/2022] Open
Abstract
GDP dissociation inhibitor (GDI) regulates the GDP/GTP exchange reaction of most Rab proteins by inhibiting GDP dissociation. This study evaluated the potential prognostic and predictive value of GDI1 in colorectal cancer (CRC). To address the prognostic power of GDI1, we performed individual and pooled survival analyses on six independent CRC microarray gene expression datasets. GDI1-enriched signatures were also analyzed. Kaplan-Meier and Cox proportional analyses were employed for survival analysis. An immunohistochemistry (IHC) analysis was performed to validate the clinical relevance and prognostic significance of the GDI1 protein level in CRC tissue samples. The results revealed that GDI1 mRNA level was significantly linked with the aggressiveness of CRC, which is compatible with gene set enrichment analysis. A meta-analysis and pooled analysis demonstrated that a higher mRNA GDI1 expression was dramatically correlated with a worse survival in a dose-dependent manner in CRC patients. Further IHC analysis validated that the protein expression of GDI1 in both cytoplasm and membrane also significantly impacted the outcome of CRC patients. In CRC patients with stage III, chemotherapy significantly reduced the relative risk of death in low-GDI1 subgroup (hazard ratio (HR) = 0.22; 95% confidence interval (95% CI) 0.09-0.56, p = 0.0003), but not in high-GDI1 subgroup (HR = 0.63; 95% CI 0.35-1.14, p = 0.1137). Therefore, both high mRNA and protein levels of GDI1 were significantly related to poor outcomes in CRC patients. GD11 may serve as a prognostic biomarker for CRC.
Collapse
Affiliation(s)
- Xiao Xie
- Department of Gastroenterology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Affiliated Huzhou Hospital Zhejiang University School of Medicine, Huzhou, Zhejiang Province, China
| | - Huajiang Lin
- Department of Gastroenterology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Affiliated Huzhou Hospital Zhejiang University School of Medicine, Huzhou, Zhejiang Province, China
| | - Xiaolei Zhang
- Department of Gastroenterology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Affiliated Huzhou Hospital Zhejiang University School of Medicine, Huzhou, Zhejiang Province, China
| | - Pengtao Song
- Department of Pathology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Affiliated Huzhou Hospital Zhejiang University School of Medicine, Huzhou, Zhejiang Province, China
| | - Xiangyi He
- Department of Pathology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Affiliated Huzhou Hospital Zhejiang University School of Medicine, Huzhou, Zhejiang Province, China
| | - Jing Zhong
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Affiliated Huzhou Hospital Zhejiang University School of Medicine, Huzhou, Zhejiang Province, China
| | - Jiemin Shi
- Department of Gastroenterology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Affiliated Huzhou Hospital Zhejiang University School of Medicine, Huzhou, Zhejiang Province, China
| |
Collapse
|
3
|
Huang J, Wang Y, Liu J, Chu M, Wang Y. TFDP3 as E2F Unique Partner, Has Crucial Roles in Cancer Cells and Testis. Front Oncol 2021; 11:742462. [PMID: 34745961 PMCID: PMC8564135 DOI: 10.3389/fonc.2021.742462] [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: 07/16/2021] [Accepted: 09/30/2021] [Indexed: 12/03/2022] Open
Abstract
Transcription factor DP family member 3 (TFDP3) is a cancer-testis antigen, mainly expressed in normal testis and multiple cancers. TFDP3 gene (Gene ID: 51270) is located on the chromosome X and shares a high degree of sequence homology with TFDP1 and TFDP2, which can form heterodimers with E2F family members and enhance DNA-binding activity of E2Fs. In contrast to TFDP1 and TFDP2, TFDP3 downregulates E2F-mediated transcriptional activation. During DNA damage response in cancer cells, TFDP3 is induced and can inhibit E2F1-mediated apoptosis. Moreover, TFDP3 is involved in cell autophagy and epithelial-mesenchymal transition. Regarding cancer therapy opportunity, the transduction of dendritic cells with recombinant adenovirus-encoding TFDP3 can activate autologous cytotoxic T lymphocytes to target hepatoma cells. Here, we review the characterization of TFDP3, with an emphasis on the biological function and molecular mechanism. A better understanding of TFDP3 will provide new insights into the pathological mechanisms and therapeutic strategies for cancers.
Collapse
Affiliation(s)
- Jiahao Huang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Yini Wang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Jinlong Liu
- Department of Basic Medicine and Forensic Medicine, Baotou Medical College, Baotou, China
| | - Ming Chu
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Yuedan Wang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| |
Collapse
|
4
|
Tang Y, Jiang L, Zhao X, Hu D, Zhao G, Luo S, Du X, Tang W. FOXO1 inhibits prostate cancer cell proliferation via suppressing E2F1 activated NPRL2 expression. Cell Biol Int 2021; 45:2510-2520. [PMID: 34459063 DOI: 10.1002/cbin.11696] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 08/14/2021] [Accepted: 08/28/2021] [Indexed: 11/11/2022]
Abstract
Previous studies in our lab suggest that nitrogen permease regulator 2-like (NPRL2) upregulation in prostate cancer is associated with malignant behavior and poor prognosis. However, the underlying mechanisms of NPRL2 dysregulation remain poorly understood. This study aimed to explore the transcription factors (TFs) contributing to NPRL2 dysregulation in prostate cancer. Potential TFs were identified using prostate tissue/cell-specific chromatin immunoprecipitation (ChIP)-seq data collected in the Cistrome Data Browser and Signaling Pathways Project. Dual-luciferase assay and ChIP-qPCR assay were conducted to assess the binding and activating effect of TFs on the gene promoter. Cell Counting Kit-8 and colony formation assays were performed to assess cell proliferation. Results showed that E2F1 is a TF that bound to the NPRL2 promoter and activated its transcription. NPRL2 inhibition significantly alleviated E2F1 enhanced cell proliferation. Kaplan-Meier survival analysis indicated that E2F1 upregulation was associated with unfavorable progression-free survival and disease-specific survival. FOXO1 interacted and E2F1 in both PC3 and LNCaP cells and weakened the binding of E2F1 to the NPRL2 promoter. Functionally, FOXO1 overexpression significantly slowed the proliferation of PC3 and LNCaP cells and also decreased E2F1 enhanced cell proliferation. In summary, this study revealed a novel FOXO1/E2F1-NPRL2 regulatory axis in prostate cancer. E2F1 binds to the NPRL2 promoter and activates its transcription, while FOXO1 interacts with E2F1 and weakens its transcriptional activating effects. These findings help expand our understanding of the prostate cancer etiology and suggest that the FOXO1/E2F1-NPRL2 signaling axis might be a potential target.
Collapse
Affiliation(s)
- Yu Tang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Jiang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Zhao
- Department of Urology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Daixing Hu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guozhi Zhao
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shengjun Luo
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoyi Du
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Tang
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
Hurtado López AM, Chen-Liang TH, Zurdo M, Carrillo-Tornel S, Panadero J, Salido EJ, Beltrán V, Muiña B, Amigo M, Navarro-Villamor N, Cifuentes R, Calabria I, Antón AI, Teruel R, Muro M, Vicente V, Jerez A. Cancer testis antigens in myelodysplastic syndromes revisited: a targeted RNA-seq approach. Oncoimmunology 2020; 9:1824642. [PMID: 33101773 PMCID: PMC7553508 DOI: 10.1080/2162402x.2020.1824642] [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] [Indexed: 11/21/2022] Open
Abstract
Cancer-Testis antigens (CTA) are named after the tissues where they are mainly expressed: in germinal and in cancer cells, a process that mimics many gametogenesis features. Mapping accurately the CTA gene expression signature in myelodysplastic syndromes (MDS) and chronic myelomonocytic leukemia (CMML) is a prerequisite for downstream immune target-discovery projects. In this study, we take advantage of the use of azacitidine to treat high-risk MDS and CMML to draw the CTAs landscape, before and after treatment, using an ad hoc targeted RNA sequencing (RNA-seq) design for this group of low transcript genes. In 19 patients, 196 CTAs were detected at baseline. Azacitidine did not change the number of CTAs expressed, but it significantly increased or decreased expression in nine and five CTAs, respectively. TFDP3 and DDX53, emerged as the main candidates for immunotherapeutic targeting, as they showed three main features: i) a significant derepression on day +28 of cycle one in those patients who achieved complete remission with hypomethylating treatment (FC = 6, p = .008; FC = 2.1, p = .008, respectively), ii) similar dynamics at the protein level to what was observed at the RNA layer, and iii) to elicit significant specific cytotoxic immune responses detected by TFDP3 and DDX53 HLA-A*0201 tetramers. Our study addresses the unmet landscape of CTAs expression in MDS and CMML and revealed a previously unrecognized TFDP3 and DDX53 reactivation, detectable in plasma and able to elicit a specific immune response after one cycle of azacitidine.
Collapse
Affiliation(s)
- Ana María Hurtado López
- Hematology and Medical Oncology Department, Hospital Universitario Morales Meseguer, IMIB, Murcia, Spain
| | - Tzu Hua Chen-Liang
- Hematology and Medical Oncology Department, Hospital Universitario Morales Meseguer, IMIB, Murcia, Spain
| | - María Zurdo
- Hematology and Medical Oncology Department, Hospital Universitario Morales Meseguer, IMIB, Murcia, Spain
| | - Salvador Carrillo-Tornel
- Hematology and Medical Oncology Department, Hospital Universitario Morales Meseguer, IMIB, Murcia, Spain
| | | | - Eduardo José Salido
- Department of Hematology, Virgen De La Arrixaca University Hospital, Murcia, Spain
| | | | - Begoña Muiña
- Hematology Unit, Hospital Rafael Méndez, Lorca, Spain
| | - MariLuz Amigo
- Hematology and Medical Oncology Department, Hospital Universitario Morales Meseguer, IMIB, Murcia, Spain
| | | | - Rosa Cifuentes
- Hematology and Medical Oncology Department, Hospital Universitario Morales Meseguer, IMIB, Murcia, Spain
| | - Inés Calabria
- Genomics Unit, Health Research Institute La Fe, Valencia, Spain
| | | | - Raúl Teruel
- Hematology and Medical Oncology Department, Hospital Universitario Morales Meseguer, IMIB, Murcia, Spain
| | - Manuel Muro
- Immunology Department, Hospital Clínico Universitario Virgen De La Arrixaca, Murcia, Spain
| | - Vicente Vicente
- Hematology and Medical Oncology Department, Hospital Universitario Morales Meseguer, IMIB, Murcia, Spain
| | - Andrés Jerez
- Hematology and Medical Oncology Department, Hospital Universitario Morales Meseguer, IMIB, Murcia, Spain.,CB15/00055-CIBERER, Murcia, Spain
| |
Collapse
|
6
|
Jin D, Jiao Y, Ji J, Jiang W, Ni W, Wu Y, Ni R, Lu C, Qu L, Ni H, Liu J, Xu W, Xiao M. Identification of prognostic risk factors for pancreatic cancer using bioinformatics analysis. PeerJ 2020; 8:e9301. [PMID: 32587798 PMCID: PMC7301898 DOI: 10.7717/peerj.9301] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 05/15/2020] [Indexed: 12/11/2022] Open
Abstract
Background Pancreatic cancer is one of the most common malignant cancers worldwide. Currently, the pathogenesis of pancreatic cancer remains unclear; thus, it is necessary to explore its precise molecular mechanisms. Methods To identify candidate genes involved in the tumorigenesis and proliferation of pancreatic cancer, the microarray datasets GSE32676, GSE15471 and GSE71989 were downloaded from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) between Pancreatic ductal adenocarcinoma (PDAC) and nonmalignant samples were screened by GEO2R. The Database for Annotation Visualization and Integrated Discovery (DAVID) online tool was used to obtain a synthetic set of functional annotation information for the DEGs. A PPI network of the DEGs was established using the Search Tool for the Retrieval of Interacting Genes (STRING) database, and a combination of more than 0.4 was considered statistically significant for the PPI. Subsequently, we visualized the PPI network using Cytoscape. Functional module analysis was then performed using Molecular Complex Detection (MCODE). Genes with a degree ≥10 were chosen as hub genes, and pathways of the hub genes were visualized using ClueGO and CluePedia. Additionally, GenCLiP 2.0 was used to explore interactions of hub genes. The Literature Mining Gene Networks module was applied to explore the cocitation of hub genes. The Cytoscape plugin iRegulon was employed to analyze transcription factors regulating the hub genes. Furthermore, the expression levels of the 13 hub genes in pancreatic cancer tissues and normal samples were validated using the Gene Expression Profiling Interactive Analysis (GEPIA) platform. Moreover, overall survival and disease-free survival analyses according to the expression of hub genes were performed using Kaplan-Meier curve analysis in the cBioPortal online platform. The relationship between expression level and tumor grade was analyzed using the online database Oncomine. Lastly, the eight snap-frozen tumorous and adjacent noncancerous adjacent tissues of pancreatic cancer patients used to detect the CDK1 and CEP55 protein levels by western blot. Conclusions Altogether, the DEGs and hub genes identified in this work can help uncover the molecular mechanisms underlying the tumorigenesis of pancreatic cancer and provide potential targets for the diagnosis and treatment of this disease.
Collapse
Affiliation(s)
- Dandan Jin
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China.,Clinical Medicine, Medical College, Nantong University, Nantong, China
| | - Yujie Jiao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China.,Clinical Medicine, Medical College, Nantong University, Nantong, China
| | - Jie Ji
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China.,Clinical Medicine, Medical College, Nantong University, Nantong, China
| | - Wei Jiang
- Department of Emergency, Affiliated Hospital of Nantong University, Nantong, China
| | - Wenkai Ni
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yingcheng Wu
- Clinical Medicine, Medical College, Nantong University, Nantong, China
| | - Runzhou Ni
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
| | - Cuihua Lu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
| | - Lishuai Qu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
| | - Hongbing Ni
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
| | - Jinxia Liu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
| | - Weisong Xu
- Department of Gastroenterology, Second People's Hospital of Nantong, Nantong, China
| | - MingBing Xiao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
7
|
Identification and Validation of an Individualized EMT-Related Prognostic Risk Score Formula in Gastric Adenocarcinoma Patients. BIOMED RESEARCH INTERNATIONAL 2020; 2020:7082408. [PMID: 32309437 PMCID: PMC7142392 DOI: 10.1155/2020/7082408] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/30/2020] [Accepted: 02/03/2020] [Indexed: 02/07/2023]
Abstract
Background The epithelial-mesenchymal transition (EMT) is a pivotal process for fibrotic disease, embryonic development, and wound healing. Moreover, some evidence has proven that the disorder of EMT also plays an important role in carcinogenesis, especially invasion and metastasis of various tumors (Ritchie et al., 2015). Additionally, gastric adenocarcinoma (GAC) is a common gastrointestinal malignancy which is the fourth most commonly diagnosed tumor. Our study is aimed at identifying the prognostic value of EMT-related genes in gastric adenocarcinoma. Methods Firstly, high-throughput and clinical data were downloaded from The Cancer Genome Atlas (TCGA) database. 99 differentially expressed EMT-related genes (ERGs) were obtained in these gastric adenocarcinoma data. Secondly, GO and KEGG enrichment analyses show that EMT may promote gastric carcinogenesis. Next, 10 ERGs associated with prognosis of gastric adenocarcinoma patients are screened out by univariate Cox regression, and 6 pivotal prognostic ERGs (MMP8, MMP11, TFDP3, MYB, F2, and CNTN1) are identified through multivariate Cox regression. These 6 genes are confirmed with significant prognostic value in gastric adenocarcinoma through overall survival (OS) analysis. Finally, a risk score formula is constructed and tested in another gastric adenocarcinoma cohort from GEO. Results 99 differentially expressed EMT-related genes (ERGs) and their enriched pathways are identified. 10 ERGs are strongly related to the prognosis of GAC patients. A risk score formula of 6 prognosis-related ERGs used to predict the prognosis of gastric adenocarcinoma patients is identified and tested (risk score = 0.448115∗expression value of MMP8 + 0.378892∗expression value of MMP11 − 0.3226∗expression value of MYB + 1.322812∗expression value of TFDP3 + 0.325063∗expression value of F2 + 0.334197∗expression value of CNTN1). Conclusion This study provides a potential prognostic signature for predicting prognosis of gastric adenocarcinoma patients and molecular insights of EMT in gastric adenocarcinoma, and the formula focusing on the prognosis of gastric adenocarcinoma can be effective.
Collapse
|
8
|
Wang Q, Zhang L. Possible Molecular Mechanisms for the Roles of MicroRNA-21 Played in Lung Cancer. Technol Cancer Res Treat 2020; 18:1533033819875130. [PMID: 31506038 PMCID: PMC6740056 DOI: 10.1177/1533033819875130] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background: We aimed to find the possible molecular mechanisms for the roles of microRNA-21 underlying lung cancer development. Methods: MicroRNA-21-5p inhibitor was transfected into A549 cells. Total RNA was isolated from 10 samples, including 3 in control group (A549 cells), 3 in negative control group (A549 cells transferred with microRNA-21 negative control), and 4 in SH group (A549 cells transferred with microRNA-21 inhibitor), followed by RNA sequencing. Then, differentially expressed genes were screened for negative control group versus control group, SH group versus control group, and SH group versus negative control group. Functional enrichment analyses, protein–protein interaction network, and modules analyses were conducted. Target genes of hsa-miR-21-5p and transcription factors were predicted, followed by the regulatory network construction. Results: Minichromosome maintenance 10 replication initiation factor and cell division cycle associated 8 were important nodes in protein–protein interaction network with higher degrees. Cell division cycle associated 8 was enriched in cell division biological process. Furthermore, maintenance 10 replication initiation factor and cell division cycle associated 8 were significantly enriched in cluster 1 and micro-RNA-transcription factor-target genes regulating network. In addition, transcription factor Dp family member 3 (transcription factor of maintenance 10 replication initiation factor and cell division cycle associated 8) and RAD21 cohesin complex component (transcription factor of maintenance 10 replication initiation factor) were target genes of hsa-miR-21-5p. Conclusions: Micro-RNA-21 may play a key role in lung cancer partly via maintenance 10 replication initiation factor and cell division cycle associated 8. Furthermore, microRNA-21 targeted cell division cycle associated 8 and then played roles in lung cancer via the process of cell division. Transcription factor Dp family member 3 and RAD21 cohesin complex component are important transcription factors in microRNA-21-interfered lung cancer.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Linyou Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
9
|
Chun JN, Cho M, Park S, So I, Jeon JH. The conflicting role of E2F1 in prostate cancer: A matter of cell context or interpretational flexibility? Biochim Biophys Acta Rev Cancer 2019; 1873:188336. [PMID: 31870703 DOI: 10.1016/j.bbcan.2019.188336] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/20/2019] [Indexed: 02/07/2023]
Abstract
The transcription factor E2F1 plays a crucial role in mediating multiple cancer hallmark capabilities that regulate cell cycle, survival, apoptosis, metabolism, and metastasis. Aberrant activation of E2F1 is closely associated with a poor clinical outcome in various human cancers. However, E2F1 has conflictingly been reported to exert tumor suppressive activity, raising a question as to the nature of its substantive role in the control of cell fate. In this review, we summarize deregulated E2F1 activity and its role in prostate cancer. We highlight the recent advances in understanding the molecular mechanism by which E2F1 regulates the development and progression of prostate cancer, providing insight into how cell context or data interpretation shapes the role of E2F1 in prostate cancer. This review will aid in translating biomedical knowledge into therapeutic strategies for prostate cancer.
Collapse
Affiliation(s)
- Jung Nyeo Chun
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea
| | - Minsoo Cho
- Undergraduate Research Program, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Soonbum Park
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Insuk So
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea
| | - Ju-Hong Jeon
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Institute of Human-Environment Interface Biology, Seoul National University, Seoul 03080, Republic of Korea.
| |
Collapse
|
10
|
Abstract
The cyclin-dependent kinase (CDK)-RB-E2F axis forms the core transcriptional machinery driving cell cycle progression, dictating the timing and fidelity of genome replication and ensuring genetic material is accurately passed through each cell division cycle. The ultimate effectors of this axis are members of a family of eight distinct E2F genes encoding transcriptional activators and repressors. E2F transcriptional activity is tightly regulated throughout the cell cycle via transcriptional and translational regulation, post-translational modifications, protein degradation, binding to cofactors and subcellular localization. Alterations in one or more key components of this axis (CDKs, cyclins, CDK inhibitors and the RB family of proteins) occur in virtually all cancers and result in heightened oncogenic E2F activity, leading to uncontrolled proliferation. In this Review, we discuss the activities of E2F proteins with an emphasis on the newest atypical E2F family members, the specific and redundant functions of E2F proteins, how misexpression of E2F transcriptional targets promotes cancer and both current and developing therapeutic strategies being used to target this oncogenic pathway.
Collapse
Affiliation(s)
- Lindsey N Kent
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Gustavo Leone
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
11
|
Expression Profiles of Long Noncoding RNAs in Intranasal LPS-Mediated Alzheimer's Disease Model in Mice. BIOMED RESEARCH INTERNATIONAL 2019; 2019:9642589. [PMID: 30809552 PMCID: PMC6369469 DOI: 10.1155/2019/9642589] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 10/23/2018] [Accepted: 12/30/2018] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD), characterized by memory loss, cognitive decline, and dementia, is a progressive neurodegenerative disease. Although the long noncoding RNAs (lncRNAs) have recently been identified to play a role in the pathogenesis of AD, the specific effects of lncRNAs in AD remain unclear. In present study, we have investigated the expression profiles of lncRNAs in hippocampal of intranasal LPS-mediated Alzheimer's disease models in mice by microarray method. A total of 395 lncRNAs and 123 mRNAs was detected to express differently in AD models and controls (>2.0 folds, p<0.05). The microarray expression was validated by Quantitative Real-Time-PCR (qRT-PCR). The pathway analysis showed the mRNAs that correlated with lncRNAs were involved in inflammation, apoptosis, and nervous system related pathways. The lncRNA-TFs network analysis suggested the lncRNAs were mostly regulated by HMGA2, ONECUT2, FOXO1, and CDC5L. Additionally, lncRNA-target-TFs network analysis indicated the FOXL1, CDC5L, ONECUT2, and CDX1 to be the TFs most likely to regulate the production of these lncRNAs. This is the first study to investigate lncRNAs expression pattern in intranasal LPS-mediated Alzheimer's disease model in mice. And these results may facilitate the understanding of the pathogenesis of AD targeting lncRNAs.
Collapse
|
12
|
Ding LY, Chu M, Jiao YS, Hao Q, Xiao P, Li HH, Guo Q, Wang YD. TFDP3 regulates the apoptosis and autophagy in breast cancer cell line MDA-MB-231. PLoS One 2018; 13:e0203833. [PMID: 30235236 PMCID: PMC6147432 DOI: 10.1371/journal.pone.0203833] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 08/28/2018] [Indexed: 01/04/2023] Open
Abstract
Cancer/testis antigen TFDP3 belongs to the transcription factor DP(TFDP) family. It can bind to E2F family molecules to form a heterodimeric transcription factor E2F/TFDP complex. The complex is an important regulatory activator of cell cycle, involved in the regulation of cell proliferation, differentiation, apoptosis and other important physiological activities. In addition, TFDP3 has also been found to be a tumor-associated antigen that only expresses in malignant tumor tissue and normal testicular tissue; Thus, it is closely related to tumor occurrence and development. In this study, our group investigated the expression of TFDP3 in mononuclear cell samples from a variety of tissue-derived malignant tumors, breast cancer and benign breast lesions. The results show that TFDP3 is expressed in the malignant form of various tissues. Moreover, our recent research had focused on the ability of TFDP3 to influence the drug resistance and apoptosis of tumor cells. To further clarify the mechanisms involved in tumor resistance, this study also examined the expression of TFDP3 and tumor cell autophagy regulation; Autophagy helps cells cope with metabolic stress (such as in cases of malnutrition, growth factor depletion, hypoxia or hypoxia) removes erroneously folded proteins or defective organelles to prevent the accumulation of abnormal proteins; and removes intracellular pathogens. Our results showed that TFDP3 expression can induce autophagy by up-regulating the expression of autophagic key protein LC3(MAP1LC3) and increasing the number of autophagosomes during chemotherapy of malignant tumors. Then, DNA and organelles damage caused by the chemotherapy medicine are repaired. Thus, TFDP3 contributes toward tumor cell resistance. When siRNA inhibits TFDP3 expression, it can reduce cell autophagy, improving the sensitivity of tumor cells to chemotherapy drugs.
Collapse
Affiliation(s)
- Ling-yu Ding
- Department of Immunology, School of Basic Medical Science, Peking University, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Beijing, China
| | - Ming Chu
- Department of Immunology, School of Basic Medical Science, Peking University, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Beijing, China
- * E-mail: (MC); (YDW)
| | - Yun-shen Jiao
- Department of Immunology, School of Basic Medical Science, Peking University, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Beijing, China
| | - Qi Hao
- Department of Medical Genetics, School of Basic Medical Science, Peking University, Beijing, China
| | - Peng Xiao
- State Key Laboratory of Genetic Engineering, School of Life Science, Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai, China
| | - Huan-huan Li
- Department of Immunology, School of Basic Medical Science, Peking University, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Beijing, China
| | - Qi Guo
- School of Basic Medical Science, Peking University, Beijing, China
| | - Yue-dan Wang
- Department of Immunology, School of Basic Medical Science, Peking University, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Beijing, China
- * E-mail: (MC); (YDW)
| |
Collapse
|
13
|
TFDP3 confers chemoresistance in minimal residual disease within childhood T-cell acute lymphoblastic leukemia. Oncotarget 2018; 8:1405-1415. [PMID: 27902457 PMCID: PMC5352064 DOI: 10.18632/oncotarget.13630] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 11/08/2016] [Indexed: 01/16/2023] Open
Abstract
Acquired drug resistance in childhood T-cell acute lymphoblastic leukemia (T-ALL) remains a significant clinical problem. In this study, a novel gene therapy target for childhood T-ALL to overcome chemoresistance was discovered: TFDP3 increased in the minimal residual disease (MRD) positive childhood T-ALL patients. Then, we established a preclinical model of resistance to induction therapy to examine the functional relevance of TFDP3 to chemoresistance in MRD derived from Jurkat/E6-1. Jurkat xenografts in NOD/SCID mice were exposed to a four drug combination (VXLD) of vincristine (VCR), dexamethasone (DEX), L-asparaginase (L-asp) and daunorubicin (DNR). During the 4-week VXLD treatment, the level of TFDP3 increased 4-fold. High expression of TFDP3 was identified in the re-emerging lines (Jurkat/MRD) with increased chemoresistance, which is correlated with partially promoter demethylation of TFDP3. Downregulation of TFDP3 by RNA interference reversed chemoresistance in Jurkat/MRD accompanied by reinstated E2F1 activity that coincided with increased levels of p53, p73, and associated proapoptotic target genes. Importantly, TFDP3 silencing in vivo induced apparent benefit to overcome chemoresistance in combination with VXLD treatment. Collectively, TFDP3 confers chemoresistance in MRD within childhood T-ALL, indicating that TFDP3 is a potential gene therapy target for residual cancer.
Collapse
|
14
|
Jiao Y, Ding L, Chu M, Wang T, Kang J, Zhao X, Li H, Chen X, Gao Z, Gao L, Wang Y. Effects of cancer-testis antigen, TFDP3, on cell cycle regulation and its mechanism in L-02 and HepG2 cell lines in vitro. PLoS One 2017; 12:e0182781. [PMID: 28797103 PMCID: PMC5552311 DOI: 10.1371/journal.pone.0182781] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 07/24/2017] [Indexed: 12/31/2022] Open
Abstract
TFDP3, also be known as HCA661, was one of the cancer-testis antigens, which only expressed in human tissues. The recent researches about TFDP3 mostly focused on its ability to control the drug resistance and apoptosis of tumor cells. However, the role of TFDP3 in the progress of the cell cycle is rarely involved. In this study, we examined the expression of TFDP3 in human liver tissues firstly. After that, we detect the expression of TFDP3 at the RNA level and protein level in L-02 cell line and HepG2 cell line, and the location of TFDP3 was defined by immunofluorescence technique. Furthermore, we synchronized the cells to G1 phase, S phase and G2 phase, and arrested cell mitosis. The localization of TFDP3 and co-localization with E2F1 molecules in different phases of hepatocyte lines. Finally, TFDP3 gene knockout was performed on L-02 and HepG2 cell lines, and detected the new cell cycles by flow cytometry. The result showed that the expression of TFDP3 molecule is negative in normal liver tissue, but positive in immortalized human hepatocyte cell line, and the expression level is lower than in hepatocellular carcinoma cell line. The expression level of TFDP3 was in the dynamic change of L-02 and HepG2 cell lines, and was related to the phase transition. TFDP3 can bind to E2F1 molecule to form E2F/TFDP3 complex; and the localizations of TFDP3 and E2F1 molecules and the co-localization were different in different phases of cell cycle in the nucleus and cytoplasm, which indicated that the E2F/TFDP3 complex involved in the process of regulating the cell cycle. By knocking down the TFDP3 expression level in L-02 and HepG2 cell lines, the cell cycle would be arrested in S phase, which confirmed that TFDP3 can be a potential target for tumor therapy.
Collapse
Affiliation(s)
- Yunshen Jiao
- Department of Immunology, School of Basic Medical Science, Peking University, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Beijing, China
| | - Lingyu Ding
- Department of Immunology, School of Basic Medical Science, Peking University, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Beijing, China
| | - Ming Chu
- Department of Immunology, School of Basic Medical Science, Peking University, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Beijing, China
- * E-mail: (MC); (YDW)
| | - Tieshan Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jiarui Kang
- Department of Pathology, the First Affiliated Hospital of General Hospital of Chinese People’s Liberation Army, Beijing, China
| | - Xiaofan Zhao
- Department of Immunology, School of Basic Medical Science, Peking University, Beijing, China
| | - Huanhuan Li
- Department of Immunology, School of Basic Medical Science, Peking University, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Beijing, China
| | - Xi Chen
- Department of Immunology, School of Basic Medical Science, Peking University, Beijing, China
| | - Zirui Gao
- Department of Immunology, School of Basic Medical Science, Peking University, Beijing, China
| | - Likai Gao
- Department of Immunology, School of Basic Medical Science, Peking University, Beijing, China
| | - Yuedan Wang
- Department of Immunology, School of Basic Medical Science, Peking University, Beijing, China
- Key Laboratory of Medical Immunology, Ministry of Health, Beijing, China
- * E-mail: (MC); (YDW)
| |
Collapse
|
15
|
Yin K, Liu Y, Chu M, Wang Y. TFDP3 Regulates Epithelial-Mesenchymal Transition in Breast Cancer. PLoS One 2017; 12:e0170573. [PMID: 28114432 PMCID: PMC5256886 DOI: 10.1371/journal.pone.0170573] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 01/06/2017] [Indexed: 11/19/2022] Open
Abstract
Breast cancer remains a lethal disease to women due to lymph node metastasis, the tumor microenvironment, secondary resistance and other unknown factors. Several important transcription factors involved in this disease, such as PTEN, p53 and beta-catenin, have been identified and researched in-depth as candidates for targeted therapy in breast cancer. TFDP3 is a new, promising candidate for transcriptional regulation in breast cancer, although it was first identified in hepatocellular carcinoma. Here, we demonstrate that TFDP3 is expressed in a variety of malignancies, normal testis tissue and breast cancer cell lines and thus provide evidence that TFDP3 is a cancer-testis antigen. We illustrate that overexpression or silencing TFDP3 interferes with epithelial-mesenchymal transition but does not influence cell proliferation, indicating that the TFDP3 protein acts as a transcription factor during epithelial-mesenchymal transition. These data highlight that TFDP3 is expressed in breast cancer, that it is a member of the cancer-testis antigen family and that it functions as a regulator in epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Kailin Yin
- Department of Immunology, School of Basic Medical Science, Peking University, Beijing, China
- * E-mail: (KLY); (YDW)
| | - Yanchen Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Liaoning, China
| | - Ming Chu
- Department of Immunology, School of Basic Medical Science, Peking University, Beijing, China
| | - Yuedan Wang
- Department of Immunology, School of Basic Medical Science, Peking University, Beijing, China
- * E-mail: (KLY); (YDW)
| |
Collapse
|
16
|
Gao H, Song X, Kang T, Yan B, Feng L, Gao L, Ai L, Liu X, Yu J, Li H. Long noncoding RNA CRNDE functions as a competing endogenous RNA to promote metastasis and oxaliplatin resistance by sponging miR-136 in colorectal cancer. Onco Targets Ther 2017; 10:205-216. [PMID: 28115855 PMCID: PMC5221653 DOI: 10.2147/ott.s116178] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Colorectal neoplasia differentially expressed (CRNDE) is a novel gene recognized as a long noncoding RNA (lncRNA) that is highly elevated in colorectal cancer and many other solid tumors but its functions on metastasis and oxaliplatin (OXA) resistance are unknown. In our study, we confirmed the upregulation of CRNDE in both primary specimens from colorectal cancer patients and colorectal cancer cell lines. Knockdown of CRNDE expression inhibited the migration and invasion potency of colorectal cancer cells with no effect on cell apoptosis. Overexpression of CRNDE promoted the migration and invasion potency of colorectal cancer cells. Furthermore, we found that CRNDE conferred chemoresistance in colorectal cancer cells. Knockdown of CRNDE with OXA treatment decreased cell viability and promoted DNA damage and cell apoptosis, while the overexpression of CRNDE with OXA treatment reduced DNA damage and cell apoptosis. Further in-depth mechanistic studies revealed that CRNDE functioned as a competing endogenous RNA for miR-136, led to the de-repression of its endogenous target, E2F transcription factor 1 (E2F1). Overall, our findings demonstrate that CRNDE functions as a competing endogenous RNA to promote metastasis and OXA resistance by sponging miR-136 in colorectal cancer.
Collapse
Affiliation(s)
- Hongyan Gao
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University
| | - Xiaodi Song
- Department of Pharmacy, Xi'an Central Hospital, Xi'an
| | - Ting Kang
- Department of Oncology, Yan'an University Affiliated Hospital, Yan'an
| | - Baohong Yan
- Department of Pharmacy, Hong-Hui Hospital, Xi'an Jiaotong University Medical College, Xi'an
| | - Li Feng
- Department of Anorectal Surgery, Ankang City Central Hospital, Ankang
| | - Li Gao
- Department of Pharmacy, Yan'an University Affiliated Hospital
| | - Liang Ai
- Department of Pharmacy, Yan'an University Affiliated Hospital
| | - Xiaoni Liu
- Department of Endocrinology, Yan'an People's Hospital, Yan'an
| | - Jie Yu
- Department of General Surgery, Nuclear Industry 215 Hospital of Shaanxi Province, Xianyang
| | - Huiqi Li
- Department of General Surgery, The People's Hospital of Baoji City, Baoji, People's Republic of China
| |
Collapse
|
17
|
Jayachandran A, Lo PH, Chueh AC, Prithviraj P, Molania R, Davalos-Salas M, Anaka M, Walkiewicz M, Cebon J, Behren A. Transketolase-like 1 ectopic expression is associated with DNA hypomethylation and induces the Warburg effect in melanoma cells. BMC Cancer 2016; 16:134. [PMID: 26907172 PMCID: PMC4763451 DOI: 10.1186/s12885-016-2185-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 02/16/2016] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND The metabolism of cancer cells is often reprogrammed by dysregulation of metabolic enzymes. Transketolase-like 1 (TKTL1) is a homodimeric transketolase linking the pentose-phosphate pathway with the glycolytic pathway. It is generally silenced at a transcriptional level in somatic tissues. However, in human cancers its expression is associated with the acquisition of a glycolytic phenotype (the Warburg effect) by cancer cells that contributes to the progression of malignant tumors. In melanoma, defective promoter methylation results in the expression of genes and their products that can affect the tumor cell's phenotype including the modification of immune and functional characteristics. The present study evaluates the role of TKTL1 as a mediator of disease progression in melanoma associated with a defective methylation phenotype. METHODS The expression of TKTL1 in metastatic melanoma tumors and cell lines was analysed by qRT-PCR and immunohistochemistry. The promoter methylation status of TKTL1 in melanoma cells was evaluated by quantitative methylation specific PCR. Using qRT-PCR, the effect of a DNA demethylating agent 5-aza-2'-deoxycytidine (5aza) on the expression of TKTL1 was examined. Biochemical and molecular analyses such as glucose consumption, lactate production, invasion, proliferation and cell cycle progression together with ectopic expression and siRNA mediated knockdown were used to investigate the role of TKTL1 in melanoma cells. RESULTS Expression of TKTL1 was highly restricted in normal adult tissues and was overexpressed in a subset of metastatic melanoma tumors and derived cell lines. The TKTL1 promoter was activated by hypomethylation and treatment with 5aza induced TKTL1 expression in melanoma cells. Augmented expression of TKTL1 in melanoma cells was associated with a glycolytic phenotype. Loss and gain of function studies revealed that TKTL1 contributed to enhanced invasion of melanoma cells. CONCLUSIONS Our data provide evidence for an important role of TKTL1 in aerobic glycolysis and tumor promotion in melanoma that may result from defective promoter methylation. This epigenetic change may enable the natural selection of tumor cells with a metabolic phenotype and thereby provide a potential therapeutic target for a subset of melanoma tumors with elevated TKTL1 expression.
Collapse
Affiliation(s)
- Aparna Jayachandran
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Heidelberg, VIC, 3084, Australia.,Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia.,Department of Medicine, University of Melbourne, Melbourne, VIC, 3010, Australia.,School of Cancer Medicine, Latrobe University, Melbourne, VIC, 3086, Australia.,School of Medicine and the Gallipoli Medical Research Foundation, The University of Queensland, Brisbane, QLD 4120, Australia
| | - Pu-Han Lo
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Heidelberg, VIC, 3084, Australia
| | - Anderly C Chueh
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Heidelberg, VIC, 3084, Australia.,ACRF Chemical Biology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Melbourne, 3010, Australia
| | - Prashanth Prithviraj
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Heidelberg, VIC, 3084, Australia.,Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia.,Department of Medicine, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Ramyar Molania
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Heidelberg, VIC, 3084, Australia.,Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - Mercedes Davalos-Salas
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Heidelberg, VIC, 3084, Australia.,Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - Matthew Anaka
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Heidelberg, VIC, 3084, Australia.,Department of Medicine, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Marzena Walkiewicz
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Heidelberg, VIC, 3084, Australia.,Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - Jonathan Cebon
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Heidelberg, VIC, 3084, Australia.,Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia.,Department of Medicine, University of Melbourne, Melbourne, VIC, 3010, Australia.,School of Cancer Medicine, Latrobe University, Melbourne, VIC, 3086, Australia
| | - Andreas Behren
- Ludwig Institute for Cancer Research, Melbourne-Austin Branch, Heidelberg, VIC, 3084, Australia. .,Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia. .,Department of Medicine, University of Melbourne, Melbourne, VIC, 3010, Australia. .,School of Cancer Medicine, Latrobe University, Melbourne, VIC, 3086, Australia. .,Cancer Immuno-biology Laboratory, Olivia Newton-John Cancer Research Institute, Level 5, Olivia Newton-John Cancer and Wellness Centre, 145 Studley Road, Heidelberg, VIC, 3084, Australia.
| |
Collapse
|
18
|
Transcriptional master regulator analysis in breast cancer genetic networks. Comput Biol Chem 2015; 59 Pt B:67-77. [PMID: 26362298 DOI: 10.1016/j.compbiolchem.2015.08.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 08/17/2015] [Accepted: 08/17/2015] [Indexed: 01/05/2023]
Abstract
Gene regulatory networks account for the delicate mechanisms that control gene expression. Under certain circumstances, gene regulatory programs may give rise to amplification cascades. Such transcriptional cascades are events in which activation of key-responsive transcription factors called master regulators trigger a series of gene expression events. The action of transcriptional master regulators is then important for the establishment of certain programs like cell development and differentiation. However, such cascades have also been related with the onset and maintenance of cancer phenotypes. Here we present a systematic implementation of a series of algorithms aimed at the inference of a gene regulatory network and analysis of transcriptional master regulators in the context of primary breast cancer cells. Such studies were performed in a highly curated database of 880 microarray gene expression experiments on biopsy-captured tissue corresponding to primary breast cancer and healthy controls. Biological function and biochemical pathway enrichment analyses were also performed to study the role that the processes controlled - at the transcriptional level - by such master regulators may have in relation to primary breast cancer. We found that transcription factors such as AGTR2, ZNF132, TFDP3 and others are master regulators in this gene regulatory network. Sets of genes controlled by these regulators are involved in processes that are well-known hallmarks of cancer. This kind of analyses may help to understand the most upstream events in the development of phenotypes, in particular, those regarding cancer biology.
Collapse
|
19
|
Fröhlich H. biRte: Bayesian inference of context-specific regulator activities and transcriptional networks. Bioinformatics 2015; 31:3290-8. [PMID: 26112290 DOI: 10.1093/bioinformatics/btv379] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 06/15/2015] [Indexed: 11/14/2022] Open
Abstract
UNLABELLED In the last years there has been an increasing effort to computationally model and predict the influence of regulators (transcription factors, miRNAs) on gene expression. Here we introduce biRte as a computationally attractive approach combining Bayesian inference of regulator activities with network reverse engineering. biRte integrates target gene predictions with different omics data entities (e.g. miRNA and mRNA data) into a joint probabilistic framework. The utility of our method is tested in extensive simulation studies and demonstrated with applications from prostate cancer and Escherichia coli growth control. The resulting regulatory networks generally show a good agreement with the biological literature. AVAILABILITY AND IMPLEMENTATION biRte is available on Bioconductor (http://bioconductor.org). CONTACT frohlich@bit.uni-bonn.de SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Holger Fröhlich
- University of Bonn, Institute for Computer Science, Römerstr. 164, 53117 Bonn, Germany
| |
Collapse
|
20
|
MiR-136 targets E2F1 to reverse cisplatin chemosensitivity in glioma cells. J Neurooncol 2014; 120:43-53. [DOI: 10.1007/s11060-014-1535-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 06/28/2014] [Indexed: 11/26/2022]
|