1
|
Rac1 as a Target to Treat Dysfunctions and Cancer of the Bladder. Biomedicines 2022; 10:biomedicines10061357. [PMID: 35740379 PMCID: PMC9219850 DOI: 10.3390/biomedicines10061357] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 12/28/2022] Open
Abstract
Bladder pathologies, very common in the aged population, have a considerable negative impact on quality of life. Novel targets are needed to design drugs and combinations to treat diseases such as overactive bladder and bladder cancers. A promising new target is the ubiquitous Rho GTPase Rac1, frequently dysregulated and overexpressed in bladder pathologies. We have analyzed the roles of Rac1 in different bladder pathologies, including bacterial infections, diabetes-induced bladder dysfunctions and bladder cancers. The contribution of the Rac1 protein to tumorigenesis, tumor progression, epithelial-mesenchymal transition of bladder cancer cells and their metastasis has been analyzed. Small molecules selectively targeting Rac1 have been discovered or designed, and two of them—NSC23766 and EHT 1864—have revealed activities against bladder cancer. Their mode of interaction with Rac1, at the GTP binding site or the guanine nucleotide exchange factors (GEF) interaction site, is discussed. Our analysis underlines the possibility of targeting Rac1 with small molecules with the objective to combat bladder dysfunctions and to reduce lower urinary tract symptoms. Finally, the interest of a Rac1 inhibitor to treat advanced chemoresistance prostate cancer, while reducing the risk of associated bladder dysfunction, is discussed. There is hope for a better management of bladder pathologies via Rac1-targeted approaches.
Collapse
|
2
|
Kim H, Momen-Heravi F, Chen S, Hoffmann P, Kebschull M, Papapanou PN. Differential DNA methylation and mRNA transcription in gingival tissues in periodontal health and disease. J Clin Periodontol 2021; 48:1152-1164. [PMID: 34101221 DOI: 10.1111/jcpe.13504] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 04/13/2021] [Accepted: 05/14/2021] [Indexed: 12/25/2022]
Abstract
AIM We investigated differential DNA methylation in gingival tissues in periodontal health, gingivitis, and periodontitis, and its association with differential mRNA expression. MATERIALS AND METHODS Gingival tissues were harvested from individuals and sites with clinically healthy and intact periodontium, gingivitis, and periodontitis. Samples were processed for differential DNA methylation and mRNA expression using the IlluminaEPIC (850 K) and the IlluminaHiSeq2000 platforms, respectively. Across the three phenotypes, we identified differentially methylated CpG sites and regions, differentially expressed genes (DEGs), and genes with concomitant differential methylation at their promoters and expression were identified. The findings were validated using our earlier databases using HG-U133Plus2.0Affymetrix microarrays and Illumina (450 K) methylation arrays. RESULTS We observed 43,631 differentially methylated positions (DMPs) between periodontitis and health, and 536 DMPs between gingivitis and health (FDR < 0.05). On the mRNA level, statistically significant DEGs were observed only between periodontitis and health (n = 126). Twelve DEGs between periodontitis and health (DCC, KCNA3, KCNA2, RIMS2, HOXB7, PNOC, IRX1, JSRP1, TBX1, OPCML, CECR1, SCN4B) were also differentially methylated between the two phenotypes. Spearman correlations between methylation and expression in the EPIC/mRNAseq dataset were largely replicated in the 450 K/Affymetrix datasets. CONCLUSIONS Concomitant study of DNA methylation and gene expression patterns may identify genes whose expression is epigenetically regulated in periodontitis.
Collapse
Affiliation(s)
- Hyunjin Kim
- Biomedical Informatics Shared Resource, Herbert Irving Comprehensive Cancer Center and Department of Systems Biology, Columbia University, New York, New York, USA.,Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Fatemeh Momen-Heravi
- Division of Periodontics, Section of Oral, Diagnostic and Rehabilitation Sciences, College of Dental Medicine, Columbia University, New York, New York, USA
| | - Steven Chen
- Division of Periodontics, Section of Oral, Diagnostic and Rehabilitation Sciences, College of Dental Medicine, Columbia University, New York, New York, USA
| | - Per Hoffmann
- Institute of Human Genetics, University of Bonn, Bonn, Germany
| | - Moritz Kebschull
- Division of Periodontics, Section of Oral, Diagnostic and Rehabilitation Sciences, College of Dental Medicine, Columbia University, New York, New York, USA.,School of Dentistry, Institute of Clinical Sciences, University of Birmingham, Birmingham, UK
| | - Panos N Papapanou
- Division of Periodontics, Section of Oral, Diagnostic and Rehabilitation Sciences, College of Dental Medicine, Columbia University, New York, New York, USA
| |
Collapse
|
3
|
Basic Hallmarks of Urothelial Cancer Unleashed in Primary Uroepithelium by Interference with the Epigenetic Master Regulator ODC1. Sci Rep 2020; 10:3808. [PMID: 32123240 PMCID: PMC7052216 DOI: 10.1038/s41598-020-60796-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 02/14/2020] [Indexed: 01/16/2023] Open
Abstract
Urothelial carcinoma (UC) is a common disease causing significant morbidity and mortality as well as considerable costs for health systems. Extensive aberrant methylation of DNA is broadly documented in early UC, contributing to genetic instability, altered gene expression and tumor progression. However the triggers initiating aberrant methylation are unknown. Recently we discovered that several genes encoding key enzymes of methyl group and polyamine metabolism, including Ornithine Decarboxylase 1 (ODC1), are affected by DNA methylation in early stage UC. In this study, we investigated the hypothesis that these epigenetic alterations act in a feed-forward fashion to promote aberrant DNA methylation in UC. We demonstrate that siRNA-mediated knockdown of ODC1 expression elicits genome-wide LINE-1 demethylation, induction of LINE-1 transcripts and double-strand DNA breaks and decreases viability in primary cultured uroepithelial cells. Similarly, following siRNA-mediated knockdown of ODC1, UC cells undergo double-strand DNA breaks and apoptosis. Collectively, our findings provide evidence that ODC1 gene hypermethylation could be a starting point for the onset of genome-wide epigenetic aberrations in urothelial carcinogenesis. Furthermore, LINE-1 induction enabled by ODC1 interference provides a new experimental model to study mechanisms and consequences of LINE-1 activation in the etiology and progression of UC as well as presumably other cancers.
Collapse
|
4
|
Ding D, Liu Z, Zhao L, Geng H, Liang Z, Yu D. Role of PI3K/Akt pathway in Benzidine-induced proliferation in SV-40 immortalized human uroepithelial cell. Transl Cancer Res 2019; 8:1301-1310. [PMID: 35116872 PMCID: PMC8798037 DOI: 10.21037/tcr.2019.07.14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 06/24/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND Long term exposure to benzidine has been determined as a cause of urothelial carcinoma. But how it works in the process of cell proliferation that involves in tumor growth is not examined yet. In the current research, the effect of PI3K/Akt on cell proliferation mediated by benzidine was confirmed. METHODS The immortalized SV-40 human uroepithelial cells (SV-HUC-1) had been subjected to 6 days of benzidine treatment at various contents, then MTT assay, together with subsequent flow cytometry assay were used for observing effects on cell proliferation. Further Western blots were used to detect the expression of total-Akt, phospho-Akt and specific proteins of cell cycle. The Akt, Cyclin D1, PCNA and P21 mRNA levels were detected through RT-PCR. In addition, the blocker-LY294002 was used to cut down the PI3K/Akt signaling pathway. And then those parameters were detected using the same methods as above. RESULTS Results showed that benzidine acted to induce cell proliferation at low doses (P<0.05 vs. controls) via MTT and flow cytometry assay. The expression of phospho-Akt, Cyclin D1, and PCNA were significantly enhanced compared with that of control (P<0.05; P<0.01), but total-Akt and P21 levels were reduced. Whereas, inhibitor of PI3K/Akt suppressed the proliferating procedure when cells were treated with the blocker (LY294002) and also inhibited the expression of related cycle proteins. CONCLUSIONS Activated PI3K/Akt signal pathway promotes benzidine-triggered cell proliferation. It may shed light on the molecular mechanisms that the activated PI3K/Akt pathway promotes benzidine-triggered cell proliferation and intervention of its target.
Collapse
Affiliation(s)
- Demao Ding
- Department of Urologic Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Zhiqi Liu
- Department of Urologic Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Li Zhao
- Department of Urologic Surgery, Nanjing Jiangbei People’s Hospital, Nanjing 210048, China
| | - Hao Geng
- Department of Urologic Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| | - Zhaofeng Liang
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Dexin Yu
- Department of Urologic Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei 230032, China
| |
Collapse
|
5
|
Roy NK, Monisha J, Padmavathi G, Lalhruaitluanga H, Kumar NS, Singh AK, Bordoloi D, Baruah MN, Ahmed GN, Longkumar I, Arfuso F, Kumar AP, Kunnumakkara AB. Isoform-Specific Role of Akt in Oral Squamous Cell Carcinoma. Biomolecules 2019; 9:E253. [PMID: 31252679 PMCID: PMC6681224 DOI: 10.3390/biom9070253] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 06/05/2019] [Accepted: 06/22/2019] [Indexed: 12/15/2022] Open
Abstract
Protein kinase B (Akt) plays a very significant role in various cancers including oral cancer. However, it has three isoforms (Akt1, Akt2, and Akt3) and they perform distinct functions and even play contrasting roles in different cancers. Therefore, it becomes essential to evaluate the isoform-specific role of Akt in oral cancer. In the present study, an attempt has been made to elucidate the isoform-specific role of Akt in oral cancer. The immunohistochemical analysis of oral cancer tissues showed an overexpression of Akt1 and 2 isoforms but not Akt3. Moreover, the dataset of "The Cancer Genome Atlas" for head and neck cancer has suggested the genetic alterations of Akt1 and 2 tend to be associated with the utmost poor clinical outcome in oral cancer. Further, treatment of oral cancer cells with tobacco and its components such as benzo(a)pyrene and nicotine caused increased mRNA levels of Akt1 and 2 isoforms and also enhanced the aggressiveness of oral cancer cells in terms of proliferation, and clonogenic and migration potential. Finally, silencing of Akt1 and 2 isoforms caused decreased cell survival and induced cell cycle arrest at the G2/M phase. Akt1/2 silencing also reduced tobacco-induced aggressiveness by decreasing the clonogenic and migration potential of oral cancer cells. Moreover, silencing of Akt1 and 2 isoforms was found to decrease the expression of proteins regulating cancer cell survival and proliferation such as cyclooxygenase-2, B-cell lymphoma 2 (Bcl-2), cyclin D1, and survivin. Thus, the important role of Akt1 and 2 isoforms have been elucidated in oral cancer with in-depth mechanistic analysis.
Collapse
Affiliation(s)
- Nand Kishor Roy
- Cancer Biology Laboratory & DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Javadi Monisha
- Cancer Biology Laboratory & DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Ganesan Padmavathi
- Cancer Biology Laboratory & DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - H Lalhruaitluanga
- Department of Biotechnology, Mizoram University, Aizawl, Mizoram 796 004, India
| | | | - Anuj Kumar Singh
- Cancer Biology Laboratory & DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Devivasha Bordoloi
- Cancer Biology Laboratory & DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | | | - Gazi Naseem Ahmed
- North-East Cancer Hospital and Research Institute, Guwahati, Assam 781023, India
| | - Imliwati Longkumar
- North-East Cancer Hospital and Research Institute, Guwahati, Assam 781023, India
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6009, Australia
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
- Medical Science Cluster, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Medical School, Faculty of Health Sciences, Curtin University, Perth, WA 6845, Australia
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory & DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India.
| |
Collapse
|
6
|
Fragou D, Pakkidi E, Aschner M, Samanidou V, Kovatsi L. Smoking and DNA methylation: Correlation of methylation with smoking behavior and association with diseases and fetus development following prenatal exposure. Food Chem Toxicol 2019; 129:312-327. [PMID: 31063835 DOI: 10.1016/j.fct.2019.04.059] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 04/29/2019] [Accepted: 04/30/2019] [Indexed: 12/13/2022]
Abstract
Among epigenetic mechanisms, DNA methylation has been widely studied with respect to many environmental factors. Smoking is a common factor which affects both global and gene-specific DNA methylation. It is supported that smoking directly affects DNA methylation, and these effects contribute to the development and progression of various diseases, such as cancer, lung and cardiovascular diseases and male infertility. In addition, prenatal smoking influences the normal development of the fetus via DNA methylation changes. The DNA methylation profile and its smoking-induced alterations helps to distinguish current from former smokers and non-smokers and can be used to predict the risk for the development of a disease. This review summarizes the DNA methylation changes induced by smoking, their correlation with smoking behavior and their association with various diseases and fetus development.
Collapse
Affiliation(s)
- Domniki Fragou
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, Greece
| | - Eleni Pakkidi
- Laboratory of Analytical Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, Greece
| | - Michael Aschner
- Departments of Molecular Pharmacology, Neuroscience, and Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Victoria Samanidou
- Laboratory of Analytical Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, Greece
| | - Leda Kovatsi
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, Greece.
| |
Collapse
|
7
|
Tsay MD, Hsieh MJ, Wang SS, Wang WC, Chou YY, Shih CH, Yang SF, Chou YE. Impact of endothelial nitric oxide synthase polymorphisms on urothelial cell carcinoma development. Urol Oncol 2019; 37:293.e1-293.e9. [PMID: 30611644 DOI: 10.1016/j.urolonc.2018.12.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 12/03/2018] [Accepted: 12/22/2018] [Indexed: 01/04/2023]
Abstract
OBJECTIVES Urothelial cell carcinoma (UCC), a major malignancy of the genitourinary tract, is induced through carcinogenic etiological factors. Endothelial nitric oxide synthase (eNOS) is one of the major isoforms of nitric oxide synthase and is involved in various pathophysiologic and physiologic processes. In this study, eNOS single-nucleotide polymorphisms were investigated to evaluate UCC susceptibility and clinicopathological characteristics. MATERIALS AND METHODS Two single-nucleotide polymorphisms of eNOS in 431 patients with UCC and 862 controls without cancer were analyzed using real-time polymerase chain reaction. RESULTS The results showed that 272 men with UCC having eNOS 894 G > T rs1799983 "GT + TT" variants had a high risk of developing a large tumor (T1-T4, P = 0.038). Furthermore, a correlation was observed between the expressions of eNOS and invasive tumor, metastasis and poor survival in urothelial carcinoma in The Cancer Genome Atlas data set. CONCLUSION Our results indicated that male patients with UCC carrying eNOS 894 G > T rs1799983 "GT + TT" genetic variants have a high risk of developing a large tumor, and eNOS polymorphisms may serve as a marker or therapeutic target in UCC treatment.
Collapse
Affiliation(s)
- Ming-Dow Tsay
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Family medicine, Tungs' Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Ming-Ju Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Shian-Shiang Wang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Wen-Chen Wang
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Ya-Yi Chou
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Chen-Ho Shih
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ying-Erh Chou
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW Epigenetics refers to processes that alter gene expression without altering primary DNA. Over that past decade, there is a growing focus on epigenetic mechanisms in cancer research and its importance in cancer biology. This review summarizes epigenetic dysregulation in bladder cancer. RECENT FINDINGS Epigenetic alterations are overall shared across various grades and stages of bladder cancer. High grade invasive tumors demonstrate a greater degree and intensity of methylation and may have a unique methylation pattern. Environmental exposures may influence epigenetic alterations directly independent of genomic change. Non-coding RNAs play an important role in cancer phenotype, especially in the context of integrative genomic analyses. DNA hypermethylation and non-coding RNAs have potential as robust bladder cancer biomarkers; however, they require further study and validation. Changes in chromatin and histone modification are attractive targets for therapy and are currently in clinical trials. Epigenetic dysregulation may be an important key in improving the understanding of bladder cancer pathogenesis, especially through integrative genomic analyses. Deeper understanding of these pathways can help identify clinically relevant biomarkers and therapeutic targets to validate for diagnosis, monitoring, prognosis, and treatment for bladder cancer.
Collapse
Affiliation(s)
- Sima P Porten
- Department of Urology, University of California San Francisco (UCSF), Mailbox Code 1695, 550 16th Street, 6th Floor, San Francisco, CA, 94143, USA.
| |
Collapse
|
9
|
MicroRNAs in Smoking-Related Carcinogenesis: Biomarkers, Functions, and Therapy. J Clin Med 2018; 7:jcm7050098. [PMID: 29723992 PMCID: PMC5977137 DOI: 10.3390/jcm7050098] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 04/21/2018] [Accepted: 04/25/2018] [Indexed: 12/11/2022] Open
Abstract
Long-term heavy cigarette smoking is a well-known high-risk factor for carcinogenesis in various organs such as the head and neck, lungs, and urinary bladder. Furthermore, cigarette smoking can systemically accelerate aging, and as the result, promoting carcinogenesis via changing the host microenvironment. Various inflammatory factors, hormones, and chemical mediators induced by smoking mediate carcinoma-related molecules and induce carcinogenesis. MicroRNAs (miRNAs) are a family of short noncoding RNA molecules that bind to mRNAs and inhibit their expression. Cigarette smoke induces the expression of various miRNAs, many of which are known to function in the post-transcriptional silencing of anticancer molecules, thereby leading to smoking-induced carcinogenesis. Analysis of expression profiles of smoking-induced miRNAs can help identify biomarkers for the diagnosis and prognosis of smoking-related cancers and prediction of therapeutic responses, as well as revealing promising therapeutic targets. Here, we introduce the most recent and useful findings of miRNA analyses focused on lung cancer and urinary bladder cancer, which are strongly associated with cigarette smoking, and discuss the utility of miRNAs as clinical biomarkers.
Collapse
|
10
|
Falzone L, Candido S, Salemi R, Basile MS, Scalisi A, McCubrey JA, Torino F, Signorelli SS, Montella M, Libra M. Computational identification of microRNAs associated to both epithelial to mesenchymal transition and NGAL/MMP-9 pathways in bladder cancer. Oncotarget 2018; 7:72758-72766. [PMID: 27602581 PMCID: PMC5341942 DOI: 10.18632/oncotarget.11805] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 08/25/2016] [Indexed: 12/20/2022] Open
Abstract
Bladder cancer is one of the leading cancer of the urinary tract. It is often diagnosed at advanced stage of the disease. To date, no specific and effective early detection biomarkers are available. Cancer development and progression are associated with the involvement of both epithelial-mesenchymal transition (EMT) and tumor microenvironment of which NGAL/MMP-9 complex represents the main player in bladder cancer. It is known that change in microRNAs (miRNAs) expression may result in gene modulation. Therefore, the identification of specific miRNAs associated with EMT pathway and NGAL/MMP-9 complex may be useful to detect the development of bladder cancer at early stages. On this ground, the expression levels of miRNAs in public available datasets of bladder cancer containing data of non-coding RNA profiling was evaluated. This analysis revealed a group of 16 miRNAs differentially expressed between bladder cancer patients and related healthy controls. By miRNA prediction tool (mirDIP), the relationship between the identified miRNAs and the EMT genes was established. Using the DIANA-mirPath (v.2) software, miRNAs, able to modulate the expression of NGAL and MMP-9 genes, were recognized. The results of this study provide evidence that the downregulated hsa-miR-145-5p and hsa-miR-214-3p may modulate the expression of both EMT and NGAL/MMP-9 pathways. Therefore, further validation analyses may confirm the usefulness of these selected miRNAs for predicting the development of bladder cancer at the early stage of the disease.
Collapse
Affiliation(s)
- Luca Falzone
- Department of Biomedical and Biotechnological Sciences, Laboratory of Translational Oncology and Functional Genomics, Section of General and Clinical Pathology and Oncology, University of Catania, Catania, Italy
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences, Laboratory of Translational Oncology and Functional Genomics, Section of General and Clinical Pathology and Oncology, University of Catania, Catania, Italy
| | - Rossella Salemi
- Department of Biomedical and Biotechnological Sciences, Laboratory of Translational Oncology and Functional Genomics, Section of General and Clinical Pathology and Oncology, University of Catania, Catania, Italy
| | - Maria S Basile
- Department of Biomedical and Biotechnological Sciences, Laboratory of Translational Oncology and Functional Genomics, Section of General and Clinical Pathology and Oncology, University of Catania, Catania, Italy
| | | | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Francesco Torino
- Department of Systems Medicine, Chair of Medical Oncology, Tor Vergata University of Rome, Rome, Italy
| | - Salvatore S Signorelli
- Department of Clinical and Experimental Medicine, University of Catania, Medical Angiology Unit, Garibaldi Hospital, Catania, Italy
| | - Maurizio Montella
- Unit of Epidemiology, 'Fondazione G. Pascale', Istituto Nazionale Tumori, Naples, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, Laboratory of Translational Oncology and Functional Genomics, Section of General and Clinical Pathology and Oncology, University of Catania, Catania, Italy
| |
Collapse
|
11
|
Erichsen L, Ghanjati F, Beermann A, Poyet C, Hermanns T, Schulz WA, Seifert HH, Wild PJ, Buser L, Kröning A, Braunstein S, Anlauf M, Jankowiak S, Hassan M, Bendhack ML, Araúzo-Bravo MJ, Santourlidis S. Aberrant methylated key genes of methyl group metabolism within the molecular etiology of urothelial carcinogenesis. Sci Rep 2018; 8:3477. [PMID: 29472622 PMCID: PMC5823913 DOI: 10.1038/s41598-018-21932-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 02/12/2018] [Indexed: 12/14/2022] Open
Abstract
Urothelial carcinoma (UC), the most common cancer of the urinary bladder causes severe morbidity and mortality, e.g. about 40.000 deaths in the EU annually, and incurs considerable costs for the health system due to the need for prolonged treatments and long-term monitoring. Extensive aberrant DNA methylation is described to prevail in urothelial carcinoma and is thought to contribute to genetic instability, altered gene expression and tumor progression. However, it is unknown how this epigenetic alteration arises during carcinogenesis. Intact methyl group metabolism is required to ensure maintenance of cell-type specific methylomes and thereby genetic integrity and proper cellular function. Here, using two independent techniques for detecting DNA methylation, we observed DNA hypermethylation of the 5'-regulatory regions of the key methyl group metabolism genes ODC1, AHCY and MTHFR in early urothelial carcinoma. These hypermethylation events are associated with genome-wide DNA hypomethylation which is commonly associated with genetic instability. We therefore infer that hypermethylation of methyl group metabolism genes acts in a feed-forward cycle to promote additional DNA methylation changes and suggest a new hypothesis on the molecular etiology of urothelial carcinoma.
Collapse
Affiliation(s)
- Lars Erichsen
- Epigenetics Core Laboratory, Institute of Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich-Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Foued Ghanjati
- Epigenetics Core Laboratory, Institute of Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich-Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Agnes Beermann
- Epigenetics Core Laboratory, Institute of Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich-Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Cedric Poyet
- Department of Urology, University Hospital, University of Zurich, Zurich, Switzerland
| | - Thomas Hermanns
- Department of Urology, University Hospital, University of Zurich, Zurich, Switzerland
| | - Wolfgang A Schulz
- Department of Urology, Medical Faculty, Heinrich-Heine University Duesseldorf, Duesseldorf, Germany
| | | | - Peter J Wild
- Institute of Surgical Pathology, University Hospital, University of Zurich, 8091, Zurich, Switzerland
| | - Lorenz Buser
- Institute of Surgical Pathology, University Hospital, University of Zurich, 8091, Zurich, Switzerland
| | - Alexander Kröning
- Epigenetics Core Laboratory, Institute of Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich-Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Stefan Braunstein
- Department of Pathology, Medical Faculty, Heinrich-Heine University Duesseldorf, Duesseldorf, Germany
| | - Martin Anlauf
- Department of Pathology, Medical Faculty, Heinrich-Heine University Duesseldorf, Duesseldorf, Germany
| | - Silvia Jankowiak
- Department of Pathology, Medical Faculty, Heinrich-Heine University Duesseldorf, Duesseldorf, Germany
| | - Mohamed Hassan
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, 70112, USA
- Institut National de la Santé et de la Recherché Médicale, University of Strasbourg, 67000, Strasbourg, France
| | - Marcelo L Bendhack
- Department of Urology, University Hospital, Positivo University, Curitiba, Brazil
| | - Marcos J Araúzo-Bravo
- Group of Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, 20014, San Sebastián, Spain
- IKERBASQUE, Basque Foundation for Science, 48009, Bilbao, Spain
| | - Simeon Santourlidis
- Epigenetics Core Laboratory, Institute of Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich-Heine University Duesseldorf, Moorenstr. 5, 40225, Duesseldorf, Germany.
| |
Collapse
|
12
|
Lee HL, Chiou HL, Wang SS, Hung SC, Chou MC, Yang SF, Hsieh MJ, Chou YE. WISP1 genetic variants as predictors of tumor development with urothelial cell carcinoma. Urol Oncol 2017; 36:160.e15-160.e21. [PMID: 29277583 DOI: 10.1016/j.urolonc.2017.11.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/11/2017] [Accepted: 11/30/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Urothelial cell carcinoma (UCC) of the urinary bladder is a major malignancy of the genitourinary tract. Etiological factors, such as the environment, ethnicity, genetics, and diet, contribute to UCC carcinogenesis. WNT1-inducible signaling pathway protein 1 (WISP1), also known as CCN4, a cysteine-rich protein belonging to the Cyr61, CTGF, Nov (CCN) family of matricellular proteins, has many developmental functions and might be involved in carcinogenesis. This study investigated WISP1 single-nucleotide polymorphisms to evaluate UCC susceptibility and clinicopathological characteristics. MATERIALS AND METHODS Real-time polymerase chain reaction was used to analyze 4 single-nucleotide polymorphisms of WISP1 in 369 patients with UCC and 738 controls without cancer. RESULTS The results showed that in 128 women with UCC who carried WISP1 rs2929973 (AG + GG) variants had a higher risk of developing an advanced muscle-invasive tumor stage (pT2-pT4, P = 0.007) and a large tumor (T1-T4, P = 0.030). Further analyses revealed that a correlation between the expressions of WISP1 and invasive tumor and large tumor size in urothelial carcinoma was observed in the TCGA (The Cancer Genome Atlas) dataset. CONCLUSIONS Our results indicated that patients with UCC carrying rs2977530 genetic variants (AG + GG) have a higher risk of developing a more invasive tumor stage and a large tumor. WISP1 polymorphisms may serve as a marker or a therapeutic target in UCC.
Collapse
Affiliation(s)
- Hsiang-Lin Lee
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Hui-Ling Chiou
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan; Department of Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shian-Shiang Wang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Sheng-Chun Hung
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Ming-Chih Chou
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ming-Ju Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
| | - Ying-Erh Chou
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
13
|
Promoter hypermethylation of LGALS4 correlates with poor prognosis in patients with urothelial carcinoma. Oncotarget 2017; 8:23787-23802. [PMID: 28423602 PMCID: PMC5410344 DOI: 10.18632/oncotarget.15865] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 02/13/2017] [Indexed: 01/09/2023] Open
Abstract
Galectine-4 (gal-4), encoded by the LGALS4 gene, was recently shown to exhibit a tumor suppressive effect in colorectal carcinoma and pancreatic adenocarcinoma, although how the expression of this gene is regulated remains unknown. No reports describe the significance of gal-4 in the malignant potential of urothelial tumors. Thus, we analyzed LGALS4 methylation and gene expression and their clinical relevance and biological function in urothelial carcinoma (UC). LGALS4 methylation was initially identified as a progression biomarker for UC patients through genome-wide DNA methylation profiling of 16 tumor samples. Bisulfite sequencing PCR and immunohistochemistry were performed to validate the promoter methylation and expression of LGALS4. We used quantitative methylation-specific PCR to determine the methylation levels of LGALS4 normalized to ACTB in the tumor samples of 79 UC patients and compared the levels between patients with different clinicopathological characteristics. The association with survival probability was analyzed with the Kaplan-Meier method and Cox regression analysis. The ectopic expression of gal-4 in cancer cell lines was used to address its biological function in UC in vitro. The promoter hypermethylation of LGALS4 (>2.51, log10 scale) revealed a positive correlation with high levels of both histological grade and tumor T category and with lymph node metastasis (all P≤0.001). In addition, LGALS4 hypermethylation was an independent predictor of inferior survival in UC patients (P<0.05). The ectopic expression studies demonstrated that gal-4 suppressed urothelial cancer cell growth, migration, and invasion. Thus, LGALS4 may function as a tumor suppressor gene in UC progression. Our findings provide evidence that methylation-mediated LGALS4 gene repression may be involved in urothelial tumor progression.
Collapse
|
14
|
Hoque MO. Differences in the Molecular Characteristics of Bladder Cancer between Smokers and Nonsmokers. Eur Urol Focus 2017; 4:98-99. [PMID: 28753779 DOI: 10.1016/j.euf.2016.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 07/09/2016] [Indexed: 11/30/2022]
Affiliation(s)
- Mohammad Obaidul Hoque
- Department of Otolaryngology and Head and Neck Surgery, Urology, and Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
15
|
Association of Smoking, Alcohol Use, and Betel Quid Chewing with Epigenetic Aberrations in Cancers. Int J Mol Sci 2017; 18:ijms18061210. [PMID: 28587272 PMCID: PMC5486033 DOI: 10.3390/ijms18061210] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 05/26/2017] [Accepted: 06/02/2017] [Indexed: 12/16/2022] Open
Abstract
Numerous environmental factors such as diet, alcohol use, stress, and environmental chemicals are known to elicit epigenetic changes, leading to increased rates of cancers and other diseases. The incidence of head and neck cancer, one of the most common cancers in Taiwanese males, is increasing: oral cancer and nasopharyngeal carcinoma are ranked fourth and tenth respectively, among the top ten cancers in this group, and a major cause of cancer-related deaths in Taiwanese males. Previous studies have identified smoking, alcohol use, and betel quid chewing as the three major causes of head and neck cancers; these three social habits are commonly observed in Taiwanese males, resulting in an increasing morbidity rate of head and neck cancers in this population. In this literature review, we discuss the association between specific components of betel quid, alcohol, and tobacco, and the occurrence of head and neck cancers, lung cancer, gastrointestinal cancers, and urethral cancer. We focus on regulatory mechanisms at the epigenetic level and their oncogenic effects. The review further discusses the application of FDA-approved epigenetic drugs as therapeutic strategies against cancer.
Collapse
|
16
|
Brait M, Banerjee M, Maldonado L, Ooki A, Loyo M, Guida E, Izumchenko E, Mangold L, Humphreys E, Rosenbaum E, Partin A, Sidransky D, Hoque MO. Promoter methylation of MCAM, ERα and ERβ in serum of early stage prostate cancer patients. Oncotarget 2017; 8:15431-15440. [PMID: 28147335 PMCID: PMC5362497 DOI: 10.18632/oncotarget.14873] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 12/31/2016] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Prostate cancer (PC) is the second most common cancer among men worldwide. Currently, the most common non-invasive approach for screening and risk assessment of PC is measuring the level of serum prostate-specific antigen (PSA). However, the sensitivity of PSA is 42.8 % and specificity is 41.1%. As a result, the serum PSA test leads to numerous unneeded biopsies. Therefore, a rigorous search for biomarkers for early detection of PC is ongoing. In this study, we aim to assess a panel of epigenetic markers in an intend to develop an early detection test for PC. RESULTS The sensitivity and specificity of hypermethylation of MCAM was 66% and 73% respectively which is an improvement from the sensitivity and specificity of PSA. Considering a combination marker panel of MCAM, ERα and ERβ increased the sensitivity to 75% and the specificity became 70% for the minimally invasive early detection test of PC. MATERIALS AND METHODS Sixteen primary matched tumor and serum were analyzed by quantitative methylation specific PCR (QMSP) to determine analytical and clinical sensitivity of the genes tested (SSBP2, MCAM, ERα, ERβ, APC, CCND2, MGMT, GSTP1, p16 and RARβ2). Additionally, serum samples from eighty four cases of PC, thirty controls and seven cases diagnosed as high grade Prostatic Intraepithelial Neoplasia (HGPIN) were analyzed. CONCLUSIONS Promoter methylation of MCAM, ERα and ERβ have a potential to be utilized as biomarker for the early detection of prostate PC as their sensitivity and specificity seem to be better than serum PSA in our cohort of samples. After robust validation in a larger prospective cohort, our findings may reduce the numbers of unwarranted prostate biopsies.
Collapse
Affiliation(s)
- Mariana Brait
- Department of Otolaryngology and Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mithu Banerjee
- Department of Otolaryngology and Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Leonel Maldonado
- Department of Otolaryngology and Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, University of South Alabama Medical Center, Mobile, Alabama, USA
| | - Akira Ooki
- Department of Otolaryngology and Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Myriam Loyo
- Department of Otolaryngology and Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Elisa Guida
- Department of Otolaryngology and Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Evgeny Izumchenko
- Department of Otolaryngology and Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Leslie Mangold
- Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Elizabeth Humphreys
- Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Eli Rosenbaum
- Department of Urological Oncology, Davidoff Center, Beilinson Hospital, Eliahu Hakim, Ramat Aviv, Israel
| | - Alan Partin
- Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - David Sidransky
- Department of Otolaryngology and Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mohammad Obaidul Hoque
- Department of Otolaryngology and Head and Neck Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
17
|
Glaser AP, Fantini D, Shilatifard A, Schaeffer EM, Meeks JJ. The evolving genomic landscape of urothelial carcinoma. Nat Rev Urol 2017; 14:215-229. [PMID: 28169993 DOI: 10.1038/nrurol.2017.11] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Survival of patients with urothelial carcinoma (including bladder cancer and upper tract urothelial carcinoma) is limited by our current approaches to staging, surgery, and chemotherapy. Large-scale, next-generation sequencing collaborations, such as The Cancer Genome Atlas, have already identified drivers and vulnerabilities of urothelial carcinoma. This disease has a high degree of mutational heterogeneity and a high frequency of somatic mutations compared with other solid tumours, potentially resulting in an increased neoantigen burden. Mutational heterogeneity is mediated by multiple factors including the apolipoprotein B mRNA editing enzyme catalytic polypeptide family of enzymes, smoking exposure, viral integrations, and intragene and intergene fusion proteins. The mutational landscape of urothelial carcinoma, including specific mutations in pathways and driver genes, such as FGFR3, ERBB2, PIK3CA, TP53, and STAG2, affects tumour aggressiveness and response to therapy. The next generation of therapies for urothelial carcinoma will be based on patient-specific targetable mutations found in individual tumours. This personalized-medicine approach to urothelial carcinoma has already resulted in unique clinical trial design and has the potential to improve patient outcomes and survival.
Collapse
Affiliation(s)
- Alexander P Glaser
- Northwestern University, Department of Urology, 303 E. Chicago Avenue, Tarry 16-703, Chicago, Illinois 60611, USA
| | - Damiano Fantini
- Northwestern University, Department of Urology, 303 E. Chicago Avenue, Tarry 16-703, Chicago, Illinois 60611, USA
| | - Ali Shilatifard
- Northwestern University, Department of Urology, 303 E. Chicago Avenue, Tarry 16-703, Chicago, Illinois 60611, USA
| | - Edward M Schaeffer
- Northwestern University, Department of Urology, 303 E. Chicago Avenue, Tarry 16-703, Chicago, Illinois 60611, USA
| | - Joshua J Meeks
- Northwestern University, Department of Urology, 303 E. Chicago Avenue, Tarry 16-703, Chicago, Illinois 60611, USA
| |
Collapse
|
18
|
Abstract
DNA methylation alterations are common in urothelial carcinoma, a prevalent cancer worldwide caused predominantly by chemical carcinogens. Recent studies have proposed sets of hypermethylated genes as promising diagnostic and prognostic biomarkers from urine or tissue samples, which require validation. Other studies have revealed intriguing links between specific carcinogens and DNA methylation alterations in cancer tissues or blood that might clarify carcinogenesis mechanisms and aid prevention. Like DNA methylation alterations, mutations in chromatin regulators are frequent, underlining the importance of epigenetic changes. However, the relations between the two changes and their functions in urothelial carcinogenesis remain unclear. Transcription factor genes with altered methylation deserve particular interest. Elucidating the functional impact of methylation changes is a prerequisite for their therapeutic targeting.
Collapse
Affiliation(s)
- Wolfgang A Schulz
- Department of Urology, Medical Faculty, Heinrich Heine University Duesseldorf, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Wolfgang Goering
- Department of Pathology, Medical Faculty, Heinrich Heine University Duesseldorf, Germany
| |
Collapse
|
19
|
Lu L, Xu H, Luo F, Liu X, Lu X, Yang Q, Xue J, Chen C, Shi L, Liu Q. Epigenetic silencing of miR-218 by the lncRNA CCAT1, acting via BMI1, promotes an altered cell cycle transition in the malignant transformation of HBE cells induced by cigarette smoke extract. Toxicol Appl Pharmacol 2016; 304:30-41. [PMID: 27212446 DOI: 10.1016/j.taap.2016.05.012] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 05/02/2016] [Accepted: 05/18/2016] [Indexed: 01/17/2023]
Abstract
Cigarette smoking is the strongest risk factor for the development of lung cancer, the leading cause of cancer-related deaths. However, the molecular mechanisms leading to lung cancer are largely unknown. A long-noncoding RNA (lncRNA), CCAT1, regarded as cancer-associated, has been investigated extensively. Moreover, the molecular mechanisms of lncRNAs in regulation of microRNAs (miRNAs) induced by cigarette smoke remain unclear. In the present investigation, cigarette smoke extract (CSE) caused an altered cell cycle and increased CCAT1 levels and decreased miR-218 levels in human bronchial epithelial (HBE) cells. Depletion of CCAT1 attenuated the CSE-induced decreases of miR-218 levels, suggesting that miR-218 is negatively regulated by CCAT1 in HBE cells exposed to CSE. The CSE-induced increases of BMI1 levels and blocked by CCAT1 siRNA were attenuated by an miR-218 inhibitor. Moreover, in CSE-transformed HBE cells, the CSE-induced cell cycle changes and elevated neoplastic capacity were reversed by CCAT1 siRNA or BMI1 siRNA. This epigenetic silencing of miR-218 by CCAT1 induces an altered cell cycle transition through BMI1 and provides a new mechanism for CSE-induced lung carcinogenesis.
Collapse
Affiliation(s)
- Lu Lu
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Hui Xu
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Fei Luo
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Xinlu Liu
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Xiaolin Lu
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Qianlei Yang
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Junchao Xue
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Chao Chen
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Le Shi
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Qizhan Liu
- Institute of Toxicology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China; The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China.
| |
Collapse
|
20
|
Delitto D, Zhang D, Han S, Black BS, Knowlton AE, Vlada AC, Sarosi GA, Behrns KE, Thomas RM, Lu X, Liu C, George TJ, Hughes SJ, Wallet SM, Trevino JG. Nicotine Reduces Survival via Augmentation of Paracrine HGF-MET Signaling in the Pancreatic Cancer Microenvironment. Clin Cancer Res 2016; 22:1787-1799. [PMID: 26667487 PMCID: PMC4818664 DOI: 10.1158/1078-0432.ccr-15-1256] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Accepted: 11/26/2015] [Indexed: 01/18/2023]
Abstract
PURPOSE The relationship between smoking and pancreatic cancer biology, particularly in the context of the heterogeneous microenvironment, remains incompletely defined. We hypothesized that nicotine exposure would lead to the augmentation of paracrine growth factor signaling between tumor-associated stroma (TAS) and pancreatic cancer cells, ultimately resulting in accelerated tumor growth and metastasis. EXPERIMENTAL DESIGN The effect of tobacco use on overall survival was analyzed using a prospectively maintained database of surgically resected patients with pancreatic cancer. Nicotine exposure was evaluated in vitro using primary patient-derived TAS and pancreatic cancer cells independently and in coculture. Nicotine administration was then assessed in vivo using a patient-derived pancreatic cancer xenograft model. RESULTS Continued smoking was associated with reduced overall survival after surgical resection. In culture, nicotine-stimulated hepatocyte growth factor (HGF) secretion in primary patient-derived TAS and nicotine stimulation was required for persistent pancreatic cancer cell c-Met activation in a coculture model. c-Met activation in this manner led to the induction of inhibitor of differentiation-1 (Id1) in pancreatic cancer cells, previously established as a mediator of growth, invasion and chemoresistance. HGF-induced Id1 expression was abrogated by both epigenetic and pharmacologic c-Met inhibition. In patient-derived pancreatic cancer xenografts, nicotine treatment augmented tumor growth and metastasis; tumor lysates from nicotine-treated mice demonstrated elevated HGF expression by qRT-PCR and phospho-Met levels by ELISA. Similarly, elevated levels of phospho-Met in surgically resected pancreatic cancer specimens correlated with reduced overall survival. CONCLUSIONS Taken together, these data demonstrate a novel, microenvironment-dependent paracrine signaling mechanism by which nicotine exposure promotes the growth and metastasis of pancreatic cancer.
Collapse
Affiliation(s)
- Daniel Delitto
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - Dongyu Zhang
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - Song Han
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - Brian S Black
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - Andrea E Knowlton
- Department of Periodontology and Oral Biology, University of Florida Health Science Center, Gainesville, Florida
| | - Adrian C Vlada
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - George A Sarosi
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida. North Florida/South Georgia Veterans Health System, University of Florida Health Science Center, Gainesville, Florida
| | - Kevin E Behrns
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - Ryan M Thomas
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida. North Florida/South Georgia Veterans Health System, University of Florida Health Science Center, Gainesville, Florida
| | - Xiaomin Lu
- Department of Biostatistics and Children's Oncology Group, University of Florida Health Science Center, Gainesville, Florida
| | - Chen Liu
- Department of Pathology, Immunology, Laboratory Medicine, Colleges of Medicine, Dentistry and Public Health and Health Professions, University of Florida Health Science Center, Gainesville, Florida
| | - Thomas J George
- Department of Internal Medicine, University of Florida Health Science Center, Gainesville, Florida
| | - Steven J Hughes
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida
| | - Shannon M Wallet
- Department of Periodontology and Oral Biology, University of Florida Health Science Center, Gainesville, Florida
| | - Jose G Trevino
- Department of Surgery, University of Florida Health Science Center, Gainesville, Florida.
| |
Collapse
|
21
|
Nanjappa V, Renuse S, Sathe GJ, Raja R, Syed N, Radhakrishnan A, Subbannayya T, Patil A, Marimuthu A, Sahasrabuddhe NA, Guerrero-Preston R, Somani BL, Nair B, Kundu GC, Prasad TK, Califano JA, Gowda H, Sidransky D, Pandey A, Chatterjee A. Chronic exposure to chewing tobacco selects for overexpression of stearoyl-CoA desaturase in normal oral keratinocytes. Cancer Biol Ther 2015; 16:1593-603. [PMID: 26391970 PMCID: PMC4846103 DOI: 10.1080/15384047.2015.1078022] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 06/24/2015] [Accepted: 07/26/2015] [Indexed: 01/10/2023] Open
Abstract
Chewing tobacco is a common practice in certain socio-economic sections of southern Asia, particularly in the Indian subcontinent and has been well associated with head and neck squamous cell carcinoma. The molecular mechanisms of chewing tobacco which leads to malignancy remains unclear. In large majority of studies, short-term exposure to tobacco has been evaluated. From a biological perspective, however, long-term (chronic) exposure to tobacco mimics the pathogenesis of oral cancer more closely. We developed a cell line model to investigate the chronic effects of chewing tobacco. Chronic exposure to tobacco resulted in higher cellular proliferation and invasive ability of the normal oral keratinocytes (OKF6/TERT1). We carried out quantitative proteomic analysis of OKF6/TERT1 cells chronically treated with chewing tobacco compared to the untreated cells. We identified a total of 3,636 proteins among which expression of 408 proteins were found to be significantly altered. Among the overexpressed proteins, stearoyl-CoA desaturase (SCD) was found to be 2.6-fold overexpressed in the tobacco treated cells. Silencing/inhibition of SCD using its specific siRNA or inhibitor led to a decrease in cellular proliferation, invasion and colony forming ability of not only the tobacco treated cells but also in a panel of head and neck cancer cell lines. These findings suggest that chronic exposure to chewing tobacco induced carcinogenesis in non-malignant oral epithelial cells and SCD plays an essential role in this process. The current study provides evidence that SCD can act as a potential therapeutic target in head and neck squamous cell carcinoma, especially in patients who are users of tobacco.
Collapse
Affiliation(s)
- Vishalakshi Nanjappa
- Institute of Bioinformatics; International Technology Park; Bangalore, India
- Amrita School of Biotechnology; Amrita University; Kollam, India
| | - Santosh Renuse
- Institute of Bioinformatics; International Technology Park; Bangalore, India
- Amrita School of Biotechnology; Amrita University; Kollam, India
| | - Gajanan J Sathe
- Institute of Bioinformatics; International Technology Park; Bangalore, India
- Manipal University; Madhav Nagar; Manipal, India
| | - Remya Raja
- Institute of Bioinformatics; International Technology Park; Bangalore, India
| | - Nazia Syed
- Institute of Bioinformatics; International Technology Park; Bangalore, India
- Department of Biochemistry and Molecular Biology; Pondicherry University; Puducherry, India
| | - Aneesha Radhakrishnan
- Institute of Bioinformatics; International Technology Park; Bangalore, India
- Department of Biochemistry and Molecular Biology; Pondicherry University; Puducherry, India
| | - Tejaswini Subbannayya
- Institute of Bioinformatics; International Technology Park; Bangalore, India
- Amrita School of Biotechnology; Amrita University; Kollam, India
| | - Arun Patil
- Institute of Bioinformatics; International Technology Park; Bangalore, India
- School of Biotechnology; KIIT University; Bhubaneswar, India
| | | | | | - Rafael Guerrero-Preston
- Department of Otolaryngology-Head and Neck Surgery; Johns Hopkins University School of Medicine; Baltimore, MD USA
| | - Babu L Somani
- Institute of Bioinformatics; International Technology Park; Bangalore, India
| | - Bipin Nair
- Amrita School of Biotechnology; Amrita University; Kollam, India
| | - Gopal C Kundu
- National Center for Cell Science (NCCS); NCCS Complex; Pune, India
| | - T Keshava Prasad
- Institute of Bioinformatics; International Technology Park; Bangalore, India
- Amrita School of Biotechnology; Amrita University; Kollam, India
- YU-IOB Center for Systems Biology and Molecular Medicine; Yenepoya University; Mangalore, India
| | - Joseph A Califano
- Department of Otolaryngology-Head and Neck Surgery; Johns Hopkins University School of Medicine; Baltimore, MD USA
- Milton J. Dance Head and Neck Center; Greater Baltimore Medical Center; Baltimore, MD USA
| | - Harsha Gowda
- Institute of Bioinformatics; International Technology Park; Bangalore, India
- YU-IOB Center for Systems Biology and Molecular Medicine; Yenepoya University; Mangalore, India
| | - David Sidransky
- Department of Otolaryngology-Head and Neck Surgery; Johns Hopkins University School of Medicine; Baltimore, MD USA
| | - Akhilesh Pandey
- McKusick-Nathans Institute of Genetic Medicine; Johns Hopkins University School of Medicine; Baltimore, MD USA
- Department of Biological Chemistry; Johns Hopkins University School of Medicine; Baltimore, MD USA
- Department of Pathology; Johns Hopkins University School of Medicine; Baltimore, MD USA
| | - Aditi Chatterjee
- Institute of Bioinformatics; International Technology Park; Bangalore, India
- YU-IOB Center for Systems Biology and Molecular Medicine; Yenepoya University; Mangalore, India
| |
Collapse
|
22
|
Yuge K, Kikuchi E, Hagiwara M, Yasumizu Y, Tanaka N, Kosaka T, Miyajima A, Oya M. Nicotine Induces Tumor Growth and Chemoresistance through Activation of the PI3K/Akt/mTOR Pathway in Bladder Cancer. Mol Cancer Ther 2015; 14:2112-20. [PMID: 26184482 DOI: 10.1158/1535-7163.mct-15-0140] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 07/06/2015] [Indexed: 11/16/2022]
Abstract
Continued smoking is highly associated with not only a higher incidence but also greater risk of tumor recurrence, progression, and acquired chemoresistance of urothelial carcinoma. We investigated whether nicotine affects urothelial carcinoma, and the detailed mechanism by which nicotine could induce tumor growth and any associated chemoresistance. Cell viability was evaluated in the human bladder cancer cell line T24 exposed to nicotine with or without cisplatin (CDDP) and NVP-BEZ235 as a PI3K/mTOR dual inhibitor by the WST-1 assay. Protein expression of the PI3K/Akt/mTOR pathway was investigated by Western blotting or immunohistochemical analysis. The influence of nicotine on tumor growth was also evaluated with or without CDDP and/or NVP-BEZ235 in a subcutaneous bladder tumor model. The result demonstrated that cell proliferation was increased in T24 cells after exposure to nicotine. Phospho-specific Akt (pAkt) and phospho-specific p70 S6 kinase (pS6) were significantly upregulated by nicotine exposure. Tumor growth in vivo was significantly induced by nicotine exposure in accordance with increased pS6 expression. Nicotine attenuated inhibition of T24 cell growth by CDDP and further upregulated pS6 expression in vitro and in vivo. NVP-BZE235 inhibited T24 cell proliferation and pAkt and pS6 expression induced after exposure to nicotine and/or CDDP. In conclusion, nicotine increases tumor growth and induces acquired chemoresistance through activation of the PI3K/Akt/mTOR pathway in bladder cancer.
Collapse
Affiliation(s)
- Kazuyuki Yuge
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Eiji Kikuchi
- Department of Urology, Keio University School of Medicine, Tokyo, Japan.
| | - Masayuki Hagiwara
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Yota Yasumizu
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Nobuyuki Tanaka
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Takeo Kosaka
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Akira Miyajima
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
23
|
Chehab M, Caza T, Skotnicki K, Landas S, Bratslavsky G, Mollapour M, Bourboulia D. Targeting Hsp90 in urothelial carcinoma. Oncotarget 2015; 6:8454-73. [PMID: 25909217 PMCID: PMC4496161 DOI: 10.18632/oncotarget.3502] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 03/09/2015] [Indexed: 02/06/2023] Open
Abstract
Urothelial carcinoma, or transitional cell carcinoma, is the most common urologic malignancy that carries significant morbidity, mortality, recurrence risk and associated health care costs. Despite use of current chemotherapies and immunotherapies, long-term remission in patients with muscle-invasive or metastatic disease remains low, and disease recurrence is common. The molecular chaperone Heat Shock Protein-90 (Hsp90) may offer an ideal treatment target, as it is a critical signaling hub in urothelial carcinoma pathogenesis and potentiates chemoradiation. Preclinical testing with Hsp90 inhibitors has demonstrated reduced proliferation, enhanced apoptosis and synergism with chemotherapies and radiation. Despite promising preclinical data, clinical trials utilizing Hsp90 inhibitors for other malignancies had modest efficacy. Therefore, we propose that Hsp90 inhibition would best serve as an adjuvant treatment in advanced muscle-invasive or metastatic bladder cancers to potentiate other therapies. An overview of bladder cancer biology, current treatments, molecular targeted therapies, and the role for Hsp90 inhibitors in the treatment of urothelial carcinoma is the focus of this review.
Collapse
MESH Headings
- Angiogenesis Inhibitors/therapeutic use
- Antineoplastic Agents/therapeutic use
- Apoptosis
- BCG Vaccine/therapeutic use
- Carcinoma, Transitional Cell/epidemiology
- Carcinoma, Transitional Cell/metabolism
- Carcinoma, Transitional Cell/pathology
- Carcinoma, Transitional Cell/therapy
- Cell Cycle/drug effects
- Cell Division
- Cell Transformation, Neoplastic
- Chemoradiotherapy
- Chemotherapy, Adjuvant
- Clinical Trials as Topic
- Combined Modality Therapy
- Cystectomy
- Drug Resistance, Neoplasm
- Drugs, Investigational/therapeutic use
- HSP90 Heat-Shock Proteins/antagonists & inhibitors
- HSP90 Heat-Shock Proteins/chemistry
- HSP90 Heat-Shock Proteins/physiology
- Histone Code/drug effects
- Humans
- Models, Biological
- Molecular Targeted Therapy
- Muscle, Smooth/pathology
- Neoplasm Invasiveness
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/physiology
- Protein Kinase Inhibitors/therapeutic use
- Signal Transduction/drug effects
- Transcription, Genetic/drug effects
- Urologic Neoplasms/epidemiology
- Urologic Neoplasms/metabolism
- Urologic Neoplasms/pathology
- Urologic Neoplasms/therapy
Collapse
Affiliation(s)
- Mahmoud Chehab
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Tiffany Caza
- Department of Pathology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Kamil Skotnicki
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Steve Landas
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Pathology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Gennady Bratslavsky
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Upstate Cancer Research Institute, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Mehdi Mollapour
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Upstate Cancer Research Institute, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Dimitra Bourboulia
- Department of Urology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Upstate Cancer Research Institute, SUNY Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
24
|
Michailidi C, Hayashi M, Datta S, Sen T, Zenner K, Oladeru O, Brait M, Izumchenko E, Baras A, VandenBussche C, Argos M, Bivalacqua TJ, Ahsan H, Hahn NM, Netto GJ, Sidransky D, Hoque MO. Involvement of epigenetics and EMT-related miRNA in arsenic-induced neoplastic transformation and their potential clinical use. Cancer Prev Res (Phila) 2015; 8:208-21. [PMID: 25586904 PMCID: PMC4355280 DOI: 10.1158/1940-6207.capr-14-0251] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Exposure to toxicants leads to cumulative molecular changes that overtime increase a subject's risk of developing urothelial carcinoma. To assess the impact of arsenic exposure at a time progressive manner, we developed and characterized a cell culture model and tested a panel of miRNAs in urine samples from arsenic-exposed subjects, urothelial carcinoma patients, and controls. To prepare an in vitro model, we chronically exposed an immortalized normal human bladder cell line (HUC1) to arsenic. Growth of the HUC1 cells was increased in a time-dependent manner after arsenic treatment and cellular morphology was changed. In a soft agar assay, colonies were observed only in arsenic-treated cells, and the number of colonies gradually increased with longer periods of treatment. Similarly, invaded cells in an invasion assay were observed only in arsenic-treated cells. Withdrawal of arsenic treatment for 2.5 months did not reverse the tumorigenic properties of arsenic-treated cells. Western blot analysis demonstrated decreased PTEN and increased AKT and mTOR in arsenic-treated HUC1 cells. Levels of miR-200a, miR-200b, and miR-200c were downregulated in arsenic-exposed HUC1 cells by quantitative RT-PCR. Furthermore, in human urine, miR-200c and miR-205 were inversely associated with arsenic exposure (P = 0.005 and 0.009, respectively). Expression of miR-205 discriminated cancer cases from controls with high sensitivity and specificity (AUC = 0.845). Our study suggests that exposure to arsenic rapidly induces a multifaceted dedifferentiation program and miR-205 has potential to be used as a marker of arsenic exposure as well as a maker of early urothelial carcinoma detection.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Apoptosis
- Arsenic/adverse effects
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Blotting, Western
- Case-Control Studies
- Cell Movement
- Cell Proliferation
- Cell Transformation, Neoplastic/chemically induced
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/pathology
- Cells, Cultured
- Cohort Studies
- DNA Methylation
- Epigenesis, Genetic/genetics
- Epithelial-Mesenchymal Transition
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Immunoenzyme Techniques
- Male
- MicroRNAs/analysis
- MicroRNAs/genetics
- Middle Aged
- Neoplasm Invasiveness
- PTEN Phosphohydrolase/genetics
- PTEN Phosphohydrolase/metabolism
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- TOR Serine-Threonine Kinases/genetics
- TOR Serine-Threonine Kinases/metabolism
- Urinary Bladder/drug effects
- Urinary Bladder/metabolism
- Urinary Bladder/pathology
- Urinary Bladder Neoplasms/drug therapy
- Urinary Bladder Neoplasms/genetics
- Urinary Bladder Neoplasms/pathology
- Young Adult
Collapse
Affiliation(s)
- Christina Michailidi
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Masamichi Hayashi
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Sayantan Datta
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Tanusree Sen
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Kaitlyn Zenner
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Oluwadamilola Oladeru
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Mariana Brait
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Evgeny Izumchenko
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Alexander Baras
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | | | - Maria Argos
- Department of Health Studies, The University of Chicago, Chicago, Illinois
| | | | - Habibul Ahsan
- Department of Health Studies, The University of Chicago, Chicago, Illinois. Departments of Medicine and Human Genetics and Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois
| | - Noah M Hahn
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland
| | - George J Netto
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland. Department of Urology, Johns Hopkins University, Baltimore, Maryland
| | - David Sidransky
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Mohammad Obaidul Hoque
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, Maryland. Department of Urology, Johns Hopkins University, Baltimore, Maryland. Department of Oncology, Johns Hopkins University, Baltimore, Maryland.
| |
Collapse
|
25
|
Ichimura M, Minami A, Nakano N, Kitagishi Y, Murai T, Matsuda S. Cigarette smoke may be an exacerbation factor in nonalcoholic fatty liver disease via modulation of the PI3K/AKT pathway. AIMS MOLECULAR SCIENCE 2015. [DOI: 10.3934/molsci.2015.4.427] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
26
|
Millis SZ, Bryant D, Basu G, Bender R, Vranic S, Gatalica Z, Vogelzang NJ. Molecular profiling of infiltrating urothelial carcinoma of bladder and nonbladder origin. Clin Genitourin Cancer 2014; 13:e37-49. [PMID: 25178641 DOI: 10.1016/j.clgc.2014.07.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Accepted: 07/29/2014] [Indexed: 12/23/2022]
Abstract
BACKGROUND Infiltrating UC represents the second most common genitourinary malignancy. Advanced UC has a poor prognosis and new treatments are needed. Molecular profiling of UC might identify biomarkers associated with targeted therapies or chemotherapeutics, providing physicians with new treatment options. MATERIALS AND METHODS Five hundred thirty-seven cases of locally advanced or metastatic UC of the bladder, 74 nonbladder, and 55 nonurothelial bladder cancers were profiled using mutation analysis, in situ hybridization, and immunohistochemistry assays for biomarkers predictive of therapy response. RESULTS Molecular profiling of UC showed high overexpression of topoisomerase 2α, common phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha and/or phosophatase and tensin homolog (PTEN) alterations in nonbladder (27%) and bladder UC (21%), and rare gene mutations across subtypes. Compared with nonbladder, bladder UC consistently exhibited more frequent abnormal protein expression, including HER2 (10% vs. 3%; P = .04), tyrosine protein c-Kit receptor kinases (11% vs. 5%), c-Met proto-oncogene, receptor tyrosine kinases (25% vs. 8%), androgen receptor (16% vs. 6%), O(6)-methylguanine-methyltransferase (63% vs. 43%), ribonucleotide reductase M1 (32% vs. 11%), Serum protein acidic and rich in cysteine (SPARC) (69% vs. 33%), and topoisomerase 1 (63% vs. 39%). Bladder UC also exhibited increased amplification of HER2 (12% vs. 2%; P = .06). CONCLUSION Comprehensive molecular profiling of UC identified a large number of biomarkers aberrations that might direct treatment in conventional chemotherapies and targeted therapies, not currently recommended in this population. As a group, bladder UC exhibited higher levels of actionable biomarkers, suggesting that UC from different primary sites and non-UC are driven by different molecular pathways. These differences could have clinical implications resulting in different treatment regimens depending on the site of origin of UC.
Collapse
Affiliation(s)
| | | | | | | | - Semir Vranic
- Department of Pathology, Clinical Center, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | | | | |
Collapse
|
27
|
Kagohara LT, Schussel JL, Subbannayya T, Sahasrabuddhe N, Lebron C, Brait M, Maldonado L, Valle BL, Pirini F, Jahuira M, Lopez J, Letelier P, Brebi-Mieville P, Ili C, Pandey A, Chatterjee A, Sidransky D, Guerrero-Preston R. Global and gene-specific DNA methylation pattern discriminates cholecystitis from gallbladder cancer patients in Chile. Future Oncol 2014; 11:233-49. [PMID: 25066711 PMCID: PMC4332836 DOI: 10.2217/fon.14.165] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AIM The aim of the study was to evaluate the use of global and gene-specific DNA methylation changes as potential biomarkers for gallbladder cancer (GBC) in a cohort from Chile. MATERIAL & METHODS DNA methylation was analyzed through an ELISA-based technique and quantitative methylation-specific PCR. RESULTS Global DNA Methylation Index (p = 0.02) and promoter methylation of SSBP2 (p = 0.01) and ESR1 (p = 0.05) were significantly different in GBC when compared with cholecystitis. Receiver curve operator analysis revealed promoter methylation of APC, CDKN2A, ESR1, PGP9.5 and SSBP2, together with the Global DNA Methylation Index, had 71% sensitivity, 95% specificity, a 0.97 area under the curve and a positive predictive value of 90%. CONCLUSION Global and gene-specific DNA methylation may be useful biomarkers for GBC clinical assessment.
Collapse
Affiliation(s)
- Luciane Tsukamoto Kagohara
- Department of Otolaryngology–Head & Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Juliana L Schussel
- Department of Otolaryngology–Head & Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Oral Medicine, Post Graduation Program in Dentistry, Universidade Federal do Paraná, Curitiba, Brazil
| | | | | | - Cynthia Lebron
- Department of Otolaryngology–Head & Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Mariana Brait
- Department of Otolaryngology–Head & Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Leonel Maldonado
- Department of Otolaryngology–Head & Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Blanca L Valle
- Department of Otolaryngology–Head & Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Francesca Pirini
- Department of Otolaryngology–Head & Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Martha Jahuira
- Department of Otolaryngology–Head & Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Jaime Lopez
- Universidad de La Frontera, Facultad de Medicina, Departamento de Anatomía Patológica, Laboratorio Patología Molecular, BIOREN-CEGIN, Temuco, Chile
| | - Pablo Letelier
- Universidad de La Frontera, Facultad de Medicina, Departamento de Anatomía Patológica, Laboratorio Patología Molecular, BIOREN-CEGIN, Temuco, Chile
- Escuela Ciencias de la Salud, Universidad Católica de Temuco, Temuco, Chile
| | - Priscilla Brebi-Mieville
- Department of Otolaryngology–Head & Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Universidad de La Frontera, Facultad de Medicina, Departamento de Anatomía Patológica, Laboratorio Patología Molecular, BIOREN-CEGIN, Temuco, Chile
| | - Carmen Ili
- Department of Otolaryngology–Head & Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Universidad de La Frontera, Facultad de Medicina, Departamento de Anatomía Patológica, Laboratorio Patología Molecular, BIOREN-CEGIN, Temuco, Chile
| | - Akhilesh Pandey
- McKusick–Nathans Institute of Genetic Medicine, Baltimore, MD 21205, USA
- Department of Biological Chemistry, Baltimore, MD 21205, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Aditi Chatterjee
- Department of Otolaryngology–Head & Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Institute of Bioinformatics, Whitefield, Bangalore 560066, Karnataka, India
| | - David Sidransky
- Department of Otolaryngology–Head & Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Rafael Guerrero-Preston
- Department of Otolaryngology–Head & Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- University of Puerto Rico School of Medicine, Department of Obstetrics & Gynecology, San Juan, Puerto Rico
| |
Collapse
|
28
|
Pinto-Leite R, Carreira I, Melo J, Ferreira SI, Ribeiro I, Ferreira J, Filipe M, Bernardo C, Arantes-Rodrigues R, Oliveira P, Santos L. Genomic characterization of three urinary bladder cancer cell lines: understanding genomic types of urinary bladder cancer. Tumour Biol 2014; 35:4599-617. [PMID: 24459064 DOI: 10.1007/s13277-013-1604-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 12/30/2013] [Indexed: 11/25/2022] Open
Abstract
Several genomic regions are frequently altered and associated with the type, stage and progression of urinary bladder cancer (UBC). We present the characterization of 5637, T24 and HT1376 UBC cell lines by karyotyping, fluorescence in situ hybridization (FISH), array comparative genomic hybridization (aCGH) and multiplex ligation-dependent probe amplification (MLPA) analysis. Some cytogenetic anomalies present in UBC were found in the three cell lines, such as chromosome 20 aneuploidy and the loss of 9p21. Some gene loci losses (e.g. CDKN2A) and gains (e.g. HRAS, BCL2L1 and PTPN1) were coincident across all cell lines. Although some significant heterogeneity and complexity were detected between them, their genomic profiles exhibited a similar pattern to UBC. We suggest that 5637 and HT1376 represent the E2F3/RB1 pathway due to amplification of 6p22.3, concomitant with loss of one copy of RB1 and mutation of the remaining copy. The HT1376 presented a 10q deletion involving PTEN region and no alteration of PIK3CA region which, in combination with the inactivation of TP53, bears more invasive and metastatic properties than 5637. The T24 belongs to the alternative pathway of FGFR3/CCND1 by presenting mutated HRAS and over-represented CCND1. These cell lines cover the more frequent subtypes of UBC and are reliable models that can be used, as a group, in preclinical studies.
Collapse
Affiliation(s)
- Rosário Pinto-Leite
- Cytogenetic Laboratory, Department of Human Genetics, Hospital Center of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Peluso MEM, Munnia A, Bollati V, Srivatanakul P, Jedpiyawongse A, Sangrajrang S, Ceppi M, Giese RW, Boffetta P, Baccarelli AA. Aberrant methylation of hypermethylated-in-cancer-1 and exocyclic DNA adducts in tobacco smokers. Toxicol Sci 2013; 137:47-54. [PMID: 24154486 DOI: 10.1093/toxsci/kft241] [Citation(s) in RCA: 206] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Tobacco smoke has been shown to produce both DNA damage and epigenetic alterations. However, the potential role of DNA damage in generating epigenetic changes is largely underinvestigated in human studies. We examined the effects of smoking on the levels of DNA methylation in genes for tumor protein p53, cyclin-dependent kinase inhibitor2A, hypermethylated-in-cancer-1 (HIC1), interleukin-6, Long Interspersed Nuclear Element type1, and Alu retrotransposons in blood of 177 residents in Thailand using bisulfite-PCR andpyrosequencing. Then, we analyzed the relationship of this methylation with the oxidative DNA adduct, M₁dG (a malondialdehyde adduct), measured by ³²P-postlabeling. Multivariate statistical analyses showed that HIC1 methylation levels were significantly increased in smokers compared with nonsmokers (p ≤ .05). A dose response was observed, with the highest HIC1 methylation levels in smokers of ≥ 10 cigarettes/day relative to nonsmokers and intermediate values in smokers of 1-9 cigarettes/day (p for trend ≤ .001). No additional relationships were observed. We also evaluated correlations between M₁dG and the methylation changes at each HIC1 CpG site individually. The levels of this adduct in smokers showed a significant linear correlation with methylation at one of the 3 CpGs evaluated in HIC1: hypermethylation at position 1904864340 was significantly correlated with the adduct M₁dG (covariate-adjusted regression coefficient (β) = .224 ± .101 [SE], p ≤ .05). No other correlations were detected. Our study extends prior work by others associating hypermethylation of HIC1 with smoking; shows that a very specific hypermethylation event can arise from smoking; and encourages future studies that explore a possible role for M₁dG in connecting smoking to this latter hypermethylation.
Collapse
Affiliation(s)
- Marco E M Peluso
- * Cancer Risk Factor Branch, Cancer Prevention and Research Institute, Florence, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Kreimer U, Schulz WA, Koch A, Niegisch G, Goering W. HERV-K and LINE-1 DNA Methylation and Reexpression in Urothelial Carcinoma. Front Oncol 2013; 3:255. [PMID: 24133654 PMCID: PMC3783855 DOI: 10.3389/fonc.2013.00255] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 09/11/2013] [Indexed: 11/16/2022] Open
Abstract
Changes in DNA methylation frequently accompany cancer development. One prominent change is an apparently genome-wide decrease in methylcytosine that is often ascribed to DNA hypomethylation at retroelements comprising nearly half the genome. DNA hypomethylation may allow reactivation of retroelements, enabling retrotransposition, and causing gene expression disturbances favoring tumor development. However, neither the extent of hypomethylation nor of retroelement reactivation are precisely known. We therefore assessed DNA methylation and expression of three major classes of retroelements (LINE-1, HERV-K, and AluY) in human urinary bladder cancer tissues and cell lines by pyrosequencing and quantitative reverse transcription–polymerase chain reaction, respectively. We found substantial global LINE-1 DNA hypomethylation in bladder cancer going along with a shift toward full-length LINE-1 expression. Thus, pronounced differences in LINE-1 expression were observed, which may be promoted, among others, by LINE-1 hypomethylation. Significant DNA hypomethylation was found at the HERV-K_22q11.23 proviral long terminal repeat (LTR) in bladder cancer tissues but without reactivation of its expression. DNA methylation of HERVK17, essentially absent from normal urothelial cells, was elevated in cell lines from invasive bladder cancers. Accordingly, the faint expression of HERVK17 in normal urothelial cells disappeared in such cancer cell lines. Of 16 additional HERV-Ks, expression of 7 could be detected in the bladder, albeit generally at low levels. Unlike in prostate cancers, none of these showed significant expression changes in bladder cancer. In contrast, expression of the AluYb8 but not of the AluYa5 family was significantly increased in bladder cancer tissues. Collectively, our findings demonstrate a remarkable specificity of changes in expression and DNA methylation of retroelements in bladder cancer with a significantly different pattern from that in prostate cancer.
Collapse
Affiliation(s)
- Ulrike Kreimer
- Department of Urology, Medical Faculty, Heinrich Heine University , Düsseldorf , Germany
| | | | | | | | | |
Collapse
|
31
|
DNA methylation in complex disease: Applications in nursing research, practice, and policy. Nurs Outlook 2013; 61:235-241.e4. [DOI: 10.1016/j.outlook.2013.04.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 04/23/2013] [Accepted: 04/28/2013] [Indexed: 12/31/2022]
|
32
|
Martignoni G. Discovering smoking-related pathway alterations in urothelial cell carcinoma pathogenesis. Cell Cycle 2013; 12:1483. [PMID: 23652923 PMCID: PMC3680525 DOI: 10.4161/cc.24852] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Guido Martignoni
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy.
| |
Collapse
|
33
|
|