1
|
Qian Y, Zhao X, Wu F, Wang X, Chen T. TGF-β-induced acetylation of KLF5 drives TNFAIP2 transcription and EMT in nasopharyngeal carcinoma: Unveiling a novel regulatory mechanism. Exp Cell Res 2025; 447:114498. [PMID: 40054652 DOI: 10.1016/j.yexcr.2025.114498] [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: 12/18/2024] [Revised: 02/25/2025] [Accepted: 03/04/2025] [Indexed: 03/12/2025]
Abstract
Epithelial-mesenchymal transition (EMT) is one of the critical mechanisms underlying migration, invasion, and metastasis of nasopharyngeal carcinoma (NPC) cells. The transcription factor KLF5 plays a pivotal role in various cancers, however, its precise functions in NPC remain incompletely understood. This study aims to explore the detailed mechanisms by which TGF-β enhances TNFAIP2 transcription by acetylating KLF5, thereby inducing EMT in NPC. KLF5 was significantly overexpressed in NPC tissues and closely associated with adverse clinicopathological features of the patients. Further studies revealed that TGF-β markedly increased the expression of KLF5 and its acetylated form, Ac-KLF5, in NPC cells, with the acetylation status of KLF5 being crucial for its function. KLF5 induced EMT in NPC cells by directly binding to the TNFAIP2 promoter and promoting its transcription. The pro-migratory and pro-invasive effects of acetylated KLF5 on NPC cells depended on TNFAIP2. Additionally, in vivo experiments confirmed that TGF-β treatment induced tumors in NPC mouse models to exhibit apparent EMT characteristics. These results collectively support the central role of the TGF-β-KLF5-TNFAIP2 axis in EMT of NPC. This study elucidates the specific mechanisms by which TGF-β promotes TNFAIP2 transcription by acetylating KLF5, thereby inducing EMT in NPC. This discovery not only provides new insights into the pathogenesis of NPC but also identifies potential therapeutic targets for NPC treatment.
Collapse
Affiliation(s)
- Yi Qian
- Department of Health Management Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing City, China
| | - Xuxu Zhao
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Feiyang Wu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoqiang Wang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tao Chen
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
2
|
Zhang Z, Liu Y, Xu Y, Xu Z, Jia J, Jin Y, Wang W, Liu L. Abrogation of KLF5 sensitizes BRCA1-proficient pancreatic cancer to PARP inhibition. Acta Biochim Biophys Sin (Shanghai) 2024; 56:576-585. [PMID: 38433576 PMCID: PMC11090855 DOI: 10.3724/abbs.2023288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 12/14/2023] [Indexed: 03/05/2024] Open
Abstract
Poly ADP-ribose polymerase (PARP) inhibitor monotherapies are selectively effective in patients with pancreatic, breast, prostate, and ovarian cancers with BRCA1 mutations. Cancer patients with more frequent wild-type BRCA show poor responses to PARP inhibitors. Moreover, patients who are initially sensitive to these inhibitors eventually respond poorly to drugs. In the present study, we discover that abrogation of Kruppel-like factor 5 (KLF5) significantly inhibits homologous recombination, which is the main mechanism for DNA double-stranded repair. Furthermore, the downregulation of KLF5 expression promotes the DNA damage induced by olaparib and significantly reduces the IC 50 of the RARP inhibitor in pancreatic cancer cells. Overexpression of BRCA1 reverses the above effects caused by silencing of KLF5. Olaparib combined with a KLF5 inhibitor has an enhanced cytotoxic effect. Mechanistically, we identify BRCA1 as a KLF5 target gene. BRCA1 is positively correlated with KLF5 in PDAC tissue. Our results indicate that inhibition of KLF5 may induce BRCAness in a larger pancreatic cancer subset with proficient BRCA. The combination of KLF5 inhibitors and PARP inhibitors provides a novel treatment strategy to enhance the sensitivity of BRCA1-proficient pancreatic cancer to PARP inhibitors.
Collapse
Affiliation(s)
- Zheng Zhang
- Department of Pancreatic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
- Cancer CenterZhongshan HospitalFudan UniversityShanghai200032China
- Department of General SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Yuxin Liu
- of Liver DiseasesShanxi Medical UniversityTaiyuan030001China
| | - Yaolin Xu
- Department of Pancreatic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
- Cancer CenterZhongshan HospitalFudan UniversityShanghai200032China
- Department of General SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Zijin Xu
- Department of General SurgeryQingpu Branch of Zhongshan Hospital Affiliated to Fudan UniversityShanghai201700China
| | - Jinbin Jia
- of Liver DiseasesShanxi Medical UniversityTaiyuan030001China
| | - Yun Jin
- Department of Hepatobiliary and Pancreatic Surgerythe First People’s Hospital of Yunnan Provincethe Affiliated Hospital of Kunming University of Science and TechnologyKunming650500China
| | - Wenquan Wang
- Department of Pancreatic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
- Cancer CenterZhongshan HospitalFudan UniversityShanghai200032China
- Department of General SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Liang Liu
- Department of Pancreatic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
- Cancer CenterZhongshan HospitalFudan UniversityShanghai200032China
- Department of General SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| |
Collapse
|
3
|
Yuan M, Liu X, Wang M, Li Z, Li H, Leng L, Wang S. A Functional Variant Alters the Binding of Bone morphogenetic protein 2 to the Transcription Factor NF-κB to Regulate Bone morphogenetic protein 2 Gene Expression and Chicken Abdominal Fat Deposition. Animals (Basel) 2023; 13:3401. [PMID: 37958155 PMCID: PMC10650395 DOI: 10.3390/ani13213401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
In this study, we employed a dual-luciferase reporter assay and electrophoretic mobility shift analysis (EMSA) in vitro to explore whether a 12-base pair (bp) insertion/deletion (InDel) variant (namely g.14798187_14798188insTCCCTGCCCCCT) within intron 2 of the chicken BMP2 gene, which was significantly associated with chicken abdominal fat weight and abdominal fat percentage, is a functional marker and its potential regulatory mechanism. The reporter analysis demonstrated that the luciferase activity of the deletion allele was extremely significantly higher than that of the insertion allele (p < 0.01). A bioinformatics analysis revealed that compared to the deletion allele, the insertion allele created a transcription factor binding site of nuclear factor-kappa B (NF-κB), which exhibited an inhibitory effect on fat deposition. A dual-luciferase reporter assay demonstrated that the inhibitory effect of NF-κB on the deletion allele was stronger than that on the insertion allele. EMSA indicated that the binding affinity of NF-κB for the insertion allele was stronger than that for the deletion allele. In conclusion, the 12-bp InDel chicken BMP2 gene variant is a functional variant affecting fat deposition in chickens, which may partially regulate BMP2 gene expression by affecting the binding of transcription factor NF-κB to the BMP2 gene.
Collapse
Affiliation(s)
- Meng Yuan
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China; (M.Y.); (X.L.); (M.W.); (Z.L.); (H.L.)
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Xin Liu
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China; (M.Y.); (X.L.); (M.W.); (Z.L.); (H.L.)
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Mengdie Wang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China; (M.Y.); (X.L.); (M.W.); (Z.L.); (H.L.)
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Ziwei Li
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China; (M.Y.); (X.L.); (M.W.); (Z.L.); (H.L.)
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Hui Li
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China; (M.Y.); (X.L.); (M.W.); (Z.L.); (H.L.)
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Li Leng
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China; (M.Y.); (X.L.); (M.W.); (Z.L.); (H.L.)
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Shouzhi Wang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin 150030, China; (M.Y.); (X.L.); (M.W.); (Z.L.); (H.L.)
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin 150030, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
4
|
Shi FY, Wang Y, Huang D, Liang Y, Liang N, Chen XW, Gao G. Computational Assessment of the Expression-modulating Potential for Non-coding Variants. GENOMICS, PROTEOMICS & BIOINFORMATICS 2023; 21:662-673. [PMID: 34890839 PMCID: PMC10787178 DOI: 10.1016/j.gpb.2021.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 10/13/2021] [Accepted: 11/01/2021] [Indexed: 06/13/2023]
Abstract
Large-scale genome-wide association studies (GWAS) and expression quantitative trait locus (eQTL) studies have identified multiple non-coding variants associated with genetic diseases by affecting gene expression. However, pinpointing causal variants effectively and efficiently remains a serious challenge. Here, we developed CARMEN, a novel algorithm to identify functional non-coding expression-modulating variants. Multiple evaluations demonstrated CARMEN's superior performance over state-of-the-art tools. Applying CARMEN to GWAS and eQTL datasets further pinpointed several causal variants other than the reported lead single-nucleotide polymorphisms (SNPs). CARMEN scales well with the massive datasets, and is available online as a web server at http://carmen.gao-lab.org.
Collapse
Affiliation(s)
- Fang-Yuan Shi
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Biomedical Pioneering Innovative Center (BIOPIC) & Beijing Advanced Innovation Center for Genomics (ICG), Center for Bioinformatics (CBI), Peking University, Beijing 100871, China
| | - Yu Wang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Biomedical Pioneering Innovative Center (BIOPIC) & Beijing Advanced Innovation Center for Genomics (ICG), Center for Bioinformatics (CBI), Peking University, Beijing 100871, China
| | - Dong Huang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Yu Liang
- Human Aging Research Institute, School of Life Science, Nanchang University, Nanchang 330031, China
| | - Nan Liang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Biomedical Pioneering Innovative Center (BIOPIC) & Beijing Advanced Innovation Center for Genomics (ICG), Center for Bioinformatics (CBI), Peking University, Beijing 100871, China
| | - Xiao-Wei Chen
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Ge Gao
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Biomedical Pioneering Innovative Center (BIOPIC) & Beijing Advanced Innovation Center for Genomics (ICG), Center for Bioinformatics (CBI), Peking University, Beijing 100871, China.
| |
Collapse
|
5
|
Leeming RC, Koutros S, Karagas MR, Baris D, Schwenn M, Johnson A, Zens MS, Schned AR, Rothman N, Silverman DT, Passarelli MN. Diet quality, common genetic polymorphisms, and bladder cancer risk in a New England population-based study. Eur J Nutr 2022; 61:3905-3913. [PMID: 35759030 PMCID: PMC10329807 DOI: 10.1007/s00394-022-02932-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 05/31/2022] [Indexed: 11/04/2022]
Abstract
PURPOSE We examined the interaction between common genetic bladder cancer variants, diet quality, and bladder cancer risk in a population-based case-control study conducted in New England. METHODS At the time of enrollment, 806 bladder cancer cases and 974 controls provided a DNA sample and completed a diet history questionnaire. Diet quality was assessed using the 2010 Alternate Healthy Eating Index (AHEI-2010) score. Single nucleotide polymorphisms (SNPs) reported in genome-wide association studies to be associated with bladder cancer risk were combined into a polygenic risk score and also examined individually for interaction with the AHEI-2010. Adjusted odds ratios (OR) and 95% confidence intervals (CI) were calculated using logistic regression. RESULTS A 1-standard deviation increase in polygenic risk score was associated with higher bladder cancer risk (OR, 1.34; 95% CI 1.21-1.49). Adherence to the AHEI-2010 was not associated with bladder cancer risk (OR, 0.99; 95% CI 0.98-1.00) and the polygenic risk score did not appear to modify the association between the AHEI-2010 and bladder cancer risk. In single-SNP analyses, rs8102137 (bladder cancer risk allele, C) modified the association between the AHEI-2010 total score and bladder cancer risk, with the strongest evidence for the AHEI-2010 long chain fat guideline (OR for TT, 0.92; 95% CI 0.87-0.98; OR for CT, 1.02; 95% CI 0.96-1.08; OR for CC, 1.03; 95% CI 0.93-1.14; p for interaction, 0.02). CONCLUSIONS In conclusion, rs8102137 near the cyclin E1 gene ( CCNE1 ) may be involved in gene-diet interactions for bladder cancer risk.
Collapse
Affiliation(s)
- Reno C Leeming
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, 1 Medical Center Drive, HB 7927, Hanover, Lebanon, NH, 03756, USA
| | - Stella Koutros
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, Bethesda, MD, USA
| | - Margaret R Karagas
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, 1 Medical Center Drive, HB 7927, Hanover, Lebanon, NH, 03756, USA
| | - Dalsu Baris
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, Bethesda, MD, USA
| | | | | | - Michael S Zens
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, 1 Medical Center Drive, HB 7927, Hanover, Lebanon, NH, 03756, USA
| | - Alan R Schned
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, Bethesda, MD, USA
| | - Debra T Silverman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, Bethesda, MD, USA
| | - Michael N Passarelli
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, 1 Medical Center Drive, HB 7927, Hanover, Lebanon, NH, 03756, USA.
| |
Collapse
|
6
|
Systems Drug Design for Muscle Invasive Bladder Cancer and Advanced Bladder Cancer by Genome-Wide Microarray Data and Deep Learning Method with Drug Design Specifications. Int J Mol Sci 2022; 23:ijms232213869. [PMID: 36430344 PMCID: PMC9692470 DOI: 10.3390/ijms232213869] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/05/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022] Open
Abstract
Bladder cancer is the 10th most common cancer worldwide. Due to the lack of understanding of the oncogenic mechanisms between muscle-invasive bladder cancer (MIBC) and advanced bladder cancer (ABC) and the limitations of current treatments, novel therapeutic approaches are urgently needed. In this study, we utilized the systems biology method via genome-wide microarray data to explore the oncogenic mechanisms of MIBC and ABC to identify their respective drug targets for systems drug discovery. First, we constructed the candidate genome-wide genetic and epigenetic networks (GWGEN) through big data mining. Second, we applied the system identification and system order detection method to delete false positives in candidate GWGENs to obtain the real GWGENs of MIBC and ABC from their genome-wide microarray data. Third, we extracted the core GWGENs from the real GWGENs by selecting the significant proteins, genes and epigenetics via the principal network projection (PNP) method. Finally, we obtained the core signaling pathways from the corresponding core GWGEN through the annotations of the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway to investigate the carcinogenic mechanisms of MIBC and ABC. Based on the carcinogenic mechanisms, we selected the significant drug targets NFKB1, LEF1 and MYC for MIBC, and LEF1, MYC, NOTCH1 and FOXO1 for ABC. To design molecular drug combinations for MIBC and ABC, we employed a deep neural network (DNN)-based drug-target interaction (DTI) model with drug specifications. The DNN-based DTI model was trained by drug-target interaction databases to predict the candidate drugs for MIBC and ABC, respectively. Subsequently, the drug design specifications based on regulation ability, sensitivity and toxicity were employed as filter criteria for screening the potential drug combinations of Embelin and Obatoclax for MIBC, and Obatoclax, Entinostat and Imiquimod for ABC from their candidate drugs. In conclusion, we not only investigated the oncogenic mechanisms of MIBC and ABC, but also provided promising therapeutic options for MIBC and ABC, respectively.
Collapse
|
7
|
Secreted miR-153 Controls Proliferation and Invasion of Higher Gleason Score Prostate Cancer. Int J Mol Sci 2022; 23:ijms23116339. [PMID: 35683018 PMCID: PMC9181550 DOI: 10.3390/ijms23116339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 06/01/2022] [Accepted: 06/03/2022] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PC) is a male common neoplasm and is the second leading cause of cancer death in American men. PC is traditionally diagnosed by the evaluation of prostate secreted antigen (PSA) in the blood. Due to the high levels of false positives, digital rectal examination and transrectal ultrasound guided biopsy are necessary in uncertain cases with elevated PSA levels. Nevertheless, the high mortality rate suggests that new PC biomarkers are urgently needed to help clinical diagnosis. In a previous study, we have identified a network of genes, altered in high Gleason Score (GS) PC (GS ≥ 7), being regulated by miR-153. Until now, no publication has explained the mechanism of action of miR-153 in PC. By in vitro studies, we found that the overexpression of miR-153 in high GS cell lines is required to control cell proliferation, migration and invasion rates, targeting Kruppel-like factor 5 (KLF5). Moreover, miR-153 could be secreted by exosomes and microvesicles in the microenvironment and, once entered into the surrounding tissue, could influence cellular growth. Being upregulated in high GS human PC, miR-153 could be proposed as a circulating biomarker for PC diagnosis.
Collapse
|
8
|
Alsheikh AJ, Wollenhaupt S, King EA, Reeb J, Ghosh S, Stolzenburg LR, Tamim S, Lazar J, Davis JW, Jacob HJ. The landscape of GWAS validation; systematic review identifying 309 validated non-coding variants across 130 human diseases. BMC Med Genomics 2022; 15:74. [PMID: 35365203 PMCID: PMC8973751 DOI: 10.1186/s12920-022-01216-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 03/17/2022] [Indexed: 02/08/2023] Open
Abstract
Background The remarkable growth of genome-wide association studies (GWAS) has created a critical need to experimentally validate the disease-associated variants, 90% of which involve non-coding variants. Methods To determine how the field is addressing this urgent need, we performed a comprehensive literature review identifying 36,676 articles. These were reduced to 1454 articles through a set of filters using natural language processing and ontology-based text-mining. This was followed by manual curation and cross-referencing against the GWAS catalog, yielding a final set of 286 articles. Results We identified 309 experimentally validated non-coding GWAS variants, regulating 252 genes across 130 human disease traits. These variants covered a variety of regulatory mechanisms. Interestingly, 70% (215/309) acted through cis-regulatory elements, with the remaining through promoters (22%, 70/309) or non-coding RNAs (8%, 24/309). Several validation approaches were utilized in these studies, including gene expression (n = 272), transcription factor binding (n = 175), reporter assays (n = 171), in vivo models (n = 104), genome editing (n = 96) and chromatin interaction (n = 33). Conclusions This review of the literature is the first to systematically evaluate the status and the landscape of experimentation being used to validate non-coding GWAS-identified variants. Our results clearly underscore the multifaceted approach needed for experimental validation, have practical implications on variant prioritization and considerations of target gene nomination. While the field has a long way to go to validate the thousands of GWAS associations, we show that progress is being made and provide exemplars of validation studies covering a wide variety of mechanisms, target genes, and disease areas. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01216-w.
Collapse
Affiliation(s)
- Ammar J Alsheikh
- Genomics Research Center, AbbVie Inc, North Chicago, Illinois, 60064, USA.
| | - Sabrina Wollenhaupt
- Information Research, AbbVie Deutschland GmbH & Co. KG, 67061, Knollstrasse, Ludwigshafen, Germany
| | - Emily A King
- Genomics Research Center, AbbVie Inc, North Chicago, Illinois, 60064, USA
| | - Jonas Reeb
- Information Research, AbbVie Deutschland GmbH & Co. KG, 67061, Knollstrasse, Ludwigshafen, Germany
| | - Sujana Ghosh
- Genomics Research Center, AbbVie Inc, North Chicago, Illinois, 60064, USA
| | | | - Saleh Tamim
- Genomics Research Center, AbbVie Inc, North Chicago, Illinois, 60064, USA
| | - Jozef Lazar
- Genomics Research Center, AbbVie Inc, North Chicago, Illinois, 60064, USA
| | - J Wade Davis
- Genomics Research Center, AbbVie Inc, North Chicago, Illinois, 60064, USA
| | - Howard J Jacob
- Genomics Research Center, AbbVie Inc, North Chicago, Illinois, 60064, USA
| |
Collapse
|
9
|
Expression and Prognosis Value of the KLF Family Members in Colorectal Cancer. JOURNAL OF ONCOLOGY 2022; 2022:6571272. [PMID: 35345512 PMCID: PMC8957442 DOI: 10.1155/2022/6571272] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/03/2022] [Accepted: 01/07/2022] [Indexed: 12/17/2022]
Abstract
Krüppel-like factors (KLFs) are some kind of transcriptional regulator that regulates a broad range of cellular functions and has been linked to the development of certain malignancies. KLF expression patterns and prognostic values in colorectal cancer (CRC) have, however, been investigated rarely. To investigate the differential expression, predictive value, and gene mutations of KLFs in CRC patients, we used various online analytic tools, including ONCOMINE, TCGA, cBioPortal, and the TIMER database. KLF2-6, KLF8-10, KLF12-15, and KLF17 mRNA expression levels were dramatically downregulated in CRC tissues, but KLF1, KLF7, and KLF16 mRNA expression levels were significantly elevated in CRC tissues. According to the findings of Cox regression analysis, upregulation of KLF3, KLF5, and KLF6 and downregulation of KLF15 were linked with a better prognosis in CRC. For functional enrichment, our findings revealed that KLF members are involved in a variety of cancer-related biological processes. In colon cancer and rectal cancer, KLFs were also shown to be associated with a variety of immune cells. The findings of this research reveal that KLF family members' mRNA expression levels are possible prognostic indicators for patients with CRC.
Collapse
|
10
|
FOXC1 Binds Enhancers and Promotes Cisplatin Resistance in Bladder Cancer. Cancers (Basel) 2022; 14:cancers14071717. [PMID: 35406487 PMCID: PMC8996937 DOI: 10.3390/cancers14071717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/18/2022] [Accepted: 03/18/2022] [Indexed: 01/25/2023] Open
Abstract
Simple Summary In bladder cancer, cisplatin remains the front-line therapy, but drug resistance is common. Previously, we showed that cancer cells can spontaneously convert to an aggressive drug-resistant phenotype without mutational events. In the current work, we explored the epigenetic mechanism behind the conversion to the drug-resistant phenotype. We discovered that drug-resistant cells have differentially accessible enhancers, which are bound by FOXC1, a transcription factor that is overexpressed in these cells. Accordingly, FOXC1 knockout significantly attenuates the emergence of the drug-resistant phenotype and reduces cell survival upon cisplatin treatment. These findings suggest that FOXC1 binding at accessible enhancers promotes cisplatin drug resistance in bladder cancer cells. Therefore, FOXC1 targeting may be a new therapeutic avenue to mitigate cisplatin resistance and improve treatment efficacy in bladder cancer. Abstract Chemotherapy resistance is traditionally attributed to DNA mutations that confer a survival advantage under drug selection pressure. However, in bladder cancer and other malignancies, we and others have previously reported that cancer cells can convert spontaneously to an aggressive drug-resistant phenotype without prior drug selection or mutational events. In the current work, we explored possible epigenetic mechanisms behind this phenotypic plasticity. Using Hoechst dye exclusion and flow cytometry, we isolated the aggressive drug-resistant cells and analyzed their chromatin accessibility at regulatory elements. Compared to the rest of the cancer cell population, the aggressive drug-resistant cells exhibited enhancer accessibility changes. In particular, we found that differentially accessible enhancers were enriched for the FOXC1 transcription factor motif, and that FOXC1 was the most significantly overexpressed gene in aggressive drug-resistant cells. ChIP-seq analysis revealed that differentially accessible enhancers in aggressive drug-resistant cells had a higher FOXC1 binding, which regulated the expression of adjacent cancer-relevant genes like ABCB1 and ID3. Accordingly, cisplatin treatment of bladder cancer cells led to an increased FOXC1 expression, which mediated cell survival and conversion to a drug-resistant phenotype. Collectively, these findings suggest that FOXC1 contributes to phenotypic plasticity by binding enhancers and promoting a mutation-independent shift towards cisplatin resistance in bladder cancer.
Collapse
|
11
|
Luo Y, Chen C. The roles and regulation of the KLF5 transcription factor in cancers. Cancer Sci 2021; 112:2097-2117. [PMID: 33811715 PMCID: PMC8177779 DOI: 10.1111/cas.14910] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/27/2021] [Accepted: 03/30/2021] [Indexed: 12/11/2022] Open
Abstract
Krüppel‐like factor 5 (KLF5) is a member of the KLF family. Recent studies have suggested that KLF5 regulates the expression of a large number of new target genes and participates in diverse cellular functions, such as stemness, proliferation, apoptosis, autophagy, and migration. In response to multiple signaling pathways, various transcriptional modulation and posttranslational modifications affect the expression level and activity of KLF5. Several transgenic mouse models have revealed the physiological and pathological functions of KLF5 in different cancers. Studies of KLF5 will provide prognostic biomarkers, therapeutic targets, and potential drugs for cancers.
Collapse
Affiliation(s)
- Yao Luo
- Medical Faculty of Kunming University of Science and Technology, Kunming, China.,Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
12
|
Moritake T, Kojima J, Kubota K, Terauchi F, Isaka K, Nishi H. Krüppel-like factor 5 is upregulated and induces cell proliferation in endometrial cancer. Oncol Lett 2021; 21:484. [PMID: 33968200 PMCID: PMC8100953 DOI: 10.3892/ol.2021.12745] [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: 10/09/2020] [Accepted: 02/22/2021] [Indexed: 12/09/2022] Open
Abstract
Krüppel-like factor 5 (KLF5) is involved in various cellular processes, such as cell proliferation and survival. KLF5 has been implicated in cancer pathology. The aim of the present study was to investigate the expression levels and function of KLF5 in endometrial cancer. A total of 30 patients, including 12 patients with endometrial cancer and 18 with benign gynecological diseases (controls), were enrolled at Tokyo Medical University (Tokyo, Japan) between March 2017 and May 2018. Endometrial cancer and control endometrium tissues were collected, and the expression levels of KLF5 were determined using reverse transcription-quantitative PCR, western blotting and immunohistochemistry. For the functional analyses of KLF5 in endometrial cancer, the present study employed a loss-of-function strategy in the human endometrial cancer cell lines in vitro. Ishikawa and HEC1 cells were transduced with lentiviral constructs expressing shRNAs targeting KLF5. MTT and TUNEL assays were performed in cells after knockdown to analyze the role of KLF5 in cell proliferation and survival. The results revealed that the mRNA and protein expression levels of KLF5 were increased in endometrial cancer tissues. In vitro analyses demonstrated that depletion of KLF5 inhibited cell proliferation and decreased the expression levels of cyclin E1. However, silencing KLF5 did not induce cell death. Overall, these results indicated that KLF5 may be crucial in the tumorigenesis of endometrial cancer and has potential as a therapeutic target.
Collapse
Affiliation(s)
- Tetsuya Moritake
- Department of Obstetrics and Gynecology, Tokyo Medical University, Shinjuku, Tokyo 160-0023, Japan
| | - Junya Kojima
- Department of Obstetrics and Gynecology, Tokyo Medical University, Shinjuku, Tokyo 160-0023, Japan
| | - Kaiyu Kubota
- Department of Obstetrics and Gynecology, Tokyo Medical University, Shinjuku, Tokyo 160-0023, Japan.,Division of Grassland Farming, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Nasushiobara, Tochigi 329-2793, Japan
| | - Fumitoshi Terauchi
- Department of Obstetrics and Gynecology, Tokyo Medical University, Shinjuku, Tokyo 160-0023, Japan
| | - Keiichi Isaka
- Department of Obstetrics and Gynecology, Tokyo Medical University, Shinjuku, Tokyo 160-0023, Japan
| | - Hirotaka Nishi
- Department of Obstetrics and Gynecology, Tokyo Medical University, Shinjuku, Tokyo 160-0023, Japan
| |
Collapse
|
13
|
Wang W, Li Y, Li Z, Wang N, Xiao F, Gao H, Guo H, Li H, Wang S. Polymorphisms of KLF3 gene coding region and identification of their functionality for abdominal fat in chickens. Vet Med Sci 2020; 7:792-799. [PMID: 33369233 PMCID: PMC8136968 DOI: 10.1002/vms3.422] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/23/2020] [Accepted: 12/09/2020] [Indexed: 12/20/2022] Open
Abstract
KLF3 is a member of the Kruppel‐like factor (KLF) family of transcription factors, and plays an important role in several biological processes, including adipogenesis, erythropoiesis and B‐cell development. The purposes of this study are to search for polymorphisms of KLF3 coding region and to provide functional evidence for abdominal fat in chickens. A total of 168 SNPs in KLF3 coding region were detected in a unique chicken population, the Northeast Agricultural University broiler lines divergently selected for abdominal fat content (NEAUHLF). Of which three single nucleotide polymorphisms (g.3452T > C, g.8663A > G and g.10751G > A) were significantly correlated with abdominal fat weight (AFW) and abdominal fat percentage (AFP) of 329 birds from the 19th generation of NEAUHLF (FDR < 0.05). The reporter gene assay was performed to verify functionality of these three SNPs in both ICP‐1 and DF1 cells. Results showed that the luciferase activity of G allele was significantly higher than that of A allele in g.10751G > A (p < 0.05). However, there were no significant differences between different alleles of others two SNPs in luciferase activity. Overall, KLF3 is an important candidate gene that affects chicken abdominal fat content, and the g.10751G > A is a functional variant that potential would be applied to marker‐assisted selection (MAS) for selective breeding programme.
Collapse
Affiliation(s)
- Weijia Wang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China.,College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Yudong Li
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China.,College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Ziwei Li
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China.,College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Ning Wang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China.,College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Fan Xiao
- Fujian Sunnzer Biotechnology Development Co., Ltd., Guangze, Fujian Province, China
| | - Haihe Gao
- Fujian Sunnzer Biotechnology Development Co., Ltd., Guangze, Fujian Province, China
| | - Huaishun Guo
- Fujian Sunnzer Biotechnology Development Co., Ltd., Guangze, Fujian Province, China
| | - Hui Li
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China.,College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Shouzhi Wang
- Key Laboratory of Chicken Genetics and Breeding, Ministry of Agriculture and Rural Affairs, Harbin, China.,Key Laboratory of Animal Genetics, Breeding and Reproduction, Education Department of Heilongjiang Province, Harbin, China.,College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| |
Collapse
|
14
|
Ma P, Pan Y, Yang F, Fang Y, Liu W, Zhao C, Yu T, Xie M, Jing X, Wu X, Sun C, Li W, Xu T, Shu Y. KLF5-Modulated lncRNA NEAT1 Contributes to Tumorigenesis by Acting as a Scaffold for BRG1 to Silence GADD45A in Gastric Cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 22:382-395. [PMID: 33230443 PMCID: PMC7533296 DOI: 10.1016/j.omtn.2020.09.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 09/04/2020] [Indexed: 12/27/2022]
Abstract
Long noncoding RNAs (lncRNAs), genomic "dark matter," are deeply involved in diverse biological processes. The lncRNA nuclear paraspeckle assembly transcript 1 (NEAT1) is a highly participatory lncRNA; however, its roles in gastric cancer (GC) remain largely unexplored. Here, we demonstrated that the expression of NEAT1 was significantly increased and negatively correlated with prognosis in GC. Subsequent experiments confirmed that KLF5 can induce NEAT1 expression by binding to the NEAT1 promoter region. Further experiments revealed that NEAT1 silencing significantly suppressed cell proliferation both in vitro and in vivo and induced apoptosis. We used mRNA sequencing (mRNA-seq) to identify the preferentially affected genes linked to cell proliferation in cells with NEAT1 knockdown. Mechanistically, NEAT1 bound BRG1 (SMARCA4) directly, modulating H3K27me3 and H3K4me3 in the GADD45A promoter to regulate GADD45A-dependent G2/M cell cycle progression. In addition, BRG1 was significantly upregulated and correlated with outcomes in GC; moreover, it promoted cell proliferation both in vitro and in vivo. Taken together, our data support the importance of NEAT1 in promoting GC tumorigenesis and indicate that NEAT1 might be a diagnostic and therapeutic target in GC.
Collapse
Affiliation(s)
- Pei Ma
- Department of Oncology, Cancer Rehabilitation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People’s Republic of China
| | - Yutian Pan
- Department of Oncology, Cancer Rehabilitation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People’s Republic of China
| | - Fan Yang
- Department of Oncology, Cancer Rehabilitation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People’s Republic of China
| | - Yuan Fang
- Department of Oncology, Cancer Rehabilitation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People’s Republic of China
| | - Weitao Liu
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, People’s Republic of China
| | - Chenhui Zhao
- Department of Oncology, Cancer Rehabilitation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People’s Republic of China
| | - Tao Yu
- Department of Oncology, Cancer Rehabilitation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People’s Republic of China
| | - Mengyan Xie
- Department of Oncology, Cancer Rehabilitation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People’s Republic of China
| | - Xingming Jing
- Department of Oncology, Cancer Rehabilitation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People’s Republic of China
| | - Xi Wu
- Department of Oncology, Cancer Rehabilitation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People’s Republic of China
| | - Chongqi Sun
- Department of Oncology, Cancer Rehabilitation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People’s Republic of China
| | - Wei Li
- Department of Oncology, Cancer Rehabilitation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People’s Republic of China
- Department of Oncology, Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing 211166, People’s Republic of China
| | - Tongpeng Xu
- Department of Oncology, Cancer Rehabilitation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People’s Republic of China
| | - Yongqian Shu
- Department of Oncology, Cancer Rehabilitation Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People’s Republic of China
- Department of Oncology, Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing 211166, People’s Republic of China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing 211166, People’s Republic of China
- Corresponding author: Yongqian Shu, Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, People’s Republic of China.
| |
Collapse
|
15
|
Liao Q, Chen L, Zhang N, Xi Y, Hu S, Ng DM, Ahmed FYH, Zhao G, Fan X, Xie Y, Dai X, Jin Y, Ge J, Dong C, Zhang X, Guo J. Network analysis of KLF5 targets showing the potential oncogenic role of SNHG12 in colorectal cancer. Cancer Cell Int 2020; 20:439. [PMID: 32943987 PMCID: PMC7487661 DOI: 10.1186/s12935-020-01527-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 08/29/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND KLF5 is a member of the Kruppel-like factor, subfamily of zinc finger proteins that are involved in cancers. KLF5 functions as a transcription factor and regulates the diverse protein-coding genes (PCGs) in colorectal cancer (CRC). However, the long non-coding RNAs (lncRNAs) regulated by KLF5 in CRC are currently unknown. METHODS In this study, we first designed a computational pipeline to determine the PCG and lncRNA targets of KLF5 in CRC. Then we analyzed the motif pattern of the binding regions for the lncRNA targets. The regulatory co-factors of KLF5 were then searched for through bioinformatics analysis. We also constructed a regulatory network for KLF5 and annotated its functions. Finally, one of the KLF5 lncRNA targets, SNHG12, was selected to further explore its expression pattern and functions in CRC. RESULTS We were able to identify 19 lncRNA targets of KLF5 and found that the motifs of the lncRNA binding sites were GC-enriched. Next, we pinpointed the transcription factors AR and HSF1 as the regulatory co-factors of KLF5 through bioinformatics analysis. Then, through the analysis of the regulatory network, we found that KLF5 may be involved in DNA replication, DNA repair, and the cell cycle. Furthermore, in the cell cycle module, the SNHG12 up-regulating expression pattern was verified in the CRC cell lines and tissues, associating it to CRC invasion and distal metastasis. This indicates that SNHG12 may play a critical part in CRC tumorigenesis and progression. Additionally, expression of SNHG12 was found to be down-regulated in CRC cell lines when KLF5 expression was knocked-down by siRNA; and a strong correlation was observed between the expression levels of SNHG12 and KLF5, further alluding to their regulatory relationship. CONCLUSIONS In conclusion, the network analysis of KLF5 targets indicates that SNHG12 may be a significant lncRNA in CRC.
Collapse
Affiliation(s)
- Qi Liao
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, 315211 Zhejiang China
| | - Linbo Chen
- Department of Gastroenterology, The Affiliated People's Hospital of Ningbo University, Ningbo, 315040 Zhejiang China
| | - Ning Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Yang Xi
- Department of Biochemistry and Molecular Biology, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, 315211 Zhejiang China
| | - Shiyun Hu
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, 315211 Zhejiang China
| | - Derry Minyao Ng
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, 315211 Zhejiang China
| | - Fatma Yislam Hadi Ahmed
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, 315211 Zhejiang China
| | - Guofang Zhao
- Hua Mei Hospital, University of Chinese Academy of Science, Ningbo, 315000 China
| | - Xiaoxiang Fan
- Hua Mei Hospital, University of Chinese Academy of Science, Ningbo, 315000 China
| | - Yangyang Xie
- Hua Mei Hospital, University of Chinese Academy of Science, Ningbo, 315000 China
| | - Xiaoyu Dai
- Hua Mei Hospital, University of Chinese Academy of Science, Ningbo, 315000 China
| | - Yanping Jin
- The Affiliated Hospital of School of Medicine, Ningbo University, Ningbo, 315020 China
| | - Jiaxin Ge
- The Affiliated Hospital of School of Medicine, Ningbo University, Ningbo, 315020 China
| | - Changzheng Dong
- Department of Preventative Medicine, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, 315211 Zhejiang China
| | - Xinjun Zhang
- The Affiliated Hospital of School of Medicine, Ningbo University, Ningbo, 315020 China
| | - Junming Guo
- Department of Biochemistry and Molecular Biology, Zhejiang Provincial Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, 315211 Zhejiang China
| |
Collapse
|
16
|
Rogerson C, Ogden S, Britton E, Ang Y, Sharrocks AD. Repurposing of KLF5 activates a cell cycle signature during the progression from a precursor state to oesophageal adenocarcinoma. eLife 2020; 9:e57189. [PMID: 32880368 PMCID: PMC7544504 DOI: 10.7554/elife.57189] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 09/03/2020] [Indexed: 12/13/2022] Open
Abstract
Oesophageal adenocarcinoma (OAC) is one of the most common causes of cancer deaths. Barrett's oesophagus (BO) is the only known precancerous precursor to OAC, but our understanding about the molecular events leading to OAC development is limited. Here, we have integrated gene expression and chromatin accessibility profiles of human biopsies and identified a strong cell cycle gene expression signature in OAC compared to BO. Through analysing associated chromatin accessibility changes, we have implicated the transcription factor KLF5 in the transition from BO to OAC. Importantly, we show that KLF5 expression is unchanged during this transition, but instead, KLF5 is redistributed across chromatin to directly regulate cell cycle genes specifically in OAC cells. This new KLF5 target gene programme has potential prognostic significance as high levels correlate with poorer patient survival. Thus, the repurposing of KLF5 for novel regulatory activity in OAC provides new insights into the mechanisms behind disease progression.
Collapse
Affiliation(s)
- Connor Rogerson
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
| | - Samuel Ogden
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
| | - Edward Britton
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
| | | | - Yeng Ang
- School of Medical Sciences, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
- GI Science Centre, Salford Royal NHS FT, University of ManchesterSalfordUnited Kingdom
| | - Andrew D Sharrocks
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of ManchesterManchesterUnited Kingdom
| |
Collapse
|
17
|
Norollahi SE, Hamidian SMT, Vahidi S, Babaei K, Samadani AA. Modifications of WNT signaling pathway genes including WNT1, KLF5 and WNT16 in colorectal cancer. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
18
|
Oh M, Kim SY, Gil JE, Byun JS, Cha DW, Ku B, Lee W, Kim WK, Oh KJ, Lee EW, Bae KH, Lee SC, Han BS. Nurr1 performs its anti-inflammatory function by regulating RasGRP1 expression in neuro-inflammation. Sci Rep 2020; 10:10755. [PMID: 32612143 PMCID: PMC7329810 DOI: 10.1038/s41598-020-67549-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 06/10/2020] [Indexed: 12/21/2022] Open
Abstract
Nurr1, a transcription factor belonging to the orphan nuclear receptor, has an essential role in the generation and maintenance of dopaminergic neurons and is important in the pathogenesis of Parkinson’ disease (PD). In addition, Nurr1 has a non-neuronal function, and it is especially well known that Nurr1 has an anti-inflammatory function in the Parkinson’s disease model. However, the molecular mechanisms of Nurr1 have not been elucidated. In this study, we describe a novel mechanism of Nurr1 function. To provide new insights into the molecular mechanisms of Nurr1 in the inflammatory response, we performed Chromatin immunoprecipitation sequencing (ChIP-Seq) on LPS-induced inflammation in BV2 cells and finally identified the RasGRP1 gene as a novel target of Nurr1. Here, we show that Nurr1 directly binds to the RasGRP1 intron to regulate its expression. Moreover, we also identified that RasGRP1 regulates the Ras-Raf-MEK-ERK signaling cascade in LPS-induced inflammation signaling. Finally, we conclude that RasGRP1 is a novel regulator of Nurr1’s mediated inflammation signaling.
Collapse
Affiliation(s)
- Mihee Oh
- Biodefense Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Sun Young Kim
- Biodefense Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Jung-Eun Gil
- Biodefense Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Jeong-Su Byun
- Biodefense Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Dong-Wook Cha
- Biodefense Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea.,Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon, 34113, Republic of Korea
| | - Bonsu Ku
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | | | - Won-Kon Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea.,Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon, 34113, Republic of Korea
| | - Kyoung-Jin Oh
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea.,Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon, 34113, Republic of Korea
| | - Eun-Woo Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea.,Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon, 34113, Republic of Korea
| | - Sang Chul Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea. .,Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon, 34113, Republic of Korea.
| | - Baek-Soo Han
- Biodefense Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea. .,Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea. .,Department of Functional Genomics, University of Science and Technology (UST) of Korea, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
19
|
Huang H, Han Y, Chen Z, Pan X, Yuan P, Zhao X, Zhu H, Wang J, Sun X, Shi P. ML264 inhibits osteosarcoma growth and metastasis via inhibition of JAK2/STAT3 and WNT/β-catenin signalling pathways. J Cell Mol Med 2020; 24:5652-5664. [PMID: 32285603 PMCID: PMC7214147 DOI: 10.1111/jcmm.15226] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/25/2020] [Accepted: 02/06/2020] [Indexed: 02/06/2023] Open
Abstract
Osteosarcoma, the most common bone malignancy, has a high morbidity rate and poor prognosis. Krüppel‐like factor 5 (KLF5) is a key transcriptional regulator of cellular proliferation whose overexpression is observed in osteosarcoma cell lines (U2OS, 143B, MG63 and SAOS2). ML264, a small‐molecule inhibitor of KLF5, exerts antiproliferative effects in colorectal cancer; however, its function in osteosarcoma remains unknown. Here, we explored the possible antitumour effects of ML264 on 143B and U2OS cell lines and murine tumour xenograft model. ML264 suppressed proliferation and clonogenic ability of osteosarcoma cells in a dose‐dependent manner. Moreover, ML264 induced G0/G1 cell cycle arrest, with no influence on apoptosis, and inhibited the migratory and invasive abilities of osteosarcoma cells, as demonstrated by wound‐healing and Transwell assays. Exposure to ML264 reduced the mRNA and protein levels of molecules associated with epithelial‐mesenchymal transition phenotype, including N‐cadherin, vimentin, Snail, matrix metalloproteinase (MMP) 9 and MMP13. Inhibition of signal transducer and activator of transcription (STAT) 3 phosphorylation and Wnt signalling was also observed. In the murine model of osteosarcoma, tumour growth was efficiently suppressed following a 10‐day treatment with ML264. Collectively, our findings demonstrate the potential value of ML264 as a novel anticancer drug for osteosarcoma.
Collapse
Affiliation(s)
- Hai Huang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Ying Han
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Zhijun Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xin Pan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Putao Yuan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xiangde Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Hongfang Zhu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Jiying Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xuewu Sun
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Peihua Shi
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| |
Collapse
|
20
|
Li J, Zhang B, Liu M, Fu X, Ci X, A J, Fu C, Dong G, Wu R, Zhang Z, Fu L, Dong JT. KLF5 Is Crucial for Androgen-AR Signaling to Transactivate Genes and Promote Cell Proliferation in Prostate Cancer Cells. Cancers (Basel) 2020; 12:cancers12030748. [PMID: 32245249 PMCID: PMC7140031 DOI: 10.3390/cancers12030748] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/15/2020] [Accepted: 03/17/2020] [Indexed: 01/08/2023] Open
Abstract
Androgen/androgen receptor (AR) signaling drives both the normal prostate development and prostatic carcinogenesis, and patients with advanced prostate cancer often develop resistance to androgen deprivation therapy. The transcription factor Krüppel-like factor 5 (KLF5) also regulates both normal and cancerous development of the prostate. In this study, we tested whether and how KLF5 plays a role in the function of AR signaling in prostate cancer cells. We found that KLF5 is upregulated by androgen depending on AR in LNCaP and C4-2B cells. Silencing KLF5, in turn, reduced AR transcriptional activity and inhibited androgen-induced cell proliferation and tumor growth in vitro and in vivo. Mechanistically, KLF5 occupied the promoter of AR, and silencing KLF5 repressed AR transcription. In addition, KLF5 and AR physically interacted with each other to regulate the expression of multiple genes (e.g., MYC, CCND1 and PSA) to promote cell proliferation. These findings indicate that, while transcriptionally upregulated by AR signaling, KLF5 also regulates the expression and transcriptional activity of AR in androgen-sensitive prostate cancer cells. The KLF5-AR interaction could provide a therapeutic opportunity for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Juan Li
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China (L.F.)
- School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen, Guangdong 518055, China;
| | - Baotong Zhang
- Emory Winship Cancer Institute, Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365-C Clifton Road, Atlanta, GA 30322, USA
| | - Mingcheng Liu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China (L.F.)
- School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen, Guangdong 518055, China;
| | - Xing Fu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China (L.F.)
- School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen, Guangdong 518055, China;
| | - Xinpei Ci
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Jun A
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China (L.F.)
- School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen, Guangdong 518055, China;
| | - Changying Fu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China (L.F.)
- School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen, Guangdong 518055, China;
| | - Ge Dong
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China (L.F.)
| | - Rui Wu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China (L.F.)
- School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen, Guangdong 518055, China;
| | - Zhiqian Zhang
- School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen, Guangdong 518055, China;
| | - Liya Fu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin 300071, China (L.F.)
| | - Jin-Tang Dong
- School of Medicine, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen, Guangdong 518055, China;
- Emory Winship Cancer Institute, Department of Hematology and Medical Oncology, Emory University School of Medicine, 1365-C Clifton Road, Atlanta, GA 30322, USA
- Correspondence:
| |
Collapse
|
21
|
Vlachostergios PJ, Faltas BM, Carlo MI, Nassar AH, Alaiwi SA, Sonpavde G. The emerging landscape of germline variants in urothelial carcinoma: Implications for genetic testing. Cancer Treat Res Commun 2020; 23:100165. [PMID: 31982787 DOI: 10.1016/j.ctarc.2020.100165] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 12/30/2019] [Indexed: 06/10/2023]
Abstract
Urothelial carcinoma (UC) of the bladder and upper tract (ureter, renal pelvis) is one of the most frequently occurring malignancies. While the majority of UC are chemically induced by smoking, accumulating evidence from genetic studies have demonstrated a small, but consistent impact of heritable gene variants and family history of UC on the development of the disease. Beyond the established association between upper tract UC and germline mismatch DNA repair defects as a defining feature of Lynch syndrome, newer investigations focusing on moderate- and high-risk cancer-related gene variants in DNA damage repair and other signaling pathways are expanding our knowledge on the heritable genetic basis of UC, opening new avenues in the breadth of genetic testing and in clinical counseling of these patients. Overcoming existing challenges in the interpretation of uncertain findings and family cascade testing may help expand our testing approach and guidelines. Following the paradigm of other tumor types, such as breast and ovarian cancers, germline genetic testing, particularly when combined with somatic testing, has the potential to directly benefit affected UC patients and their families in the future through therapeutic targeting (i.e. with poly(ADP-ribose)) polymerase inhibitors, immune checkpoint inhibitors) and genetically informed screening/surveillance, respectively.
Collapse
Affiliation(s)
| | - Bishoy M Faltas
- Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, United States; Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Maria I Carlo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Amin H Nassar
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States
| | - Sarah Abou Alaiwi
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, DANA 1230, Boston, MA 02215, United States
| | - Guru Sonpavde
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, DANA 1230, Boston, MA 02215, United States.
| |
Collapse
|
22
|
Shen Y, Lu Q, Ye H, Deng Z, Ma L, Zhang Q, Tang J, Yuan L. Genetic variant of MAML2 in the NOTCH signaling pathway and the risk of bladder cancer: A STROBE-compliant study. Medicine (Baltimore) 2020; 99:e18725. [PMID: 31914088 PMCID: PMC6959859 DOI: 10.1097/md.0000000000018725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The NOTCH signaling pathway plays a crucial role in cell phenotype and transformation. Single nucleotide polymorphisms (SNPs) may regulate gene expression to trigger bladder cancer susceptibility. Here, we aimed to explore the relationships between genetic variants in the NOTCH pathway and bladder cancer progression.We screened SNPs located in NOTCH pathway genes using the 1000 Genomes Project dataset (CHB). A case-control cohort study including 580 bladder cancer cases and 1101 controls was conducted to genotype the candidate SNPs. The expression quantitative trait locus (eQTL) and bioinformatics analyses were performed to explore the biological function of the SNPs' host gene and their relationship. Kaplan-Meier analysis was performed to assess the association between host gene expression and bladder cancer patient prognosis.The rs7944701 in the intron of mastermind-like 2 (MAML2) had the strongest signal and was related to bladder cancer risk (OR = 1.329, 95% CI = 1.115-1.583, P = .001). eQTL analysis showed that rs7944701 with a C allele was negatively associated with mastermind-like 2 (MAML2) expression (TT versus TC/CC). Bioinformatics analysis indicated that MAML2expression was lower in bladder cancer tissues than in non-tumor tissues (P = 5.46 × 10). Additionally, bladder cancer patients with high MAML2 expression had a significantly poorer prognosis (HR = 1.53, 95% CI = 1.29-1.82, P = .010).The rs7944701 in MAML2 was strongly associated with bladder cancer susceptibility in a Chinese population. This genetic variant and its host gene could be a potential novel biomarker for individuals suffering from bladder cancer.
Collapse
Affiliation(s)
- Yang Shen
- Department of Urology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine
- Urological Tumor Department, Jiangsu Provincial Institute of Clinical Research
| | - Qian Lu
- Department of Urology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine
- Urological Tumor Department, Jiangsu Provincial Institute of Clinical Research
| | - Hesong Ye
- Department of Urology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Second Chinese Medicine Hospital, Nanjing, China
| | - Zhonglei Deng
- Department of Urology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine
- Urological Tumor Department, Jiangsu Provincial Institute of Clinical Research
| | - Long Ma
- Department of Urology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine
- Urological Tumor Department, Jiangsu Provincial Institute of Clinical Research
| | - Qingling Zhang
- Department of Urology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine
- Urological Tumor Department, Jiangsu Provincial Institute of Clinical Research
| | - Jingyuan Tang
- Department of Urology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine
- Urological Tumor Department, Jiangsu Provincial Institute of Clinical Research
| | - Lin Yuan
- Department of Urology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine
- Urological Tumor Department, Jiangsu Provincial Institute of Clinical Research
| |
Collapse
|
23
|
Chiang CC, Yeh CT, Hwang TL, Chu YD, Lim SN, Chen CW, Kuo CJ, Le PH, Chen TH, Lin WR. The GALNT14 Genotype Predicts Postoperative Outcome of Pancreatic Ductal Adenocarcinoma. J Clin Med 2019; 8:2225. [PMID: 31888240 PMCID: PMC6947583 DOI: 10.3390/jcm8122225] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is notorious for its poor prognosis. The current mainstay of treatment for PDA is surgical resection followed by adjuvant chemotherapy. However, it is difficult to predict the post-operative outcome because of the lack of reliable markers. The single-nucleotide polymorphism (SNP) of N-acetylgalactosaminyltransferase14 (GALNT14) has been proven to predict the progression-free survival (PFS), overall survival (OS) and response to chemotherapy in various types of gastrointestinal (GI) cancers. However, its role in PDA has not been studied. This study aims to investigate whether the GALNT14 SNP genotype can be a prognostic marker for PDA. A cohort of one hundred and three PDA patients having received surgical resection were retrospectively enrolled. GALNT14 genotypes and the clinicopathological parameters were correlated with postoperative prognosis. The genotype analysis revealed that 19.4%, 60.2% and 20.4% of patients had the GALNT14 "TT", "TG" and "GG" genotypes, respectively. The patients with the "GG" genotype had a mean OS time of 37.1 months (95% confidence interval [CI]: 18.2-56.1) and those with the "non-GG" genotype had a mean OS time of 16.1 months (95% CI: 13.1-19.2). Kaplan-Meier analysis showed that the "GG" genotype had a significantly better OS compared to the "non-GG" genotype (p = 0.005). However, there was no significant difference between the "GG" and "non-GG" genotypes in PFS (p = 0.172). The baseline characteristics between patients with the "GG" and "non-GG" genotypes were compared, and no significant difference was found. Univariate followed by multivariate Cox proportional hazard models demonstrated the GALNT14 "GG" genotype, negative resection margin, and locoregional disease as independent predictors for favorable OS (p = 0.003, p = 0.037, p = 0.021, respectively). Sensitivity analysis was performed in each subgroup to examine the relationship of GALNT14 with different clinicopathological variables and no heterogeneity was found. The GALNT14 "GG" genotype is associated with favorable survival outcome, especially OS, in patients with resected PDA and could serve as a prognostic marker.
Collapse
Affiliation(s)
- Chun-Cheng Chiang
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (C.-C.C.); (C.-T.Y.); (C.-W.C.); (C.-J.K.); (P.-H.L.); (T.-H.C.)
| | - Chau-Ting Yeh
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (C.-C.C.); (C.-T.Y.); (C.-W.C.); (C.-J.K.); (P.-H.L.); (T.-H.C.)
- Liver Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (T.-L.H.); (S.-N.L.)
| | - Tsann-Long Hwang
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (T.-L.H.); (S.-N.L.)
- Department of General Surgery, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Yu-De Chu
- Liver Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan;
| | - Siew-Na Lim
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (T.-L.H.); (S.-N.L.)
- Department of Neurology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Chun-Wei Chen
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (C.-C.C.); (C.-T.Y.); (C.-W.C.); (C.-J.K.); (P.-H.L.); (T.-H.C.)
| | - Chia-Jung Kuo
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (C.-C.C.); (C.-T.Y.); (C.-W.C.); (C.-J.K.); (P.-H.L.); (T.-H.C.)
| | - Puo-Hsien Le
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (C.-C.C.); (C.-T.Y.); (C.-W.C.); (C.-J.K.); (P.-H.L.); (T.-H.C.)
| | - Tsung-Hsing Chen
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (C.-C.C.); (C.-T.Y.); (C.-W.C.); (C.-J.K.); (P.-H.L.); (T.-H.C.)
| | - Wey-Ran Lin
- Department of Gastroenterology and Hepatology, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (C.-C.C.); (C.-T.Y.); (C.-W.C.); (C.-J.K.); (P.-H.L.); (T.-H.C.)
- Liver Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan 333, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (T.-L.H.); (S.-N.L.)
| |
Collapse
|
24
|
Morton AR, Dogan-Artun N, Faber ZJ, MacLeod G, Bartels CF, Piazza MS, Allan KC, Mack SC, Wang X, Gimple RC, Wu Q, Rubin BP, Shetty S, Angers S, Dirks PB, Sallari RC, Lupien M, Rich JN, Scacheri PC. Functional Enhancers Shape Extrachromosomal Oncogene Amplifications. Cell 2019; 179:1330-1341.e13. [PMID: 31761532 DOI: 10.1016/j.cell.2019.10.039] [Citation(s) in RCA: 218] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 09/20/2019] [Accepted: 10/29/2019] [Indexed: 12/11/2022]
Abstract
Non-coding regions amplified beyond oncogene borders have largely been ignored. Using a computational approach, we find signatures of significant co-amplification of non-coding DNA beyond the boundaries of amplified oncogenes across five cancer types. In glioblastoma, EGFR is preferentially co-amplified with its two endogenous enhancer elements active in the cell type of origin. These regulatory elements, their contacts, and their contribution to cell fitness are preserved on high-level circular extrachromosomal DNA amplifications. Interrogating the locus with a CRISPR interference screening approach reveals a diversity of additional elements that impact cell fitness. The pattern of fitness dependencies mirrors the rearrangement of regulatory elements and accompanying rewiring of the chromatin topology on the extrachromosomal amplicon. Our studies indicate that oncogene amplifications are shaped by regulatory dependencies in the non-coding genome.
Collapse
Affiliation(s)
- Andrew R Morton
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Nergiz Dogan-Artun
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Zachary J Faber
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Graham MacLeod
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Cynthia F Bartels
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Megan S Piazza
- Center for Human Genetics Laboratory, University Hospitals, Cleveland, OH 44106, USA
| | - Kevin C Allan
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA
| | - Stephen C Mack
- Department of Pediatrics, Division of Hematology and Oncology, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA
| | - Xiuxing Wang
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Ryan C Gimple
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA; Department of Pathology, Case Western Reserve University, Cleveland, OH 44120, USA
| | - Qiulian Wu
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Brian P Rubin
- Departments of Anatomic Pathology and Molecular Genetics, Cleveland Clinic, Lerner Research Institute and Taussig Cancer Center, Cleveland, OH 44195, USA
| | - Shashirekha Shetty
- Center for Human Genetics Laboratory, University Hospitals, Cleveland, OH 44106, USA
| | - Stephane Angers
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada; Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, ON M5G 0A4, Canada
| | - Peter B Dirks
- Developmental and Stem Cell Biology Program and Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | | | - Mathieu Lupien
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada; Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jeremy N Rich
- Department of Medicine, Division of Regenerative Medicine, University of California, San Diego, La Jolla, CA 92037, USA; Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA 92037, USA.
| | - Peter C Scacheri
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Case Comprehensive Cancer Center, Cleveland, OH 44106, USA.
| |
Collapse
|
25
|
The role of SOX18 in bladder cancer and its underlying mechanism in mediating cellular functions. Life Sci 2019; 232:116614. [DOI: 10.1016/j.lfs.2019.116614] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 06/20/2019] [Accepted: 06/27/2019] [Indexed: 12/14/2022]
|
26
|
Lu BS, Yin YW, Zhang YP, Guo PY, Li W, Liu KL. Upregulation of NPL4 promotes bladder cancer cell proliferation by inhibiting DXO destabilization of cyclin D1 mRNA. Cancer Cell Int 2019; 19:149. [PMID: 31164795 PMCID: PMC6543671 DOI: 10.1186/s12935-019-0874-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/27/2019] [Indexed: 12/11/2022] Open
Abstract
Background NPL4 is an important cofactor of the valosin-containing protein (VCP)–NPL4–UFD1 complex. The VCP–NPL4–UFD1 has been considered as a ubiquitin proteasome system (UPS) regulator and response to protein degradation. While NPL4 plays important roles in various diseases, little is known about its functions in bladder cancer (BC). Methods MTT assays and colony forming test were performed to evaluate cell proliferation ability and Western blotting was used to detect protein expression. Cyclin D1 mRNA expression was detected using qRT-PCR, and coimmunoprecipitation (CoIP) was used to detect protein–protein interactions. Results NPL4 was upregulated in BC tissue and correlated with poor prognosis. Upregulation of NPL4 promoted cell proliferation while suppression of NPL4 reduced BC cell proliferation. Upregulation of NPL4 led to overexpression of cyclin D1 by enhancing its mRNA stability. Moreover, NPL4 was found to bind directly to DXO and induce its degradation. DXO was downregulated in BC tissue and regulated BC cell proliferation by destabilizing cyclin D1 mRNA. DXO-mediated NPL4 regulated BC cell proliferation by stabilizing cyclin D1 expression. Conclusions The NPL4/DXO/cyclin D1 axis exert crucial role in BC cell growth and is associated with prognosis and may represent a potential therapeutic target for BC.
Collapse
Affiliation(s)
- Bao-Sai Lu
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000 Hebei People's Republic of China
| | - Yue-Wei Yin
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000 Hebei People's Republic of China
| | - Yan-Ping Zhang
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000 Hebei People's Republic of China
| | - Ping-Ying Guo
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000 Hebei People's Republic of China
| | - Wei Li
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000 Hebei People's Republic of China
| | - Kai-Long Liu
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000 Hebei People's Republic of China
| |
Collapse
|
27
|
Yang H, Chen R, Wang Q, Wei Q, Ji Y, Zheng G, Zhong X, Cox NJ, Li B. De novo pattern discovery enables robust assessment of functional consequences of non-coding variants. Bioinformatics 2019; 35:1453-1460. [PMID: 30256891 PMCID: PMC6499232 DOI: 10.1093/bioinformatics/bty826] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/17/2018] [Accepted: 09/25/2018] [Indexed: 12/13/2022] Open
Abstract
MOTIVATION Given the complexity of genome regions, prioritize the functional effects of non-coding variants remains a challenge. Although several frameworks have been proposed for the evaluation of the functionality of non-coding variants, most of them used 'black boxes' methods that simplify the task as the pathogenicity/benign classification problem, which ignores the distinct regulatory mechanisms of variants and leads to less desirable performance. In this study, we developed DVAR, an unsupervised framework that leverage various biochemical and evolutionary evidence to distinguish the gene regulatory categories of variants and assess their comprehensive functional impact simultaneously. RESULTS DVAR performed de novo pattern discovery in high-dimensional data and identified five regulatory clusters of non-coding variants. Leveraging the new insights into the multiple functional patterns, it measures both the between-class and the within-class functional implication of the variants to achieve accurate prioritization. Compared to other two-class learning methods, it showed improved performance in identification of clinically significant variants, fine-mapped GWAS variants, eQTLs and expression-modulating variants. Moreover, it has superior performance on disease causal variants verified by genome-editing (like CRISPR-Cas9), which could provide a pre-selection strategy for genome-editing technologies across the whole genome. Finally, evaluated in BioVU and UK Biobank, two large-scale DNA biobanks linked to complete electronic health records, DVAR demonstrated its effectiveness in prioritizing non-coding variants associated with medical phenotypes. AVAILABILITY AND IMPLEMENTATION The C++ and Python source codes, the pre-computed DVAR-cluster labels and DVAR-scores across the whole genome are available at https://www.vumc.org/cgg/dvar. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Hai Yang
- Department of Molecular Physiology & Biophysics, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
| | - Rui Chen
- Department of Molecular Physiology & Biophysics, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
| | - Quan Wang
- Department of Molecular Physiology & Biophysics, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
| | - Qiang Wei
- Department of Molecular Physiology & Biophysics, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
| | - Ying Ji
- Department of Molecular Physiology & Biophysics, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
| | - Guangze Zheng
- Department of Molecular Physiology & Biophysics, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
| | - Xue Zhong
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nancy J Cox
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bingshan Li
- Department of Molecular Physiology & Biophysics, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
28
|
Zhang Z, Luo G, Yu C, Yu G, Jiang R, Shi X. MicroRNA-493-5p inhibits proliferation and metastasis of osteosarcoma cells by targeting Kruppel-like factor 5. J Cell Physiol 2019; 234:13525-13533. [PMID: 30773645 DOI: 10.1002/jcp.28030] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 11/27/2018] [Indexed: 01/04/2023]
Abstract
Osteosarcoma, including spinal osteosarcoma, has properties of high degree of malignancy, high rate of recurrence, and high incidence of metastasis. microRNAs can exert oncogenic or tumor suppressive roles in cancer cells. This study explored the effects of microRNA-493-5p (miR-493-5p) on osteosarcoma cell viability, migration, invasion, and apoptosis, as well as the underlying possible mechanism. First, the expression of miR-493-5p in osteosarcoma tissues and cells was detected using quantitative reverse transcription polymerase chain reaction (qRT-PCR). Then, the effects of miR-493-5p overexpression (or suppression) on osteosarcoma cell viability, migration, invasion, and apoptosis, as well as Kruppel-like factor 5 (KLF5) expression, were assessed using the Cell Counting Kit-8 assay, two-chamber transwell assay, Annexin V-FITC/PI apoptosis detection kit, qRT-PCR, and western blotting, respectively. Finally, the roles of KLF5 in miR-493-5p suppression-induced U20S cell viability, migration, and invasion enhancement, as well as the PI3K/AKT pathway activation, were evaluated. We found that miR-493-5p had lower expression in tumor tissues of spinal osteosarcoma and osteosarcoma cells. Overexpression of miR-493-5p inhibited osteosarcoma U20S cell viability, migration, and invasion, but induced cell apoptosis. On the contrary, suppression of miR-493-5p-promoted U20S cell viability, migration, and invasion. KLF5 was a direct target gene of miR-493-5p, which participated in the effects of miR-493-5p on U20S cell viability, migration, invasion, and apoptosis. Furthermore, suppression of the miR-493-5p activated PI3K/AKT pathway in U20S cells by upregulating KLF5. In conclusion, we revealed that miR-493-5p exerted tumor suppressive roles in spinal osteosarcoma and osteosarcoma cells. Overexpression of miR-493-5p inhibited proliferation and metastasis of osteosarcoma cells by downregulating KLF5 and inactivating the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Zaiqing Zhang
- Department of Orthopaedics, Linyi Central Hospital, Yishui, China
| | - Gongzeng Luo
- Department of Orthopaedics, Linyi Central Hospital, Yishui, China
| | - Chuandong Yu
- Department of Orthopaedics, Heze Municipal Hospital, Heze, China
| | - Guisheng Yu
- Department of Orthopaedics, Heze Municipal Hospital, Heze, China
| | - Rui Jiang
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xuefeng Shi
- Department of Orthopedics, Trauma & Hand and Foot Surgery, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
29
|
de Maturana EL, Rava M, Anumudu C, Sáez O, Alonso D, Malats N. Bladder Cancer Genetic Susceptibility. A Systematic Review. Bladder Cancer 2018; 4:215-226. [PMID: 29732392 PMCID: PMC5929300 DOI: 10.3233/blc-170159] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Background: The variant/gene candidate approach to explore bladder cancer (BC) genetic susceptibility has been applied in many studies with significant findings reported. However, results are not always conclusive due to the lack of replication by subsequent studies. Objectives: To identify all epidemiological investigations on the genetic associations with BC risk, to quantify the likely magnitude of the associations by applying metaanalysis methodology and to assess whether there is a potential for publication/reporting bias. Methods: To address our aims, we have catalogued all genetic association studies published in the field of BC risk since 2000. Furthermore, we metaanalysed all polymorphisms with data available from at least three independent case-control studies with subjects of Caucasian origin analyzed under the same mode of inheritance. Results: The characterization of the genetic susceptibility of BC is composed of 28 variants, GWAS contributing most of them. Most of the significant variants associated with BC risk are located in genes belonging to chemical carcinogenesis, DNA repair, and cell cycle pathways. Causal relationship was also provided by functional analysis for GSTM1-null, NAT2-slow, APOBEC-rs1014971, CCNE1-rs8102137, SLC14A1-rs10775480, PSCA-rs2294008, UGT1A-rs1189203, and TP63-rs35592567. Conclusions: Genetic susceptibility of BC is still poorly defined, with GWAS contributing most of the strongest evidence. The systematic review did not provide evidence of further genetic associations. The potential public health translation of the existing knowledge on genetic susceptibility on BC is still limited.
Collapse
Affiliation(s)
| | - Marta Rava
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), and CIBERONC, Spain
| | - Chiaka Anumudu
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), and CIBERONC, Spain
| | - Olga Sáez
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), and CIBERONC, Spain
| | - Dolores Alonso
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), and CIBERONC, Spain
| | - Núria Malats
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre (CNIO), and CIBERONC, Spain
| |
Collapse
|