1
|
Li K, Qi L, Tang G, Xu H, Li Z, Fan B, Li Z, Li Y. Epigenetic Regulation in Urothelial Carcinoma. Curr Mol Med 2024; 24:85-97. [PMID: 36545729 DOI: 10.2174/1566524023666221221094432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 11/08/2022] [Accepted: 11/10/2022] [Indexed: 12/24/2022]
Abstract
Urothelial carcinoma (UC) is a common malignancy that remains a clinical challenge: Non-muscle-invasive urothelial carcinoma (NMIUC) has a high rate of recurrence and risk of progression, while muscle-invasive urothelial carcinoma (MIUC) has a high mortality. Although some new treatments, such as immunotherapies, have shown potential effects on some patients, most cases of advanced UC remain incurable. While treatments based on epigenetic mechanisms, whether combined with traditional platinum-based chemotherapy or emerging immunotherapy, show therapeutic advantages. With the advancement of sequencing and bioinformatics, the study of epigenomics, containing DNA methylation, histone modifications, chromatin remodeling, and non-coding RNA, is increasingly linked with the occurrence and progression of UC. Since the epigenetics of UC is a constantly developing field of medicine, this review aims to summarize the latest research on epigenetic regulation of UC, generalize the mechanism of epigenetics in UC, and reveal the potential epigenetic therapies in the clinical setting, in order to provide some new clues on the discovery of new drugs based on the epigenetics.
Collapse
Affiliation(s)
- Ke Li
- Department of Urology, Xiangya Hospital of Central South University, Changsha, China
| | - Lin Qi
- Department of Urology, Xiangya Hospital of Central South University, Changsha, China
| | - Guyu Tang
- Department of Urology, Xiangya Hospital of Central South University, Changsha, China
| | - Haozhe Xu
- Department of Urology, Xiangya Hospital of Central South University, Changsha, China
| | - Zhi Li
- Department of Urology, Xiangya Hospital of Central South University, Changsha, China
| | - Bo Fan
- Department of Urology, Xiangya Hospital of Central South University, Changsha, China
| | - Zhongbei Li
- College of Chemistry and Chemical Engineering, Central South University, Changsha, China
| | - Yuan Li
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
2
|
Petrelli A, Bellomo SE, Sarotto I, Kubatzki F, Sgandurra P, Maggiorotto F, Di Virgilio MR, Ponzone R, Geuna E, Galizia D, Nuzzo AM, Medico E, Miglio U, Berrino E, Venesio T, Ribisi S, Provero P, Sapino A, Giordano S, Montemurro F. MiR-100 is a predictor of endocrine responsiveness and prognosis in patients with operable luminal breast cancer. ESMO Open 2021; 5:e000937. [PMID: 33122354 PMCID: PMC7597498 DOI: 10.1136/esmoopen-2020-000937] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/14/2020] [Accepted: 09/16/2020] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Overexpression of miR-100 in stem cells derived from basal-like breast cancers causes loss of stemness, induction of luminal breast cancer markers and response to endocrine therapy. We, therefore, explored miR-100 as a novel biomarker in patients with luminal breast cancer. METHODS miR-100 expression was studied in 90 patients with oestrogen-receptor-positive/human-epidermal growth factor receptor 2-negative breast cancer enrolled in a prospective study of endocrine therapy given either preoperatively, or for the treatment of de novo metastatic disease. Response was defined as a Ki67 ≤2.7% after 21±3 days of treatment. The prognostic role of miR-100 expression was evaluated in the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) and The Cancer Genome Atlas (TCGA) breast cancer datasets. Additionally, we explored the correlation between miR-100 and the expression its targets reported as being associated with endocrine resistance. Finally, we evaluated whether a signature based on miR-100 and its target genes could predict the luminal A molecular subtype. RESULTS Baseline miR-100 was significantly anticorrelated with baseline and post-treatment Ki67 (p<0.001 and 0.004, respectively), and independently associated with response to treatment (OR 3.329, p=0.047). In the METABRIC dataset, high expression of miR-100 identified women with luminal A tumours treated with adjuvant endocrine therapy with improved overall survival (HR 0.55, p<0.001). miR-100 was negatively correlated with PLK1, FOXA1, mTOR and IGF1R expression, potentially explaining its prognostic effect. Finally, a miR-100-based signature developed in patients enrolled in the prospective study outperformed Ki67 alone in predicting the luminal A phenotype. CONCLUSIONS Our findings suggest that miR-100 should be further explored as a biomarker in patients with luminal breast cancer.
Collapse
Affiliation(s)
- Annalisa Petrelli
- Cancer Molecular Biology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | | | - Ivana Sarotto
- Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Franziska Kubatzki
- Gynaecological Oncology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Paola Sgandurra
- Gynaecological Oncology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Furio Maggiorotto
- Gynaecological Oncology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | | | - Riccardo Ponzone
- Gynaecological Oncology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Elena Geuna
- Multidisciplinary Outpatient Oncology Clinic, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Danilo Galizia
- Multidisciplinary Outpatient Oncology Clinic, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Anna Maria Nuzzo
- Clinical Research Office, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Enzo Medico
- Department of Oncology, University of Turin, Torino, Italy; Oncogenomics Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Umberto Miglio
- Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Enrico Berrino
- Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy; Department of Medical Sciences, University of Turin, Torino, Italy
| | - Tiziana Venesio
- Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Salvatore Ribisi
- Cancer Molecular Biology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Paolo Provero
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy; Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Anna Sapino
- Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy; Department of Medical Sciences, University of Turin, Torino, Italy
| | - Silvia Giordano
- Cancer Molecular Biology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy; Department of Oncology, University of Turin, Torino, Italy
| | - Filippo Montemurro
- Multidisciplinary Outpatient Oncology Clinic, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.
| |
Collapse
|
3
|
Crabb SJ, Danson S, Catto JWF, Hussain S, Chan D, Dunkley D, Downs N, Marwood E, Day L, Saunders G, Light M, Whitehead A, Ellis D, Sarwar N, Enting D, Birtle A, Johnson B, Huddart R, Griffiths G. Phase I Trial of DNA Methyltransferase Inhibitor Guadecitabine Combined with Cisplatin and Gemcitabine for Solid Malignancies Including Urothelial Carcinoma (SPIRE). Clin Cancer Res 2021; 27:1882-1892. [PMID: 33472913 PMCID: PMC7611191 DOI: 10.1158/1078-0432.ccr-20-3946] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/22/2020] [Accepted: 01/15/2021] [Indexed: 01/02/2023]
Abstract
PURPOSE Preclinical data indicate that DNA methyltransferase inhibition will circumvent cisplatin resistance in various cancers. PATIENT AND METHODS SPIRE comprised a dose-escalation phase for incurable metastatic solid cancers, followed by a randomized dose expansion phase for neoadjuvant treatment of T2-4a N0 M0 bladder urothelial carcinoma. The primary objective was a recommended phase II dose (RP2D) for guadecitabine combined with gemcitabine and cisplatin. Treatment comprised 21-day gemcitabine and cisplatin cycles (cisplatin 70 mg/m2, i.v., day 8 and gemcitabine 1,000 mg/m2, i.v., days 8 + 15). Guadecitabine was injected subcutaneously on days 1-5, within escalation phase cohorts, and to half of 20 patients in the expansion phase. Registration ID: ISRCTN 16332228. RESULTS Within the escalation phase, dose-limiting toxicities related predominantly to myelosuppression requiring G-CSF prophylaxis from cohort 2 (guadecitabine 20 mg/m2, days 1-5). The most common grade ≥3 adverse events in 17 patients in the dose-escalation phase were neutropenia (76.5%), thrombocytopenia (64.7%), leukopenia (29.4%), and anemia (29.4%). Addition of guadecitabine to gemcitabine and cisplatin in the expansion phase resulted in similar rates of severe hematologic adverse events, similar cisplatin dose intensity, but modestly reduced gemcitabine dose intensity. Radical treatment options after chemotherapy were not compromised. Pharmacodynamics evaluations indicated guadecitabine maximal target effect at the point of cisplatin administration. Pharmacokinetics were consistent with prior data. No treatment-related deaths occurred. CONCLUSIONS The guadecitabine RP2D was 20 mg/m2, days 1-5, in combination with gemcitabine and cisplatin and required GCSF prophylaxis. Gene promoter methylation pharmacodynamics are optimal with this schedule. Addition of guadecitabine to gemcitabine and cisplatin was tolerable, despite some additional myelosuppression, and warrants further investigation to assess efficacy.
Collapse
Affiliation(s)
- Simon J Crabb
- Southampton Clinical Trials Unit, University of Southampton, Southampton, England, United Kingdom.
- University Hospital Southampton NHS Foundation Trust, Southampton, England, United Kingdom
- Southampton Experimental Cancer Medicine Centre, University of Southampton, Southampton, England, United Kingdom
| | - Sarah Danson
- Sheffield Experimental Cancer Medicine Centre, Weston Park Hospital, University of Sheffield, Sheffield, England, United Kingdom
| | - James W F Catto
- Academic Urology Unit, University of Sheffield, Sheffield, England, United Kingdom
| | - Syed Hussain
- Sheffield Experimental Cancer Medicine Centre, Weston Park Hospital, University of Sheffield, Sheffield, England, United Kingdom
| | - Danna Chan
- Astex Pharmaceuticals, Inc., Pleasanton, California
| | - Denise Dunkley
- Southampton Clinical Trials Unit, University of Southampton, Southampton, England, United Kingdom
- University Hospital Southampton NHS Foundation Trust, Southampton, England, United Kingdom
- Southampton Experimental Cancer Medicine Centre, University of Southampton, Southampton, England, United Kingdom
| | - Nichola Downs
- Southampton Clinical Trials Unit, University of Southampton, Southampton, England, United Kingdom
- University Hospital Southampton NHS Foundation Trust, Southampton, England, United Kingdom
| | - Ellice Marwood
- Southampton Clinical Trials Unit, University of Southampton, Southampton, England, United Kingdom
- University Hospital Southampton NHS Foundation Trust, Southampton, England, United Kingdom
| | - Laura Day
- Southampton Clinical Trials Unit, University of Southampton, Southampton, England, United Kingdom
- University Hospital Southampton NHS Foundation Trust, Southampton, England, United Kingdom
| | - Geoff Saunders
- Southampton Clinical Trials Unit, University of Southampton, Southampton, England, United Kingdom
- University Hospital Southampton NHS Foundation Trust, Southampton, England, United Kingdom
| | - Michelle Light
- Southampton Clinical Trials Unit, University of Southampton, Southampton, England, United Kingdom
- University Hospital Southampton NHS Foundation Trust, Southampton, England, United Kingdom
| | - Amy Whitehead
- Southampton Clinical Trials Unit, University of Southampton, Southampton, England, United Kingdom
- University Hospital Southampton NHS Foundation Trust, Southampton, England, United Kingdom
| | - Deborah Ellis
- Southampton Clinical Trials Unit, University of Southampton, Southampton, England, United Kingdom
- University Hospital Southampton NHS Foundation Trust, Southampton, England, United Kingdom
| | - Naveed Sarwar
- Department of Oncology, Charing Cross Hospital, London, England, United Kingdom
| | - Deborah Enting
- Department of Oncology, Guy's and St Thomas' NHS Foundation Trust, London, England, United Kingdom
| | - Alison Birtle
- Lancashire Teaching Hospitals NHS Foundation Trust, Preston, England, United Kingdom
| | | | - Robert Huddart
- The Institute of Cancer Research, Sutton, England, United Kingdom
| | - Gareth Griffiths
- Southampton Clinical Trials Unit, University of Southampton, Southampton, England, United Kingdom
- University Hospital Southampton NHS Foundation Trust, Southampton, England, United Kingdom
| |
Collapse
|
4
|
Kardoust Parizi M, Margulis V, Lotan Y, Mori K, Shariat SF. Fibroblast growth factor receptor: A systematic review and meta-analysis of prognostic value and therapeutic options in patients with urothelial bladder carcinoma. Urol Oncol 2021; 39:409-421. [PMID: 33642228 DOI: 10.1016/j.urolonc.2021.01.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/16/2020] [Accepted: 01/21/2021] [Indexed: 01/21/2023]
Abstract
To evaluate the oncologic prognostic value of fibroblast growth factor receptor (FGFR) and to assess the safety and efficacy of its inhibitors in patients with urothelial bladder carcinoma. A literature search using PubMed, Scopus, and Cochrane Library was conducted on June 2020 to identify relevant studies according to the Preferred Reporting Items for Systematic Review and Meta-Analysis guidelines. The pooled recurrence-free survival (RFS), progression-free survival (PFS), and cancer-specific survival (CSS) were calculated using a fixed or random effects model in patients with nonmuscle invasive bladder cancer (NMIBC). Overall, 62 studies comprising 9,229 patients were eligible and included in this systematic review and meta-analysis. Both FGFR3 mutation and protein overexpression were significantly associated with RFS, PFS, CSS, and overall survival. FGFR3 mutation was associated with worse RFS and better PFS (pooled hazard ratio: 1.30; 95% confidence interval: 1.08-1.57, and pooled hazard ratio: 0.62; 95% confidence interval: 0.42-0.92, respectively) in patients with NMIBC. In 11 studies reporting on the response to FGFR inhibitors, complete response rates, disease control rates, and overall response rate of 0% to 8%, 59.3% to 64.2%, and 40% were reported for dovitinib, infigratinib, and erdafitinib, respectively. Based on this study, FGFR3 mutation is a statistically significant prognostic factor for RFS in NMIBC. FGFR inhibitors have measurable benefit in patients with advanced and metastatic urothelial carcinoma. However, the results of ongoing RCTs and future well-designed studies are awaited to capture the differential biologic and clinical behavior of tumors harboring FGFR while helping to identify those who are most likely to benefit from FGFR inhibitors.
Collapse
Affiliation(s)
- Mehdi Kardoust Parizi
- Department of Urology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran; Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Vitaly Margulis
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Yair Lotan
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Keiichiro Mori
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Shahrokh F Shariat
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX; Department of Urology, Weill Cornell Medical College, New York, NY; Department of Urology, Second Faculty of Medicine, Charles University, Prag, Czech Republic; Institute for Urology and Reproductive Health, I.M. Sechenov First Moscow State Medical University, Moscow, Russia; Division of Urology, Department of Special Surgery, Jordan University Hospital, The University of Jordan, Amman, Jordan; European Association of Urology research foundation, Arnhem, The Netherlands.
| |
Collapse
|
5
|
Abstract
The identification of mutations in FGFR3 in bladder tumors in 1999 led to major interest in this receptor and during the subsequent 20 years much has been learnt about the mutational profiles found in bladder cancer, the phenotypes associated with these and the potential of this mutated protein as a target for therapy. Based on mutational and expression data, it is estimated that >80% of non-muscle-invasive bladder cancers (NMIBC) and ∼40% of muscle-invasive bladder cancers (MIBC) have upregulated FGFR3 signalling, and these frequencies are likely to be even higher if alternative splicing of the receptor, expression of ligands and changes in regulatory mechanisms are taken into account. Major efforts by the pharmaceutical industry have led to development of a range of agents targeting FGFR3 and other FGF receptors. Several of these have entered clinical trials, and some have presented very encouraging early results in advanced bladder cancer. Recent reviews have summarised the drugs and related clinical trials in this area. This review will summarise what is known about the effects of FGFR3 and its mutant forms in normal urothelium and bladder tumors, will suggest when and how this protein contributes to urothelial cancer pathogenesis and will highlight areas that may benefit from further study.
Collapse
Affiliation(s)
- Margaret A. Knowles
- Division of Molecular Medicine, Leeds Institute of Medical Research at St James’s, St James’s University Hospital, Leeds LS9 7TF, UK
| |
Collapse
|
6
|
Oleanolic acid inhibits cell proliferation migration and invasion and induces SW579 thyroid cancer cell line apoptosis by targeting forkhead transcription factor A. Anticancer Drugs 2020; 30:812-820. [PMID: 30882397 DOI: 10.1097/cad.0000000000000777] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Oleanolic acid (OA) is a naturally occurring triterpenoid that possesses antitumor activity against several tumor cell lines. However, the potential mechanism underlying OA-induced thyroid carcinoma cell death is poorly understood. We investigated the biological functions of OA by performing migration, invasion, colony formation, and apoptosis assays on SW579 cells. Forkhead box A1 (FOXA1) expression was used to predict poor prognosis in patients with thyroid carcinoma among the TCGA samples from the UALCAN and gene expression profiling interactive analysis databases. Western blot was used to detect protein expression level. Results revealed that OA inhibited the migration, colony formation, and invasion of thyroid carcinoma cells in a dose-dependent manner. Further investigation verified that OA treatment induced significant apoptosis of thyroid carcinoma cells. Moreover, high FOXA1 expression predicted the poor prognosis of patients with thyroid cancer. The proliferation, migration, and invasion of thyroid carcinoma cells were significantly decreased when FOXA1 was silenced. OA significantly increased Akt phosphorylation and reduced FOXA1 expression in SW579 cells, but only PI3K/Akt inhibitor LY294002 attenuated OA-induced FOXA1 downregulation. Furthermore, Akt overexpression suppressed the FOXA1 expression in SW579 cells. In addition, molecular docking assay revealed that OA possessed high affinity toward FOXA1 with a low binding energy. OA may be a potential chemotherapeutic agent against thyroid carcinoma cells.
Collapse
|
7
|
Mehus AA, Bergum N, Knutson P, Shrestha S, Zhou XD, Garrett SH, Sens DA, Sens MA, Somji S. Activation of PPARγ and inhibition of cell proliferation reduces key proteins associated with the basal subtype of bladder cancer in As3+-transformed UROtsa cells. PLoS One 2020; 15:e0237976. [PMID: 32822399 PMCID: PMC7444546 DOI: 10.1371/journal.pone.0237976] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 07/30/2020] [Indexed: 11/25/2022] Open
Abstract
Environmental exposure to arsenite (As3+) has a strong association with the development of human urothelial cancer (UC) and is the 5th most common cancer in men and the 12th most common cancer in women. Muscle invasive urothelial cancer (MIUC) are grouped into basal or luminal molecular subtypes based on their gene expression profile. The basal subtype is more aggressive and can be associated with squamous differentiation, characterized by high expression of keratins (KRT1, 5, 6, 14, and 16) and epidermal growth factor receptor (EGFR) within the tumors. The luminal subtype is less aggressive and is predominately characterized by elevated gene expression of peroxisome proliferator-activated receptor- gamma (PPARγ) and forkhead box protein A1 (FOXA1). We have previously shown that As3+-transformed urothelial cells (As-T) exhibit a basal subtype of UC expressing genes associated with squamous differentiation. We hypothesized that the molecular subtype of the As-T cells could be altered by inducing the expression of PPARγ and/or inhibiting the proliferation of the cells. Non-transformed and As-T cells were treated with Troglitazone (TG, PPARG agonist, 10 μM), PD153035 (PD, an EGFR inhibitor, 1 μM) or a combination of TG and PD for 3 days. The results obtained demonstrate that treatment of the As-T cells with TG upregulated the expression of PPARγ and FOXA1 whereas treatment with PD decreased the expression of some of the basal keratins. However, a combined treatment of TG and PD resulted in a consistent decrease of several proteins associated with the basal subtype of bladder cancers (KRT1, KRT14, KRT16, P63, and TFAP2A). Our data suggests that activation of PPARγ while inhibiting cell proliferation facilitates the regulation of genes involved in maintaining the luminal subtype of UC. In vivo animal studies are needed to address the efficacy of using PPARγ agonists and/or proliferation inhibitors to reduce tumor grade/stage of MIUC.
Collapse
Affiliation(s)
- Aaron A. Mehus
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Nicholas Bergum
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Peter Knutson
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Swojani Shrestha
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Xu Dong Zhou
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Scott H. Garrett
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Donald A. Sens
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Mary Ann Sens
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
| | - Seema Somji
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, North Dakota, United States of America
- * E-mail:
| |
Collapse
|
8
|
Osei-Amponsa V, Buckwalter JM, Shuman L, Zheng Z, Yamashita H, Walter V, Wildermuth T, Ellis-Mohl J, Liu C, Warrick JI, Shantz LM, Feehan RP, Al-Ahmadie H, Mendelsohn C, Raman JD, Kaestner KH, Wu XR, DeGraff DJ. Hypermethylation of FOXA1 and allelic loss of PTEN drive squamous differentiation and promote heterogeneity in bladder cancer. Oncogene 2019; 39:1302-1317. [PMID: 31636388 DOI: 10.1038/s41388-019-1063-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 10/02/2019] [Accepted: 10/07/2019] [Indexed: 01/04/2023]
Abstract
Intratumoral heterogeneity in bladder cancer is a barrier to accurate molecular sub-classification and treatment efficacy. However, individual cellular and mechanistic contributions to tumor heterogeneity are controversial. We examined potential mechanisms of FOXA1 and PTEN inactivation in bladder cancer and their contribution to tumor heterogeneity. These analyses were complemented with inactivation of FOXA1 and PTEN in intermediate and luminal mouse urothelium. We show inactivation and reduced expression of FOXA1 and PTEN is prevalent in human disease, where PTEN and FOXA1 are downregulated by allelic loss and site-specific DNA hypermethylation, respectively. Conditional inactivation of both Foxa1 and Pten in intermediate/luminal cells in mice results in development of bladder cancer exhibiting squamous features as well as enhanced sensitivity to a bladder-specific carcinogen. In addition, FOXA1 is hypermethylated in basal bladder cancer cell lines, and this is reversed by treatment with DNA methyltransferase inhibitors. By integrating human correlative and in vivo studies, we define a critical role for PTEN loss and epigenetic silencing of FOXA1 in heterogeneous human disease and show genetic targeting of luminal/intermediate cells in mice drives squamous differentiation.
Collapse
Affiliation(s)
- Vasty Osei-Amponsa
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Jenna M Buckwalter
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA.,Department of Surgery, Division of Urology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Lauren Shuman
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA.,Department of Surgery, Division of Urology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Zongyu Zheng
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Hironobu Yamashita
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA.,Department of Surgery, Division of Urology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Vonn Walter
- Department of Public Health Sciences, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Thomas Wildermuth
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA.,Department of Surgery, Division of Urology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Justine Ellis-Mohl
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA.,Department of Surgery, Division of Urology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Chang Liu
- Department of Urology, Columbia University, New York, NY, USA
| | - Joshua I Warrick
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA.,Department of Surgery, Division of Urology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Lisa M Shantz
- Department of Cellular and Molecular Physiology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Robert P Feehan
- Department of Cellular and Molecular Physiology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Hikmat Al-Ahmadie
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Jay D Raman
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA.,Department of Surgery, Division of Urology, The Pennsylvania State University, College of Medicine, Hershey, PA, USA
| | - Klaus H Kaestner
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Xue-Ru Wu
- Departments of Urology and Pathology, New York University School of Medicine, New York, NY, USA.,Veterans Affairs New York Harbor Healthcare System, Manhattan Campus, New York, NY, USA
| | - David J DeGraff
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, College of Medicine, Hershey, PA, USA. .,Department of Surgery, Division of Urology, The Pennsylvania State University, College of Medicine, Hershey, PA, 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
|
Tsourlakis MC, Eleftheriadou A, Stender A, Weigand P, Grupp K, Hube-Magg C, Kluth M, Schroeder C, Steurer S, Hinsch A, Luebke A, Angerer A, Wittmer C, Friedrich E, Göbel C, Büscheck F, Heinzer H, Graefen M, Simon R, Sauter G, Wilczak W, Minner S, Schlomm T, Jacobsen F. FOXA1 expression is a strong independent predictor of early PSA recurrence in ERG negative prostate cancers treated by radical prostatectomy. Carcinogenesis 2017; 38:1180-1187. [PMID: 29029032 DOI: 10.1093/carcin/bgx105] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 09/20/2017] [Indexed: 12/24/2022] Open
Abstract
FOXA1 (Fork-head box protein A1, HNF-3a) is a transcription factor involved in androgen signaling with relevance for lineage-specific gene expression of the prostate. The expression was analyzed by immunohistochemistry on a tissue microarray containing 11152 prostate cancer specimens. Results were compared with tumor phenotype, biochemical recurrence, androgen receptor expression, ETS-related gene (ERG) status and other recurrent genomic alterations. FOXA1 expression was detectable in 97.6% of 8227 interpretable cancers and considered strong in 28.5%, moderate in 46.2% and weak in 22.9% of cases. High FOXA1 expression was associated with TMPRSS2:ERG rearrangement and ERG expression (P < 0.0001). High FOXA1 expression was linked to high Gleason grade, advanced pathological tumor (pT) stage and early PSA recurrence in ERG negative cancers (P < 0.0001), while these associations were either weak or absent in ERG positive cancers. In ERG negative cancers, the prognostic role of FOXA1 expression was independent of Gleason grade, pathological tumor stage, lymph node stage, surgical margin status and preoperative PSA. Independent prognostic value became even more evident if the analysis was limited to preoperatively available features such as biopsy Gleason grade, preoperative PSA, cT stage and FOXA1 expression (P < 0.0001). Within ERG negative cancers, FOXA1 expression was also strongly associated with PTEN and 5q21 deletions (P < 0.0001). High expression of FOXA1 is an independent prognostic parameter in ERG negative prostate cancer. Thus, FOXA1 measurement might provide clinically useful information in prostate cancer.
Collapse
Affiliation(s)
| | - Agapi Eleftheriadou
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Annegret Stender
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Philipp Weigand
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Katharina Grupp
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Germany.,General, Visceral and Thoracic Surgery Department and Clinic, University Medical Center Hamburg-Eppendorf, Germany
| | - Claudia Hube-Magg
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Martina Kluth
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Cornelia Schroeder
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Germany.,General, Visceral and Thoracic Surgery Department and Clinic, University Medical Center Hamburg-Eppendorf, Germany
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Andrea Hinsch
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Andreas Luebke
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Alexander Angerer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Corinna Wittmer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Emily Friedrich
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Cosima Göbel
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Franziska Büscheck
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Hans Heinzer
- Martini-Clinic, Prostate Cancer Center and University Medical Center Hamburg-Eppendorf, Germany
| | - Markus Graefen
- Martini-Clinic, Prostate Cancer Center and University Medical Center Hamburg-Eppendorf, Germany
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Waldemar Wilczak
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Thorsten Schlomm
- Martini-Clinic, Prostate Cancer Center and University Medical Center Hamburg-Eppendorf, Germany.,Department of Urology, Section for translational Prostate Cancer Research, University Medical Center Hamburg-Eppendorf, Germany.,Centre for Cancer Biomarkers CCBIO, University of Bergen, Norway
| | - Frank Jacobsen
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Germany
| |
Collapse
|
11
|
Wang D, Feng JF, Yuan GY, Yang YH, Liu YS, Yang YW. Association between chromosomal aberration of exfoliated bladder cells in the urine and oxidative stress in patients with bladder transitional cell carcinoma. Oncol Lett 2017; 14:137-144. [PMID: 28693145 PMCID: PMC5494805 DOI: 10.3892/ol.2017.6128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 03/09/2017] [Indexed: 01/10/2023] Open
Abstract
The aim of the current study was to investigate the chromosomal aberrations of exfoliated bladder cells in the urine and blood oxidative stress in patients with bladder transitional cell carcinoma (BTCC). A total of 40 healthy controls and 246 patients with BTCC were recruited. Abnormal levels of CSP3, CSP7, CSP17 and GLPp16 were detected by fluorescence in situ hybridization (FISH) in exfoliated bladder cells in the urine of patients with BTCC. Serum total oxidant status (TOS), total antioxidant status (TAS) and oxidative stress index (OSI) were measured. Significant differences were observed in the abnormal CSP3, CSP7, CSP17, GLPp16 signals and FISH positive rate between patients with BTCC and healthy controls (P<0.001). Serum TOS, TAS and OSI were also significantly different between the two groups (P<0.001). The clinical stage of BTCC was not associated with abnormal CSP3, CSP7, CSP17, GLPp16 or FISH positive rate and oxidative stress (P>0.05). A Gamma rank correlation analysis revealed an association between the pathological grade of BTCC with abnormal CSP3, CSP7 and CSP17 as well as FISH positive rate (P<0.001). In addition, the clinical stage of BTCC was associated with serum TOS, TAS and OSI (P<0.001). Evaluation of the association between chromosomal aberrations and oxidative stress revealed that abnormal CSP3, CSP7 and CSP17 were positively associated with serum TOS and OSI (P<0.001), abnormal CSP7 and CSP17 were negatively associated with serum TAS (P<0.001), but abnormal GLPp16 was not associated with serum TOS, TAS or OSI (P>0.05). Therefore, the chromosomal aberrations of exfoliated bladder cells in the urine are associated with blood oxidative stress in patients with BTCC, and these factors may contribute to the occurrence and development of BTCC.
Collapse
Affiliation(s)
- Dong Wang
- Department of Oncology, Mianyang Central Hospital, Affiliated to Southwest Medical University, Mianyang, Sichuan 621000, P.R. China
| | - Jia-Fu Feng
- Department of Clinical Laboratory, Mianyang Central Hospital, Affiliated to Southwest Medical University, Mianyang, Sichuan 621000, P.R. China
| | - Guang-Ya Yuan
- Department of Urology, Mianyang Central Hospital, Affiliated to Southwest Medical University, Mianyang, Sichuan 621000, P.R. China
| | - Yong-Hong Yang
- Department of Pathology, Mianyang Central Hospital, Affiliated to Southwest Medical University, Mianyang, Sichuan 621000, P.R. China
| | - Yun-Shuang Liu
- Department of Clinical Laboratory, Mianyang Central Hospital, Affiliated to Southwest Medical University, Mianyang, Sichuan 621000, P.R. China
| | - Yu-Wei Yang
- Department of Clinical Laboratory, Mianyang Central Hospital, Affiliated to Southwest Medical University, Mianyang, Sichuan 621000, P.R. China
| |
Collapse
|
12
|
Wang JG, Tang WP, Liao MC, Liu YP, Ai XH. MiR-99a suppresses cell invasion and metastasis in nasopharyngeal carcinoma through targeting HOXA1. Onco Targets Ther 2017; 10:753-761. [PMID: 28228659 PMCID: PMC5312690 DOI: 10.2147/ott.s126781] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Background Recent studies reported that miRNAs play important roles in the carcinogenesis and progression of nasopharyngeal carcinoma (NPC). Therefore, further studies are warranted to better elucidate the function and mechanism of miRNAs in NPC. Methods Quantitative reverse transcription-polymerase chain reaction (RT-PCR) was used to detect the miR-99a expression in NPC cell lines and tissue samples. Wound healing, transwell migration and invasion, and lung metastatic colonization assays were performed to determine NPC cell migratory, invasive and metastatic abilities of NPC cells. Luciferase reporter assays, quantitative RT-PCR and Western blotting were used to validate the target of miR-99a. Results We found that miR-99a was significantly downregulated in NPC cell lines and tissue samples. Ectopic overexpression of miR-99a significantly inhibited NPC cell migration and invasion in vitro, and suppressed lung macroscopic and microscopic metastatic colonization in vivo. Conversely, silencing of miR-99a significantly promoted the migratory and invasive abilities of NPC cells. Furthermore, HOXA1 was validated as a direct target of miR-99a, and ectopic expression of HOXA1 could rescue the suppressive effect of miR-99a overexpression on NPC cell migration and invasion. Conclusion Together, these results indicated that miR-99a could inhibit NPC invasion and metastasis by targeting HOXA1, thus providing a novel potential target for miRNA-based treatment for NPC patients in the future.
Collapse
Affiliation(s)
- Jian-Gang Wang
- Department of Radiation Oncology, The First Affiliated Hospital of University of South China, Hengyang; Department of Oncology, Shaoyang Hospital of TCM, Shaoyang, People's Republic of China
| | - Wu-Ping Tang
- Department of Oncology, Shaoyang Hospital of TCM, Shaoyang, People's Republic of China
| | - Ming-Chu Liao
- Department of Radiation Oncology, The First Affiliated Hospital of University of South China, Hengyang
| | - Yan-Ping Liu
- Department of Radiation Oncology, The First Affiliated Hospital of University of South China, Hengyang
| | - Xiao-Hong Ai
- Department of Radiation Oncology, The First Affiliated Hospital of University of South China, Hengyang
| |
Collapse
|
13
|
Warrick JI, Walter V, Yamashita H, Chung E, Shuman L, Amponsa VO, Zheng Z, Chan W, Whitcomb TL, Yue F, Iyyanki T, Kawasawa YI, Kaag M, Guo W, Raman JD, Park JS, DeGraff DJ. FOXA1, GATA3 and PPARɣ Cooperate to Drive Luminal Subtype in Bladder Cancer: A Molecular Analysis of Established Human Cell Lines. Sci Rep 2016; 6:38531. [PMID: 27924948 PMCID: PMC5141480 DOI: 10.1038/srep38531] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 11/10/2016] [Indexed: 12/22/2022] Open
Abstract
Discrete bladder cancer molecular subtypes exhibit differential clinical aggressiveness and therapeutic response, which may have significant implications for identifying novel treatments for this common malignancy. However, research is hindered by the lack of suitable models to study each subtype. To address this limitation, we classified bladder cancer cell lines into molecular subtypes using publically available data in the Cancer Cell Line Encyclopedia (CCLE), guided by genomic characterization of bladder cancer by The Cancer Genome Atlas (TCGA). This identified a panel of bladder cancer cell lines which exhibit genetic alterations and gene expression patterns consistent with luminal and basal molecular subtypes of human disease. A subset of bladder cancer cell lines exhibit in vivo histomorphologic patterns consistent with luminal and basal subtypes, including papillary architecture and squamous differentiation. Using the molecular subtype assignments, and our own RNA-seq analysis, we found overexpression of GATA3 and FOXA1 cooperate with PPARɣ activation to drive transdifferentiation of a basal bladder cancer cells to a luminial phenotype. In summary, our analysis identified a set of human cell lines suitable for the study of molecular subtypes in bladder cancer, and furthermore indicates a cooperative regulatory network consisting of GATA3, FOXA1, and PPARɣ drive luminal cell fate.
Collapse
Affiliation(s)
- Joshua I Warrick
- Department of Pathology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA.,Department of Surgery, Division of Urology, Pennsylvania State University College of Medicine, PA, USA
| | - Vonn Walter
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, PA, USA.,Department of Public Health Sciences, Pennsylvania State University College of Medicine, PA, USA
| | - Hironobu Yamashita
- Department of Pathology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Eunah Chung
- Division of Pediatric Urology and Developmental Biology, Cincinnati Children's Hospital Medical Center, OH, USA
| | - Lauren Shuman
- Department of Pathology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA.,Department of Surgery, Division of Urology, Pennsylvania State University College of Medicine, PA, USA
| | - Vasty Osei Amponsa
- Department of Pathology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Zongyu Zheng
- Department of Pathology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Wilson Chan
- Department of Pathology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA.,Department of Surgery, Division of Urology, Pennsylvania State University College of Medicine, PA, USA
| | - Tiffany L Whitcomb
- Department of Comparative Medicine, Pennsylvania State University College of Medicine, PA, USA
| | - Feng Yue
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, PA, USA.,Institute for Personalized Medicine, Pennsylvania State University College of Medicine, PA, USA
| | - Tejaswi Iyyanki
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, PA, USA
| | - Yuka I Kawasawa
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, PA, USA.,Institute for Personalized Medicine, Pennsylvania State University College of Medicine, PA, USA
| | - Matthew Kaag
- Department of Surgery, Division of Urology, Pennsylvania State University College of Medicine, PA, USA
| | - Wansong Guo
- Department of Surgery, Division of Urology, Changchun Central Hospital, Changchun, China
| | - Jay D Raman
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, PA, USA
| | - Joo-Seop Park
- Division of Pediatric Urology and Developmental Biology, Cincinnati Children's Hospital Medical Center, OH, USA
| | - David J DeGraff
- Department of Pathology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA.,Department of Surgery, Division of Urology, Pennsylvania State University College of Medicine, PA, USA.,Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, PA, USA
| |
Collapse
|
14
|
Abstract
Genomic and transcriptional studies have identified discrete molecular subtypes of bladder cancer. These observations could be the starting point to identify new treatments. Several members of the forkhead box (FOX) superfamily of transcription factors have been found to be differentially expressed in the different bladder cancer subtypes. In addition, the FOXA protein family are key regulators of embryonic bladder development and patterning. Both experimental and clinical data support a role for FOXA1 and FOXA2 in urothelial carcinoma. FOXA1 is expressed in embryonic and adult urothelium and its expression is altered in urothelial carcinomas and across disparate molecular bladder cancer subtypes. FOXA2 is normally absent from the adult urothelium, but developmental studies identified FOXA2 as a marker of a transient urothelial progenitor cell population during bladder development. Studies also implicate FOXA2 in bladder cancer and several other FOX proteins might be involved in development and/or progression of this disease; for example, FOXA1 and FOXO3A have been associated with clinical patient outcomes. Future studies should investigate to what extent and by which mechanisms FOX proteins might be directly involved in bladder cancer pathogenesis and treatment responses.
Collapse
|
15
|
FOXA1 overexpression mediates endocrine resistance by altering the ER transcriptome and IL-8 expression in ER-positive breast cancer. Proc Natl Acad Sci U S A 2016; 113:E6600-E6609. [PMID: 27791031 DOI: 10.1073/pnas.1612835113] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Forkhead box protein A1 (FOXA1) is a pioneer factor of estrogen receptor α (ER)-chromatin binding and function, yet its aberration in endocrine-resistant (Endo-R) breast cancer is unknown. Here, we report preclinical evidence for a role of FOXA1 in Endo-R breast cancer as well as evidence for its clinical significance. FOXA1 is gene-amplified and/or overexpressed in Endo-R derivatives of several breast cancer cell line models. Induced FOXA1 triggers oncogenic gene signatures and proteomic profiles highly associated with endocrine resistance. Integrated omics data reveal IL8 as one of the most perturbed genes regulated by FOXA1 and ER transcriptional reprogramming in Endo-R cells. IL-8 knockdown inhibits tamoxifen-resistant cell growth and invasion and partially attenuates the effect of overexpressed FOXA1. Our study highlights a role of FOXA1 via IL-8 signaling as a potential therapeutic target in FOXA1-overexpressing ER-positive tumors.
Collapse
|
16
|
Ji J, Xu M, Tu J, Zhao Z, Gao J, Chen M, Song J, Zhu H, Cheng X, Hui J, Lan X, Yang X. MiR-155 and its functional variant rs767649 contribute to the susceptibility and survival of hepatocellular carcinoma. Oncotarget 2016; 7:60303-60309. [PMID: 27531892 PMCID: PMC5312385 DOI: 10.18632/oncotarget.11206] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 07/12/2016] [Indexed: 12/25/2022] Open
Abstract
Hepatocellular carcinoma (HCC) ranks the fourth common cancer and the third common cause of cancer mortality among Chinese population. The development of hepatocellular carcinoma (HCC) were confirmed to be involved in complex interactions between environmental and genetic factors. MicroRNAs (miRNAs) have been found to play an important role in tumorigenesis and metastasis. Emerging evidence suggested that upregulation of miR-155, one of the best characterized miRNAs, could serve as a promising marker for the diagnosis and prognosis of many cancers, except for HCC. In current we tested the hypothesis that functional variant rs767649 located in the flanking region of miR-155 gene contributes to the development and survival of HCC. We identified that functional variant rs767649 in miR-155 regulation region was associated with risk and survival of HCC. The minor allele of rs767649 was significantly associated with an increased risk of HCC (OR=1.23, 95% CI=1.11-1.36, P = 7.97x10-5). The genotype TT of rs767649 was significantly associated with a 1.94 fold poor survival of HCC (HR=1.94, 95% CI=1.01-3.79), while 1.15 fold for genotype AT (HR=1.15, 95% CI=1.06-1.25). Results showed that miR-155 was highly overexpressed in HCC tissues, compared with the adjacent normal tissues (P<0.001). The allele T contributes to higher expression of miR-155 in both the HCC tissues and the adjacent non-tumor tissues (P< 0.01). Our findings suggested that miR-155 and its functional variant rs767649 might contribute to the increased risk and poor prognosis of HCC, highlighting the importance of miR-155 in the prevention and prognosis of HCC.
Collapse
Affiliation(s)
- Jiansong Ji
- Department of Radiology, Affiliated Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, The Central Hospital of Zhejiang, Zhejiang 323000, P. R. China
| | - Min Xu
- Department of Radiology, Affiliated Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, The Central Hospital of Zhejiang, Zhejiang 323000, P. R. China
| | - Jianfei Tu
- Department of Radiology, Affiliated Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, The Central Hospital of Zhejiang, Zhejiang 323000, P. R. China
| | - Zhongwei Zhao
- Department of Radiology, Affiliated Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, The Central Hospital of Zhejiang, Zhejiang 323000, P. R. China
| | - Jun Gao
- Department of Hepatobiliary Surgery, Beijing Chao-Yang Hospital Affiliated with Capital Medical University, Beijing 100043, P. R. China
| | - Minjiang Chen
- Department of Radiology, Affiliated Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, The Central Hospital of Zhejiang, Zhejiang 323000, P. R. China
| | - Jingjing Song
- Department of Radiology, Affiliated Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, The Central Hospital of Zhejiang, Zhejiang 323000, P. R. China
| | - Haidong Zhu
- Department of Radiology, Zhong-da Hospital, Medical School, Southeast University, Nanjing 210009, P. R. China
| | - Xingyao Cheng
- Department of Radiology, Affiliated Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, The Central Hospital of Zhejiang, Zhejiang 323000, P. R. China
| | - Junguo Hui
- Department of Radiology, Affiliated Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, The Central Hospital of Zhejiang, Zhejiang 323000, P. R. China
| | - Xilin Lan
- Department of Radiology, Affiliated Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, The Central Hospital of Zhejiang, Zhejiang 323000, P. R. China
| | - Xiaoming Yang
- Department of Radiology, Lab-Yang, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
17
|
Wang ZQ, Faddaoui A, Bachvarova M, Plante M, Gregoire J, Renaud MC, Sebastianelli A, Guillemette C, Gobeil S, Macdonald E, Vanderhyden B, Bachvarov D. BCAT1 expression associates with ovarian cancer progression: possible implications in altered disease metabolism. Oncotarget 2016; 6:31522-43. [PMID: 26372729 PMCID: PMC4741622 DOI: 10.18632/oncotarget.5159] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 08/28/2015] [Indexed: 12/17/2022] Open
Abstract
Previously, we have identified the branched chain amino-acid transaminase 1 (BCAT1) gene as notably hypomethylated in low-malignant potential (LMP) and high-grade (HG) serous epithelial ovarian tumors, compared to normal ovarian tissues. Here we show that BCAT1 is strongly overexpressed in both LMP and HG serous epithelial ovarian tumors, which probably correlates with its hypomethylated status. Knockdown of the BCAT1 expression in epithelial ovarian cancer (EOC) cells led to sharp decrease of cell proliferation, migration and invasion and inhibited cell cycle progression. BCAT1 silencing was associated with the suppression of numerous genes and pathways known previously to be implicated in ovarian tumorigenesis, and the induction of some tumor suppressor genes (TSGs). Moreover, BCAT1 suppression resulted in downregulation of numerous genes implicated in lipid production and protein synthesis, suggesting its important role in controlling EOC metabolism. Further metabolomic analyses were indicative for significant depletion of most amino acids and different phospho- and sphingolipids following BCAT1 knockdown. Finally, BCAT1 suppression led to significantly prolonged survival time in xenograft model of advanced peritoneal EOC. Taken together, our findings provide new insights about the functional role of BCAT1 in ovarian carcinogenesis and identify this transaminase as a novel EOC biomarker and putative EOC therapeutic target.
Collapse
Affiliation(s)
- Zhi-Qiang Wang
- Department of Molecular Medicine, Laval University, Québec PQ, Canada.,Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec PQ, Canada
| | - Adnen Faddaoui
- Department of Molecular Medicine, Laval University, Québec PQ, Canada.,Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec PQ, Canada
| | | | - Marie Plante
- Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec PQ, Canada.,Department of Obstetrics and Gynecology, Laval University, Québec PQ, Canada
| | - Jean Gregoire
- Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec PQ, Canada.,Department of Obstetrics and Gynecology, Laval University, Québec PQ, Canada
| | - Marie-Claude Renaud
- Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec PQ, Canada.,Department of Obstetrics and Gynecology, Laval University, Québec PQ, Canada
| | - Alexandra Sebastianelli
- Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec PQ, Canada.,Department of Obstetrics and Gynecology, Laval University, Québec PQ, Canada
| | - Chantal Guillemette
- Centre de recherche du CHU de Québec, CHUL, Québec PQ, Canada.,Faculty of Pharmacy, Laval University, Québec PQ, Canada
| | - Stéphane Gobeil
- Department of Molecular Medicine, Laval University, Québec PQ, Canada.,Centre de recherche du CHU de Québec, CHUL, Québec PQ, Canada
| | - Elizabeth Macdonald
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Barbara Vanderhyden
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Dimcho Bachvarov
- Department of Molecular Medicine, Laval University, Québec PQ, Canada.,Centre de recherche du CHU de Québec, L'Hôtel-Dieu de Québec, Québec PQ, Canada
| |
Collapse
|
18
|
Lv ZD, Kong B, Liu XP, Jin LY, Dong Q, Li FN, Wang HB. miR-655 suppresses epithelial-to-mesenchymal transition by targeting Prrx1 in triple-negative breast cancer. J Cell Mol Med 2016; 20:864-73. [PMID: 26820102 PMCID: PMC4831358 DOI: 10.1111/jcmm.12770] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 11/28/2015] [Indexed: 12/14/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive breast cancer subtype that lacks effective targeted therapies. The epithelial-to-mesenchymal transition (EMT) is a key contributor in the metastatic process. In this study, we found that miR-655 was down-regulated in TNBC, and its expression levels were associated with molecular-based classification and lymph node metastasis in breast cancer. These findings led us to hypothesize that miR-655 overexpression may inhibit EMT and its associated traits of TNBC. Ectopic expression of miR-655 not only induced the up-regulation of cytokeratin and decreased vimentin expression but also suppressed migration and invasion of mesenchymal-like cancer cells accompanied by a morphological shift towards the epithelial phenotype. In addition, we found that miR-655 was negatively correlated with Prrx1 in cell lines and clinical samples. Overexpression of miR-655 significantly suppressed Prrx1, as demonstrated by Prrx1 3'-untranslated region luciferase report assay. Our study demonstrated that miR-655 inhibits the acquisition of the EMT phenotype in TNBC by down-regulating Prrx1, thereby inhibiting cell migration and invasion during cancer progression.
Collapse
MESH Headings
- 3' Untranslated Regions
- Adult
- Animals
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Cell Line, Tumor
- Cell Movement
- Epithelial-Mesenchymal Transition/genetics
- Female
- Gene Expression Regulation, Neoplastic
- Genes, Reporter
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Humans
- Keratins/genetics
- Keratins/metabolism
- Luciferases/genetics
- Luciferases/metabolism
- Lymphatic Metastasis
- Mice
- Mice, Nude
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Middle Aged
- Neoplasm Invasiveness
- Protein Binding
- Signal Transduction
- Vimentin/genetics
- Vimentin/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Zhi-Dong Lv
- Department of Breast Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bin Kong
- Department of Breast Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiang-Ping Liu
- Central Laboratory of Molecular Biology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Li-Ying Jin
- Cerebrovascular Disease Research Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qian Dong
- Department of Pediatric Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fu-Nian Li
- Department of Breast Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hai-Bo Wang
- Department of Breast Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
19
|
CAO QIFENG, WANG NING, QI JUAN, GU ZHENGQIN, SHEN HAIBO. Long non‑coding RNA‑GAS5 acts as a tumor suppressor in bladder transitional cell carcinoma via regulation of chemokine (C‑C motif) ligand 1 expression. Mol Med Rep 2016; 13:27-34. [PMID: 26548923 PMCID: PMC4686088 DOI: 10.3892/mmr.2015.4503] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 07/21/2015] [Indexed: 12/31/2022] Open
Abstract
Long non‑coding RNAs (lncRNAs) have important roles in diverse biological processes, including transcriptional regulation, cell growth and tumorigenesis. The present study aimed to investigate whether lncRNA‑growth arrest‑specific (GAS)5 regulated bladder cancer progression via regulation of chemokine (C‑C) ligand (CCL)1 expression. The viability of BLX bladder cancer cells was detected using a Cell Counting kit‑8 assay, and cell apoptosis was assessed by annexin V‑propidium iodide double‑staining. The expression levels of specific genes and proteins were analyzed by reverse transcription‑quantitative polymerase chain reaction and western blotting, respectively. In addition, cells were transfected with small interfering (si)RNAs or recombinant GAS5 in order to silence or overexpress GAS5, respectively. The results of the present study demonstrated that knockdown of GAS5 expression promoted bladder cancer cell proliferation, whereas overexpression of GAS5 suppressed cell proliferation. Furthermore, knockdown of GAS5 resulted in an increased percentage of cells in S and G2 phase, and a decreased percentage of cells in G1 phase. In addition, the present study performed a hierarchical cluster analysis of differentially expressed lncRNAs in bladder cancer cells and detected that CCL1 overexpression resulted in an upregulation of GAS5, which may improve the ability of cells to regulate a stress response in vitro. Furthermore, knockdown of GAS5 expression increased the mRNA and protein expression of CCL1 in bladder cancer cells. Gain‑of‑function and loss‑of‑function studies demonstrated that GAS5 was able to inhibit bladder cancer cell proliferation, at least in part, by suppressing the expression of CCL1. The results of the present study demonstrated that GAS5 was able to suppress bladder cancer cell proliferation, at least partially, by suppressing the expression of CCL1. The results of the present study may provide a basis for developing novel effective treatment strategies against bladder cancer.
Collapse
Affiliation(s)
| | | | - JUAN QI
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - ZHENGQIN GU
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| | - HAIBO SHEN
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, P.R. China
| |
Collapse
|
20
|
Yang J, Yuan D, Li J, Zheng S, Wang B. miR-186 downregulates protein phosphatase PPM1B in bladder cancer and mediates G1-S phase transition. Tumour Biol 2015; 37:4331-41. [DOI: 10.1007/s13277-015-4117-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 09/20/2015] [Indexed: 02/05/2023] Open
|
21
|
He Y, Chevillet JR, Liu G, Kim TK, Wang K. The effects of microRNA on the absorption, distribution, metabolism and excretion of drugs. Br J Pharmacol 2015; 172:2733-47. [PMID: 25296724 PMCID: PMC4439871 DOI: 10.1111/bph.12968] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 09/18/2014] [Accepted: 09/26/2014] [Indexed: 12/17/2022] Open
Abstract
The importance of genetic factors (e.g. sequence variation) in the absorption, distribution, metabolism, excretion (ADME) and overall efficacy of therapeutic agents is well established. Our ability to identify, interpret and utilize these factors is the subject of much clinical investigation and therapeutic development. However, drug ADME and efficacy are also heavily influenced by epigenetic factors such as DNA/histone methylation and non-coding RNAs [especially microRNAs (miRNAs)]. Results from studies using tools, such as in silico miRNA target prediction, in vitro functional assays, nucleic acid profiling/sequencing and high-throughput proteomics, are rapidly expanding our knowledge of these factors and their effects on drug metabolism. Although these studies reveal a complex regulation of drug ADME, an increased understanding of the molecular interplay between the genome, epigenome and transcriptome has the potential to provide practically useful strategies to facilitate drug development, optimize therapeutic efficacy, circumvent adverse effects, yield novel diagnostics and ultimately become an integral component of personalized medicine.
Collapse
Affiliation(s)
- Y He
- Institute of Medical Systems Biology, Guangdong Medical CollegeDongguan, Guangdong, China
| | | | - G Liu
- Department of Chemistry and Biochemistry, North Dakota State UniversityFargo, ND, USA
| | - T K Kim
- Institute for Systems BiologySeattle, WA, USA
| | - K Wang
- Institute for Systems BiologySeattle, WA, USA
| |
Collapse
|
22
|
Regulation of growth of human bladder cancer by miR-192. Tumour Biol 2015; 36:3791-7. [PMID: 25566965 DOI: 10.1007/s13277-014-3020-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 12/23/2014] [Indexed: 01/30/2023] Open
Abstract
The regulation of microRNA-192 (miR-192) is impaired in many cancers. Here, we investigated the role of miR-192 in the proliferation, cell cycle progression, and apoptosis of bladder cancer cells. Human bladder cancer cells were transfected with human miR-192 precursor or non-specific control miRNA. The effect of miR-192 on cell proliferation was assessed by a MTT assay. The effects of miR-192 on cell cycle regulation and apoptosis were evaluated by flow cytometry. Western blot was used to analyze the protein levels of cyclin D1, p21, p27, Bcl-2, Bax, and Mcl-1. We found that overexpression of miR-192 significantly decreased the proliferation of bladder cancer cells by 22 and 54 % at 48 and 72 h, respectively. MiR-192-overexpressing cells exhibited a significant increase in G0/G1 phase and a significant decrease in S phase compared to the control miRNA-transfected cells. Moreover, overexpression of miR-192 significantly induced apoptotic death in bladder cancer cells, increased the levels of p21, p27, and Bax, and decreased the levels of cyclin D1, Bcl-2, and Mcl-1. Taken together, these data suggest that miR-192 may be a suppressor for bladder cancer cells by cell cycle regulation.
Collapse
|