1
|
Zaman R, Islam RA, Chowdhury EH. Evolving therapeutic proteins to precisely kill cancer cells. J Control Release 2022; 351:779-804. [DOI: 10.1016/j.jconrel.2022.09.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/02/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 10/31/2022]
|
2
|
Silveira DA, Gupta S, Sinigaglia M, Mombach JCM. The Wnt pathway can stabilize hybrid phenotypes in the epithelial-mesenchymal transition: A logical modeling approach. Comput Biol Chem 2022; 99:107714. [PMID: 35763962 DOI: 10.1016/j.compbiolchem.2022.107714] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/30/2022] [Revised: 05/27/2022] [Accepted: 06/09/2022] [Indexed: 11/28/2022]
Abstract
The Wnt pathway is important to regulate a variety of biochemical functions and can contribute to cancer development through its influence on the epithelial-mesenchymal transition (EMT). Multiple circuits have been reported to participate in the regulation of the Wnt signaling, however, the way these circuits coordinately regulate this signaling is still unclear. Moreover, the mechanisms responsible for the appearance of hybrid phenotypes (cells presenting both E and M features) are not well determined. The hybrid phenotype can present much higher metastatic potential than the mesenchymal phenotype. In this study, we propose a Boolean model of the Wnt pathway signaling contemplating recent published biochemical information on hepatocarcinoma. The model presents good coherence with experimental data for perturbed and wild-type cases. With the model, we propose two new molecular circuits involving several molecules that can stabilize hybrid states during the EMT. Moreover, we found that the two well studied circuits, AKT1/β-catenin and SNAIL1/miR-34, can cooperate with the predicted ones to favor the stabilization of the hybrid states. These findings highlight some possible unrecognized mechanisms during Wnt signaling and may provide alternative therapeutic strategies to control cancer metastatization.
Collapse
Affiliation(s)
- Daner Acunha Silveira
- Department of Physics, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil; Children's Cancer Institute, Porto Alegre, Rio Grande do Sul, Brazil
| | - Shantanu Gupta
- Department of Physics, Universidade Federal de Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | | | | |
Collapse
|
3
|
Azcue P, Guerrero Setas D, Encío I, Ibáñez-Beroiz B, Mercado M, Vera R, Gómez-Dorronsoro ML. A Novel Prognostic Biomarker Panel for Early-Stage Colon Carcinoma. Cancers (Basel) 2021; 13:5909. [PMID: 34885019 PMCID: PMC8656725 DOI: 10.3390/cancers13235909] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/28/2021] [Revised: 11/18/2021] [Accepted: 11/22/2021] [Indexed: 12/09/2022] Open
Abstract
Molecular characterization of colorectal cancer has helped us understand better the biology of the disease. However, previous efforts have yet to provide significant clinical value in order to be integrated into clinical practice for patients with early-stage colon cancer (CC). The purpose of this study was to assess PD-L1, GLUT-1, e-cadherin, MUC2, CDX2, and microsatellite instability (dMMR) and to propose a risk-panel with prognostic capabilities. Biomarkers were immunohistochemically assessed through tissue microarrays in a cohort of 144 patients with stage II/III colon cancer. A biomarker panel consisting of PD-L1, GLUT-1, dMMR, and potentially CDX2 was constructed that divided patients into low, medium, and high risk of overall survival or disease-free survival (DFS) in equally sized groups. Compared with low-risk patients, medium-risk patients have almost twice the risk of death (HR = 2.10 (0.99-4.46), p = 0.054), while high-risk patients have almost four times the risk (HR = 3.79 (1.77-8.11), p = 0.001). The multivariate goodness of fit was 0.756 and was correlated with Kaplan-Meier curves (p = 0.002). Consistent results were found for DFS. This study provides a critical basis for the future development of an immunohistochemical assessment capable of discerning early-stage CC patients as a function of their prognosis. This tool may aid with treatment personalization in daily clinical practice and improve survival outcomes.
Collapse
Affiliation(s)
- Pablo Azcue
- Department of Health Science, Public University of Navarra, 31008 Pamplona, Spain; (I.E.); (B.I.-B.)
| | - David Guerrero Setas
- Department of Pathology, University Hospital of Navarra, 31008 Pamplona, Spain; (D.G.S.); (M.M.)
- Campus Arrosadia, Public University of Navarra, 31006 Pamplona, Spain
- Molecular Pathology of Cancer Group–Navarrabiomed, 31008 Pamplona, Spain
- Department of Medical Oncology, University Hospital of Navarra, 31008 Pamplona, Spain;
| | - Ignacio Encío
- Department of Health Science, Public University of Navarra, 31008 Pamplona, Spain; (I.E.); (B.I.-B.)
- Institute for Health Research Navarra (IdISNA), 31008 Pamplona, Spain
| | - Berta Ibáñez-Beroiz
- Department of Health Science, Public University of Navarra, 31008 Pamplona, Spain; (I.E.); (B.I.-B.)
- Institute for Health Research Navarra (IdISNA), 31008 Pamplona, Spain
- Unit of Methodology-Navarrabiomed-University Hospital of Navarra, 31008 Pamplona, Spain
- Research Network on Health Services Research and Chronic Diseases (REDISSEC), 31008 Pamplona, Spain
| | - María Mercado
- Department of Pathology, University Hospital of Navarra, 31008 Pamplona, Spain; (D.G.S.); (M.M.)
| | - Ruth Vera
- Department of Medical Oncology, University Hospital of Navarra, 31008 Pamplona, Spain;
- Institute for Health Research Navarra (IdISNA), 31008 Pamplona, Spain
| | - María Luisa Gómez-Dorronsoro
- Department of Pathology, University Hospital of Navarra, 31008 Pamplona, Spain; (D.G.S.); (M.M.)
- Institute for Health Research Navarra (IdISNA), 31008 Pamplona, Spain
| |
Collapse
|
4
|
Na L, Wang Z, Bai Y, Sun Y, Dong D, Wang W, Zhao C. WNT7B represses epithelial-mesenchymal transition and stem-like properties in bladder urothelial carcinoma. Biochim Biophys Acta Mol Basis Dis 2021; 1868:166271. [PMID: 34562599 DOI: 10.1016/j.bbadis.2021.166271] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/30/2021] [Revised: 09/01/2021] [Accepted: 09/15/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Recurrence and metastasis are the major problems of bladder urothelial carcinoma, which mainly attribute to tumor cell stemness, epithelial-mesenchymal transition (EMT) and chemoresistance. METHODS TCGA database was interrogated for gene mRNA expression in bladder urothelial carcinoma samples. CCLE database was interrogated for gene mRNA expression in bladder cancer cell lines. The correlation between two genes was analyzed by Pearson statistics. 37 human bladder urothelial carcinoma specimens were adopted for immunohistochemistry. Bladder cancer cells RT4, J82, and UM-UC-3 were used to carry out loss and gain of function studies. Kaplan-Meier method was performed to analyze the overall survival. FINDINGS WNT7B is downregulated in high-grade bladder urothelial carcinomas. Low WNT7B expression is associated with unfavorable prognosis. Loss and gain of function studies showed that WNT7B inhibits bladder urothelial carcinoma cell EMT, stem-like properties and chemoresistance. FZD5, a specific receptor for WNT7B, mediates WNT7B signaling. ELF3 is a downstream component of WNT7B signaling, which transcriptionally modulates NOTCH1, a tumor suppressor in bladder urothelial carcinoma. INTERPRETATION These data demonstrate that WNT7B/FZD5-ELF3-NOTCH1 signaling functions as a tumor-suppressing pathway in bladder urothelial carcinoma.
Collapse
Affiliation(s)
- Lei Na
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China; Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhuo Wang
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Yu Bai
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China; Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Sun
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Dan Dong
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Wei Wang
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China.
| | - Chenghai Zhao
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China.
| |
Collapse
|
5
|
El Aliani A, El-Abid H, El Mallali Y, Attaleb M, Ennaji MM, El Mzibri M. Association between Gene Promoter Methylation and Cervical Cancer Development: Global Distribution and A Meta-analysis. Cancer Epidemiol Biomarkers Prev 2021; 30:450-459. [DOI: 10.1158/1055-9965.epi-20-0833] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/18/2020] [Revised: 09/27/2020] [Accepted: 12/31/2020] [Indexed: 12/24/2022] Open
|
6
|
Deb B, Kumar P. Tumor Heterogeneity and Phenotypic Plasticity in Bladder Carcinoma. J Indian Inst Sci 2020. [DOI: 10.1007/s41745-020-00183-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/23/2022]
|
7
|
Jin Y, Liu J, Liu Y, Liu Y, Guo G, Yu S, An R. Oxymatrine Inhibits Renal Cell Carcinoma Progression by Suppressing β-Catenin Expression. Front Pharmacol 2020; 11:808. [PMID: 32581789 PMCID: PMC7289957 DOI: 10.3389/fphar.2020.00808] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/13/2019] [Accepted: 05/18/2020] [Indexed: 01/17/2023] Open
Abstract
Aims Oxymatrine (OMT) has been identified to possess immunomodulatory, antiinflammatory and anticancer properties. This study aimed to investigate its precise function and the underlying molecular mechanisms in renal cell carcinoma progression. Methods The antineoplastic effect of oxymatrine was investigated by CCK-8 assay, cell cycle analysis, apoptosis assay, wound healing experiment, transwell assay, and drug-sensitivity analysis in renal cancer cells following oxymatrine treatment. The modulation of oxymatrine on β-catenin was analyzed through western blot and immunofluorescence assay. β-catenin overexpression was employed to determine the key role of β-catenin in oxymatrine-inhibited renal cell carcinoma in vitro. In addition, animal model was established to investigate the effect of oxymatrine on tumor growth in vivo. Results Oxymatrine inhibited renal cell carcinoma progression in vitro, including cell proliferation, apoptosis, migration, invasion and chemotherapy sensitivity. Further mechanistic studies demonstrated that oxymatrine exerted its antineoplastic effect through suppressing the expression of β-catenin. Moreover, in nude mice model, oxymatrine exhibited remarkable inhibition of tumor growth, which was consistent with our in vitro results. Conclusions Our findings illuminate oxymatrine as an effective antitumor agent in renal cell carcinoma, and suggest it a promising therapeutic application in renal cell carcinoma treatment.
Collapse
Affiliation(s)
- Yinshan Jin
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiannan Liu
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yadong Liu
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yang Liu
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guiying Guo
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shiliang Yu
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ruihua An
- Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
8
|
Mencucci MV, Lapyckyj L, Rosso M, Besso MJ, Belgorosky D, Isola M, Vanzulli S, Lodillinsky C, Eiján AM, Tejerizo JC, Gonzalez MI, Zubieta ME, Vazquez-Levin MH. Ephrin-B1 Is a Novel Biomarker of Bladder Cancer Aggressiveness. Studies in Murine Models and in Human Samples. Front Oncol 2020; 10:283. [PMID: 32292715 PMCID: PMC7119101 DOI: 10.3389/fonc.2020.00283] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/12/2019] [Accepted: 02/18/2020] [Indexed: 01/11/2023] Open
Abstract
Bladder cancer (BC) is the ninth most common cancer worldwide, but molecular changes are still under study. During tumor progression, Epithelial cadherin (E-cadherin) expression is altered and β-catenin may be translocated to the nucleus, where it acts as co-transcription factor of tumor invasion associated genes. This investigation further characterizes E-cadherin and β-catenin associated changes in BC, by combining bioinformatics, an experimental murine cell model (MB49/MB49-I) and human BC samples. In in silico studies, a DisGeNET (gene-disease associations database) analysis identified CDH1 (E-cadherin gene) as one with highest score among 130 BC related-genes. COSMIC mutation analysis revealed CDH1 low mutations rates. Compared to MB49 control BC cells, MB49-I invasive cells showed decreased E-cadherin expression, E- to P-cadherin switch, higher β-catenin nuclear signal and lower cytoplasmic p-Ser33-β-catenin signal, higher Ephrin-B1 ligand and EphB2 receptor expression, higher Phospho-Stat3 and Urokinase-type Plasminogen Activator (UPA), and UPA receptor expression. MB49-I cells transfected with Ephrin-B1 siRNA showed lower migratory and invasive capacity than control cells (scramble siRNA). By immunohistochemistry, orthotopic MB49-I tumors had lower E-cadherin, increased nuclear β-catenin, lower pSer33-β-catenin cytoplasmic signal, and higher Ephrin-B1 expression than MB49 tumors. Similar changes were found in human BC tumors, and 83% of infiltrating tumors depicted a high Ephrin-B1 stain. An association between higher Ephrin-B1 expression and higher stage and tumor grade was found. No association was found between abnormal E-cadherin signal, Ephrin-B1 expression or clinical-pathological parameter. This study thoroughly analyzed E-cadherin and associated changes in BC, and reports Ephrin-B1 as a new marker of tumor aggressiveness.
Collapse
Affiliation(s)
- María Victoria Mencucci
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
| | - Lara Lapyckyj
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
| | - Marina Rosso
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
| | - María José Besso
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
| | - Denise Belgorosky
- Research Area, Instituto de Oncología Angel H. Roffo, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mariana Isola
- Departamento de Anatomía Patológica, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | | | - Catalina Lodillinsky
- Research Area, Instituto de Oncología Angel H. Roffo, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ana María Eiján
- Research Area, Instituto de Oncología Angel H. Roffo, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Juan Carlos Tejerizo
- Departamento de Urología, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | | | - María Ercilia Zubieta
- Departamento de Urología, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Mónica Hebe Vazquez-Levin
- Laboratorio de Estudios de la Interacción Celular en Reproducción y Cáncer, Instituto de Biología y Medicina Experimental (IBYME; CONICET-FIBYME), Buenos Aires, Argentina
| |
Collapse
|
9
|
Garg M, Maurya N. WNT/β-catenin signaling in urothelial carcinoma of bladder. World J Nephrol 2019; 8:83-94. [PMID: 31624709 PMCID: PMC6794554 DOI: 10.5527/wjn.v8.i5.83] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 04/25/2019] [Revised: 08/14/2019] [Accepted: 09/16/2019] [Indexed: 02/06/2023] Open
Abstract
Urothelial carcinoma of bladder is the second most prevalent genitourinary disease. It is a highly heterogeneous disease as it represents a spectrum of neoplasms, including non-muscle invasive bladder cancer (NMIBC), muscle invasive bladder cancer (MIBC) and metastatic lesions. Genome-wide approaches and candidate gene analysis suggest that malignant transformation of the bladder is multifactorial and a multitude of genes are involved in the development of MIBC or NMIBC phenotypes. Wnt signaling is being examined to control and maintain balance between stemness and differentiation in adult stem cell niches. Owing to its participation in urothelial development and maintenance of adult urothelial tissue homeostasis, the components of Wnt signaling are reported as an important diagnostic and prognostic markers as well as novel therapeutic targets. Mutations/epigenetic alterations in the key molecules of Wnt/β-catenin canonical pathway have been linked with tumorigenesis, development of drug resistance and enhanced survival. Present review extends our understanding on the functions of key regulatory molecules of canonical Wnt/β-catenin pathway in urothelial tumorigenesis by inducing cancer stem cell phenotype (UCSCs). UCSCs may be responsible for tumor heterogeneity, high recurrence rates and complex biological behavior of bladder cancer. Therefore, understanding the role of UCSCs and the regulatory mechanisms that are responsible for high relapse rates and metastasis could help to develop pathway inhibitors and augment current therapies. Potential implications in the treatment of urothelial carcinoma of bladder by targeting this pathway primarily in UCSCs as well as in bulk tumor population that are responsible for high relapse rates and metastasis may facilitate potential therapeutic avenues and better prognosis.
Collapse
Affiliation(s)
- Minal Garg
- Department of Biochemistry, University of Lucknow, Lucknow 226007, India
| | - Niharika Maurya
- Department of Biochemistry, University of Lucknow, Lucknow 226007, India
| |
Collapse
|
10
|
Loh CY, Chai JY, Tang TF, Wong WF, Sethi G, Shanmugam MK, Chong PP, Looi CY. The E-Cadherin and N-Cadherin Switch in Epithelial-to-Mesenchymal Transition: Signaling, Therapeutic Implications, and Challenges. Cells 2019; 8:E1118. [PMID: 31547193 PMCID: PMC6830116 DOI: 10.3390/cells8101118] [Citation(s) in RCA: 785] [Impact Index Per Article: 130.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/07/2019] [Revised: 09/16/2019] [Accepted: 09/19/2019] [Indexed: 12/17/2022] Open
Abstract
Epithelial-to-Mesenchymal Transition (EMT) has been shown to be crucial in tumorigenesis where the EMT program enhances metastasis, chemoresistance and tumor stemness. Due to its emerging role as a pivotal driver of tumorigenesis, targeting EMT is of great therapeutic interest in counteracting metastasis and chemoresistance in cancer patients. The hallmark of EMT is the upregulation of N-cadherin followed by the downregulation of E-cadherin, and this process is regulated by a complex network of signaling pathways and transcription factors. In this review, we summarized the recent understanding of the roles of E- and N-cadherins in cancer invasion and metastasis as well as the crosstalk with other signaling pathways involved in EMT. We also highlighted a few natural compounds with potential anti-EMT property and outlined the future directions in the development of novel intervention in human cancer treatments. We have reviewed 287 published papers related to this topic and identified some of the challenges faced in translating the discovery work from bench to bedside.
Collapse
Affiliation(s)
- Chin-Yap Loh
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya 47500, Malaysia.
| | - Jian Yi Chai
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya 47500, Malaysia.
| | - Ting Fang Tang
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia.
| | - Won Fen Wong
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
| | - Muthu Kumaraswamy Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
| | - Pei Pei Chong
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya 47500, Malaysia.
| | - Chung Yeng Looi
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya 47500, Malaysia.
| |
Collapse
|
11
|
Wang Y, Mo Y, Wang L, Su P, Xie Y. Let-7b contributes to hepatocellular cancer progression through Wnt/β-catenin signaling. Saudi J Biol Sci 2018; 25:953-958. [PMID: 30108446 PMCID: PMC6087813 DOI: 10.1016/j.sjbs.2018.03.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/19/2017] [Revised: 03/01/2018] [Accepted: 03/05/2018] [Indexed: 12/11/2022] Open
Abstract
Elevated evidences show that microRNAs (miRNAs) play vital roles in tumor progression regulation. However, the functional role of let-7b in hepatocellular carcinoma (HCC) is still largely unknown. In this study, we try to investigate the biological activity of let-7b in human HCC cells and try to find the potential regulatory signaling pathway. Our results indicate that let- 7b was remarkably down-regulated in human HCC tissues by qRT-PCR. In addition, let-7b overexpression decreased the expression of β-catenin and c-Myc, while upregulated E-cadherin expression in HCC cells which was verified by quantitative real-time PCR (qRT-PCR) and western blotting. Furthermore, Wnt/β-catenin was involved in let-7b biological activity which was revealed by luciferase assay. Moreover, Wnt/β-catenin signaling inhibitor blocks HCC cell proliferation which is as the same pattern as let-7b overexpression inhibits in HCC cells proliferation. In conclusion, down-regulated let-7b promotes HCC cell proliferation through Wnt/β-catenin signaling in HCC cells. These results suggested that appropriate manipulation of let-7b might be a new treatment of human HCC in the future.
Collapse
Affiliation(s)
- Yiwei Wang
- North China University of Science and Technology Affiliated Hospital, Tangshan 063000, PR China
| | - Yanbo Mo
- North China University of Science and Technology Affiliated Hospital, Tangshan 063000, PR China
| | - Lin Wang
- North China University of Science and Technology Affiliated Hospital, Tangshan 063000, PR China
| | - Peng Su
- North China University of Technology, Tangshan 063000, PR China
| | - Yuxi Xie
- North China University of Science and Technology Affiliated Hospital, Tangshan 063000, PR China
| |
Collapse
|
12
|
Yang Q, Wang Y, Pan X, Ye J, Gan S, Qu F, Chen L, Chu C, Gao Y, Cui X. Frizzled 8 promotes the cell proliferation and metastasis of renal cell carcinoma. Oncotarget 2017; 8:78989-79002. [PMID: 29108281 PMCID: PMC5668014 DOI: 10.18632/oncotarget.20742] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/31/2016] [Accepted: 08/08/2017] [Indexed: 12/23/2022] Open
Abstract
Recent reports have shown a rapid rise in the incidence of renal cell carcinoma (RCC), and Wnt (Wingless-related integration site) signaling pathway is important in RCC. Frizzled 8 (FZD8) is a member of Frizzled (FZD) receptor family which could activate canonical or non-canonical Wnt/β-catenin pathways. Nevertheless, the role of FZD8 in RCC is poorly investigated. The immunohistochemical analysis showed high expression of FZD8 in RCC tissues compared with peri-tumor tissues. FZD8 knockdown decreased the ability of proliferation and metastasis of RCC cells. Research revealed that the FZD8 regulated the transcription of Cyclin D1, c-Myc, and could promote the epithelial to mesenchymal transition (EMT) by mediating Vimentin and Snail through the Wnt/β-catenin signaling pathway. In addition, the results of our experiment revealed that FZD8 is involved in the regulation of non-canonical Wnt signaling pathway. These data suggested that the expression of FZD8 may play an important role in the proliferation and metastasis of RCC, and serve as a putative promising drug target for human RCC therapy.
Collapse
Affiliation(s)
- Qiwei Yang
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai 201805, People’s Republic of China
- Department of Urology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, People’s Republic of China
| | - Ye Wang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai 200082, People’s Republic of China
| | - Xiuwu Pan
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai 201805, People’s Republic of China
- Department of Urology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, People’s Republic of China
| | - Jianqing Ye
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai 201805, People’s Republic of China
| | - Sishun Gan
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai 201805, People’s Republic of China
| | - Fajun Qu
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai 201805, People’s Republic of China
| | - Lu Chen
- Department of Urology, Ruijin Hospital, Shanghai Jiaotong University, Shanghai 200025, People’s Republic of China
| | - Chuanmin Chu
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai 201805, People’s Republic of China
| | - Yi Gao
- Department of Urology, Ruijin Hospital, Shanghai Jiaotong University, Shanghai 200025, People’s Republic of China
| | - Xingang Cui
- Department of Urology, The Third Affiliated Hospital of Second Military Medical University (Eastern Hepatobiliary Surgery Hospital), Shanghai 201805, People’s Republic of China
| |
Collapse
|
13
|
Aberrant β-catenin expression in urothelial carcinomas in blackfoot disease-endemic areas. Kaohsiung J Med Sci 2017; 33:11-16. [PMID: 28088268 DOI: 10.1016/j.kjms.2016.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/11/2016] [Revised: 10/05/2016] [Accepted: 10/05/2016] [Indexed: 02/07/2023] Open
Abstract
Arsenic is a well-known toxic element and carcinogenic agent. The aim of this study was to investigate p63, E-cadherin, and β-catenin proteins in urothelial carcinoma (UC) in both arsenic contaminated areas [so-called blackfoot disease (BFD) area] and non-BFD areas. The expressions of p63, E-cadherin, and β-catenin proteins in 20 UC cases of blackfoot disease and 22 UC cases in non-BFD areas were detected using immunohistochemical methods. The results revealed a high p63 expression in 20 (47.6%) UC cases and high E-cadherin expression in six (14.3%) UC cases. Expressions of p63 and E-cadherin showed no significant correlations with clinicopathologic parameters. However, all 20 BFD cases and 12 of 22 (54.5%) non-BFD cases showed aberrant β-catenin expression. Ten out of 22 (45.5%) non-BFD cases also had normal membranous immunoreactivity. The β-catenin staining pattern significantly differed between cases in endemic and nonendemic areas of BFD (p=0.001). Tumor sites also significantly correlated with β-catenin expression (p=0.044). In addition, membranous localization of β-catenin was lower in UC from BFD-endemic areas compared with those from non-BFD endemic areas. In conclusion, it is suggested that relocalization of β-catenin from membrane to cytoplasm may be involved in the tumorigenesis of UC from BFD-endemic areas.
Collapse
|
14
|
Kovacs G, Billfeldt NK, Farkas N, Dergez T, Javorhazy A, Banyai D, Pusztai C, Szanto A. Cytoplasmic expression of β-catenin is an independent predictor of progression of conventional renal cell carcinoma: a simple immunostaining score. Histopathology 2016; 70:273-280. [DOI: 10.1111/his.13059] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/22/2016] [Accepted: 08/05/2016] [Indexed: 12/28/2022]
Affiliation(s)
- Gyula Kovacs
- Medical Faculty; Ruprecht-Karls-University; Heidelberg Germany
- Department of Urology; Medical School; University of Pecs; Pecs Hungary
| | | | - Nelli Farkas
- Institute of Bioanalysis; Medical School; University of Pecs; Pecs Hungary
| | - Timea Dergez
- Institute of Bioanalysis; Medical School; University of Pecs; Pecs Hungary
| | - Andras Javorhazy
- Department of Urology; Medical School; University of Pecs; Pecs Hungary
| | - Daniel Banyai
- Department of Urology; Medical School; University of Pecs; Pecs Hungary
| | - Csaba Pusztai
- Department of Urology; Medical School; University of Pecs; Pecs Hungary
| | - Arpad Szanto
- Department of Urology; Medical School; University of Pecs; Pecs Hungary
| |
Collapse
|
15
|
Taguchi YH, Iwadate M, Umeyama H. SFRP1 is a possible candidate for epigenetic therapy in non-small cell lung cancer. BMC Med Genomics 2016; 9 Suppl 1:28. [PMID: 27534621 PMCID: PMC4989892 DOI: 10.1186/s12920-016-0196-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/12/2022] Open
Abstract
Background Non-small cell lung cancer (NSCLC) remains a lethal disease despite many proposed treatments. Recent studies have indicated that epigenetic therapy, which targets epigenetic effects, might be a new therapeutic methodology for NSCLC. However, it is not clear which objects (e.g., genes) this treatment specifically targets. Secreted frizzled-related proteins (SFRPs) are promising candidates for epigenetic therapy in many cancers, but there have been no reports of SFRPs targeted by epigenetic therapy for NSCLC. Methods This study performed a meta-analysis of reprogrammed NSCLC cell lines instead of the direct examination of epigenetic therapy treatment to identify epigenetic therapy targets. In addition, mRNA expression/promoter methylation profiles were processed by recently proposed principal component analysis based unsupervised feature extraction and categorical regression analysis based feature extraction. Results The Wnt/β-catenin signalling pathway was extensively enriched among 32 genes identified by feature extraction. Among the genes identified, SFRP1 was specifically indicated to target β-catenin, and thus might be targeted by epigenetic therapy in NSCLC cell lines. A histone deacetylase inhibitor might reactivate SFRP1 based upon the re-analysis of a public domain data set. Numerical computation validated the binding of SFRP1 to WNT1 to suppress Wnt signalling pathway activation in NSCLC. Conclusions The meta-analysis of reprogrammed NSCLC cell lines identified SFRP1 as a promising target of epigenetic therapy for NSCLC. Electronic supplementary material The online version of this article (doi:10.1186/s12920-016-0196-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Y-H Taguchi
- Department of Physics, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, 112-8551, Tokyo, Japan.
| | - Mitsuo Iwadate
- Department of Biological Science, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, 112-8551, Tokyo, Japan
| | - Hideaki Umeyama
- Department of Biological Science, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, 112-8551, Tokyo, Japan
| |
Collapse
|
16
|
Skowron MA, Niegisch G, Fritz G, Arent T, van Roermund JGH, Romano A, Albers P, Schulz WA, Hoffmann MJ. Phenotype plasticity rather than repopulation from CD90/CK14+ cancer stem cells leads to cisplatin resistance of urothelial carcinoma cell lines. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:144. [PMID: 26606927 PMCID: PMC4660687 DOI: 10.1186/s13046-015-0259-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 07/10/2015] [Accepted: 11/14/2015] [Indexed: 02/12/2023]
Abstract
BACKGROUND Tumour heterogeneity and resistance to systemic treatment in urothelial carcinoma (UC) may arise from cancer stem cells (CSC). A recent model describes cellular differentiation states within UC based on corresponding expression of surface markers (CD) and cytokeratins (CK) with CD90 and CK14 positive cells representing the least differentiated and most tumourigenic population. Based on the fact that this population is postulated to constitute CSCs and the origin of cisplatin resistance, we enriched urothelial carcinoma cell lines (UCCs) for CD90 and studied the tumour-initiating potential of these separated cells in vitro. METHODS Magnetic- and fluorescence-activated- cell sorting were used for separation of CD90(+) and CD90(-) UCCs. Distribution of cell surface markers CD90, CD44, and CD49f and cytokeratins CK14, CK5, and CK20 as well as the effects of short- and long-term treatment with cisplatin were assessed in vitro and measured by qRT-PCR, immunocytochemistry, reporter assay and flow cytometry in 11 UCCs. RESULTS We observed cell populations with surface markers according to those reported in tumour xenografts. However, expression of cytokeratins did not concord regularly with that of the surface markers. In particular, expression of CD90 and CK14 diverged during enrichment of CD90(+) cells by immunomagnetic sorting or following cisplatin treatment. Enriched CD90(+) cells did not exhibit CSC-like characteristics like enhanced clonogenicity and cisplatin resistance. Moreover, selection of cisplatin-resistant sublines by long-term drug treatment did not result in enrichment of CD90(+) cells. Rather, these sublines displayed significant phenotypic plasticity expressing EMT markers, an altered pattern of CKs, and WNT-pathway target genes. CONCLUSIONS Our findings indicate that the correspondence between CD surface markers and cytokeratins reported in xenografts is not maintained in commonly used UCCs and that CD90 may not be a stable marker of CSC in UC. Moreover, UCCs cells are capable of substantial phenotypic plasticity that may significantly contribute to the emergence of cisplatin resistance.
Collapse
Affiliation(s)
- Margaretha A Skowron
- Department of Urology, Medical Faculty, Heinrich-Heine-University Duesseldorf, Universitaetsstrasse 1, 40225, Düsseldorf, Germany.
| | - Günter Niegisch
- Department of Urology, Medical Faculty, Heinrich-Heine-University Duesseldorf, Universitaetsstrasse 1, 40225, Düsseldorf, Germany.
| | - Gerhard Fritz
- Institute of Toxicology, Medical Faculty, Heinrich-Heine-University Duesseldorf, Universitaetsstrasse 1, 40225, Düsseldorf, Germany.
| | - Tanja Arent
- Department of Forensic Medicine, Medical Faculty, Heinrich-Heine-University Duesseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.
| | - Joep G H van Roermund
- Department of Urology, Maastricht University Medical Centre, P. Debyelaan 25, 6202, AZ, Maastricht, The Netherlands.
| | - Andrea Romano
- Department of Obstetrics and Gynaecology, GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, P. Debyelaan 25, 6202, AZ, Maastricht, The Netherlands.
| | - Peter Albers
- Department of Urology, Medical Faculty, Heinrich-Heine-University Duesseldorf, Universitaetsstrasse 1, 40225, Düsseldorf, Germany.
| | - Wolfgang A Schulz
- Department of Urology, Medical Faculty, Heinrich-Heine-University Duesseldorf, Universitaetsstrasse 1, 40225, Düsseldorf, Germany.
| | - Michèle J Hoffmann
- Department of Urology, Medical Faculty, Heinrich-Heine-University Duesseldorf, Universitaetsstrasse 1, 40225, Düsseldorf, Germany.
| |
Collapse
|
17
|
Rogler A, Kendziorra E, Giedl J, Stoehr C, Taubert H, Goebell PJ, Wullich B, Stöckle M, Lehmann J, Petsch S, Hartmann A, Stoehr R. Functional analyses and prognostic significance of SFRP1 expression in bladder cancer. J Cancer Res Clin Oncol 2015; 141:1779-90. [PMID: 25732201 DOI: 10.1007/s00432-015-1942-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/14/2015] [Accepted: 02/16/2015] [Indexed: 12/13/2022]
Abstract
PURPOSE We previously showed that the Wnt-signaling antagonist SFRP1 (secreted frizzled-related protein 1) is a promising marker in bladder cancer. The aim of this study was to validate the prognostic role and analyze the functional significance of SFRP1. METHODS Four bladder cancer cell lines (RT112, RT4, J82 and BFTC905) and one urothelial cell line (UROtsa) were used for functional characterization of SFRP1 expression. Effects on viability, proliferation and wound healing were investigated, and canonical Wnt-pathway activity as well as Wnt-signaling target gene expression was analyzed. Additionally, tissue micro-arrays from two different bladder tumor cohorts were evaluated for SFRP1 expression, and associations with survival and histopathological parameters were analyzed. RESULTS The cell lines RT112, RT4, J82 and UROtsa showed SFRP1 expression. In BFTC905, SFRP1 expression was inhibited by promoter hypermethylation. Wnt-pathway activity was absent in all cell lines and independent from SFRP1 expression. RT112 and BFTC905 were used for further functional characterization. SFRP1 overexpression resulted in decreased viability and migration in BFTC905 cells. Knockdown of SFRP1 expression in RT112 cells resulted only in marginal effects. In bladder tumors, SFRP1 expression was associated with lower tumor grade, but not with progression in patients with papillary bladder cancer. SFRP1 expressing papillary bladder cancer tumors also demonstrated a tendency to longer overall survival. CONCLUSIONS SFRP1 is reducing malignant potential of BFTC905 cells, but not by regulation of canonical Wnt-signaling pathway. Other pathways, like non-canonical Wnt or the MAPK pathway, could be activated via SFRP1-expression loss. In bladder tumors, SFRP1 has the potential to predict outcome for a subset of papillary bladder tumors.
Collapse
Affiliation(s)
- Anja Rogler
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Krankenhausstr. 8-10, 91054, Erlangen, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Evaluation of transforming growth factor-β1 suppress Pokemon/epithelial-mesenchymal transition expression in human bladder cancer cells. Tumour Biol 2014; 36:1155-62. [PMID: 25722217 DOI: 10.1007/s13277-014-2625-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/03/2014] [Accepted: 09/10/2014] [Indexed: 01/08/2023] Open
Abstract
Transforming growth factor-β1 (TGF-β1) plays a dual role in apoptosis and in proapoptotic responses in the support of survival in a variety of cells. The aim of this study was to determine the function of TGF-β1 in bladder cancer cells and the relationship with POK erythroid myeloid ontogenic factor (Pokemon). TGF-β1 and its receptors mediate several tumorigenic cascades that regulate cell proliferation, migration, and survival of bladder cancer cells. Bladder cancer cells T24 were treated with different levels of TGF-β1. Levels of Pokemon, E-cadherin, Snail, MMP2, MMP9, Twist, VEGF, and β-catenin messenger RNA (mRNA) and protein were examined by real-time quantitative fluorescent PCR and Western blot analysis, respectively. The effects of TGF-β1 on epithelial-mesenchymal transition of T24 cells were evaluated with wound-healing assay, proliferation of T24 was evaluated with reference to growth curves with MTT assay, and cell invasive ability was investigated by Transwell assay. Data show that Pokemon was inhibited by TGF-β1 treatment; the gene and protein of E-cadherin and β-catenin expression level showed decreased markedly after TGF-β1 treatment (P < 0.05). While the bladder cancer cell after TGF-β1 treatment showed a significantly reduced wound-closing efficiency at 6, 12, and 24 h, mechanistic analyses demonstrated that different levels of TGF-β1 promotes tumor cell growth, migration, and invasion in bladder cancer cells (P < 0.01, P < 0.05, respectively). In summary, our findings suggest that TGF-β1 may inhibit the expression of Pokemon, β-catenin, and E-cadherin. The high expression of TGF-β1 leads to an increase in the phenotype and apical-base polarity of epithelial cells. These changes of cells may result in the recurrence and progression of bladder cancer at last. Related mechanism is worthy of further investigation.
Collapse
|
19
|
Han L, Li Y, Tchao J, Kaplan AD, Lin B, Li Y, Mich-Basso J, Lis A, Hassan N, London B, Bett GCL, Tobita K, Rasmusson RL, Yang L. Study familial hypertrophic cardiomyopathy using patient-specific induced pluripotent stem cells. Cardiovasc Res 2014; 104:258-69. [PMID: 25209314 PMCID: PMC4217687 DOI: 10.1093/cvr/cvu205] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 12/23/2022] Open
Abstract
Aims Familial hypertrophic cardiomyopathy (HCM) is one the most common heart disorders, with gene mutations in the cardiac sarcomere. Studying HCM with patient-specific induced pluripotent stem-cell (iPSC)-derived cardiomyocytes (CMs) would benefit the understanding of HCM mechanism, as well as the development of personalized therapeutic strategies. Methods and results To investigate the molecular mechanism underlying the abnormal CM functions in HCM, we derived iPSCs from an HCM patient with a single missense mutation (Arginine442Glycine) in the MYH7 gene. CMs were next enriched from HCM and healthy iPSCs, followed with whole transcriptome sequencing and pathway enrichment analysis. A widespread increase of genes responsible for ‘Cell Proliferation’ was observed in HCM iPSC-CMs when compared with control iPSC-CMs. Additionally, HCM iPSC-CMs exhibited disorganized sarcomeres and electrophysiological irregularities. Furthermore, disease phenotypes of HCM iPSC-CMs were attenuated with pharmaceutical treatments. Conclusion Overall, this study explored the possible patient-specific and mutation-specific disease mechanism of HCM, and demonstrates the potential of using HCM iPSC-CMs for future development of therapeutic strategies. Additionally, the whole methodology established in this study could be utilized to study mechanisms of other human-inherited heart diseases.
Collapse
Affiliation(s)
- Lu Han
- Department of Developmental Biology, University of Pittsburgh School of Medicine, 8117 Rangos Research Center, 530 45th Street, Pittsburgh, PA 15201, USA
| | - Yang Li
- Department of Developmental Biology, University of Pittsburgh School of Medicine, 8117 Rangos Research Center, 530 45th Street, Pittsburgh, PA 15201, USA
| | - Jason Tchao
- Department of Developmental Biology, University of Pittsburgh School of Medicine, 8117 Rangos Research Center, 530 45th Street, Pittsburgh, PA 15201, USA
| | - Aaron D Kaplan
- Center for Cellular and Systems Electrophysiology, Department of Physiology and Biophysics, SUNY, Buffalo, NY 14214, USA
| | - Bo Lin
- Department of Developmental Biology, University of Pittsburgh School of Medicine, 8117 Rangos Research Center, 530 45th Street, Pittsburgh, PA 15201, USA
| | - You Li
- Department of Developmental Biology, University of Pittsburgh School of Medicine, 8117 Rangos Research Center, 530 45th Street, Pittsburgh, PA 15201, USA
| | - Jocelyn Mich-Basso
- Department of Developmental Biology, University of Pittsburgh School of Medicine, 8117 Rangos Research Center, 530 45th Street, Pittsburgh, PA 15201, USA
| | - Agnieszka Lis
- Center for Cellular and Systems Electrophysiology, Department of Physiology and Biophysics, SUNY, Buffalo, NY 14214, USA
| | - Narmeen Hassan
- Department of Developmental Biology, University of Pittsburgh School of Medicine, 8117 Rangos Research Center, 530 45th Street, Pittsburgh, PA 15201, USA
| | - Barry London
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Glenna C L Bett
- Department of Obstetrics and Gynecology, SUNY, Buffalo, NY 14214, USA
| | - Kimimasa Tobita
- Department of Developmental Biology, University of Pittsburgh School of Medicine, 8117 Rangos Research Center, 530 45th Street, Pittsburgh, PA 15201, USA
| | - Randall L Rasmusson
- Center for Cellular and Systems Electrophysiology, Department of Physiology and Biophysics, SUNY, Buffalo, NY 14214, USA
| | - Lei Yang
- Department of Developmental Biology, University of Pittsburgh School of Medicine, 8117 Rangos Research Center, 530 45th Street, Pittsburgh, PA 15201, USA
| |
Collapse
|
20
|
Pooja T, Karunagaran D. Emodin suppresses Wnt signaling in human colorectal cancer cells SW480 and SW620. Eur J Pharmacol 2014; 742:55-64. [PMID: 25205133 DOI: 10.1016/j.ejphar.2014.08.028] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/02/2014] [Revised: 08/16/2014] [Accepted: 08/21/2014] [Indexed: 12/30/2022]
Abstract
Wnt signaling is involved in the regulation of cell proliferation, differentiation and apoptosis. Its aberrant activation is a key event in the pathogenesis and progression of human colorectal cancers. Dietary phytochemicals are gaining importance as chemotherapeutic agents owing to their potential to prevent, delay or reverse oncogenesis. Here we demonstrate that emodin (1,3,8-trihydroxy-6-methylanthraquinone), an anthraquinone present in the roots and bark of several medicinal plants, down regulates Wnt signaling pathway in human colorectal cancer cells (SW480 and SW620) by down regulating TCF/LEF transcriptional activity. Emodin significantly down regulated the expression of key players of Wnt signaling (β-catenin and TCF7L2) and also that of its various downstream targets (cyclin D1, c-Myc, snail, vimentin, MMP-2 and MMP-9). Two novel targets of emodin׳s action were discovered namely Wnt co-activator p300 (down regulated) and repressor HBP1 (up regulated). Morphological changes induced by emodin suggest mesenchymal to epithelial transition accompanied by the increase in E-cadherin expression in human colorectal cancer cells but a differentiation marker (alkaline phosphatase) was activated only in SW620 cells (metastatic origin) and not in SW480 cells (primary tumor-derived). Moreover, our data indicate that reactive oxygen species plays a key role in emodin-mediated down regulation of Wnt signaling as emodin-mediated inhibition of migration and induction of growth arrest were partially rescued by the reactive oxygen species scavenger ascorbic acid. Effects of emodin shown in this study may provide important insights for the use of this anthraquinone as a potential complementary and integrated medicine for the treatment of human colorectal cancer.
Collapse
Affiliation(s)
- Thacker Pooja
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India.
| | - Devarajan Karunagaran
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, India.
| |
Collapse
|
21
|
Du W, Liu X, Fan G, Zhao X, Sun Y, Wang T, Zhao R, Wang G, Zhao C, Zhu Y, Ye F, Jin X, Zhang F, Zhong Z, Li X. From cell membrane to the nucleus: an emerging role of E-cadherin in gene transcriptional regulation. J Cell Mol Med 2014; 18:1712-9. [PMID: 25164084 PMCID: PMC4196647 DOI: 10.1111/jcmm.12340] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/23/2014] [Accepted: 05/05/2014] [Indexed: 01/06/2023] Open
Abstract
E-cadherin is a well-known mediator of cell–cell adherens junctions. However, many other functions of E-cadherin have been reported. Collectively, the available data suggest that E-cadherin may also act as a gene transcriptional regulator. Here, evidence supporting this claim is reviewed, and possible mechanisms of action are discussed. E-cadherin has been shown to modulate the activity of several notable cell signalling pathways, and given that most of these pathways in turn regulate gene expression, we proposed that E-cadherin may regulate gene transcription by affecting these pathways. Additionally, E-cadherin has been shown to accumulate in the nucleus where documentation of an E-cadherin fragment bound to DNA suggests that E-cadherin may directly regulate gene transcription. In summary, from the cell membrane to the nucleus, a role for E-cadherin in gene transcription may be emerging. Studies specifically focused on this potential role would allow for a more thorough understanding of this transmembrane glycoprotein in mediating intra- and intercellular activities.
Collapse
Affiliation(s)
- Wenjun Du
- Department of Digestion, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong Province, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Toyokawa G, Cho HS, Masuda K, Yamane Y, Yoshimatsu M, Hayami S, Takawa M, Iwai Y, Daigo Y, Tsuchiya E, Tsunoda T, Field HI, Kelly JD, Neal DE, Maehara Y, Ponder BAJ, Nakamura Y, Hamamoto R. Histone lysine methyltransferase Wolf-Hirschhorn syndrome candidate 1 is involved in human carcinogenesis through regulation of the Wnt pathway. Neoplasia 2011; 13:887-98. [PMID: 22028615 PMCID: PMC3201566 DOI: 10.1593/neo.11048] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/27/2011] [Revised: 09/01/2011] [Accepted: 09/06/2011] [Indexed: 11/18/2022]
Abstract
A number of histone methyltransferases have been identified and biochemically characterized, but the pathologic roles of their dysfunction in human diseases like cancer are not well understood. Here, we demonstrate that Wolf-Hirschhorn syndrome candidate 1 (WHSC1) plays important roles in human carcinogenesis. Transcriptional levels of this gene are significantly elevated in various types of cancer including bladder and lung cancers. Immunohistochemical analysis using a number of clinical tissues confirmed significant up-regulation of WHSC1 expression in bladder and lung cancer cells at the protein level. Treatment of cancer cell lines with small interfering RNA targeting WHSC1 significantly knocked down its expression and resulted in the suppression of proliferation. Cell cycle analysis by flow cytometry indicated that knockdown of WHSC1 decreased the cell population of cancer cells at the S phase while increasing that at the G(2)/M phase. WHSC1 interacts with some proteins related to the WNT pathway including β-catenin and transcriptionally regulates CCND1, the target gene of the β-catenin/Tcf-4 complex, through histone H3 at lysine 36 trimethylation. This is a novel mechanism for WNT pathway dysregulation in human carcinogenesis, mediated by the epigenetic regulation of histone H3. Because expression levels of WHSC1 are significantly low in most normal tissue types, it should be feasible to develop specific and selective inhibitors targeting the enzyme as antitumor agents that have a minimal risk of adverse reaction.
Collapse
Affiliation(s)
- Gouji Toyokawa
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Surgery and Science, Graduate School of Medical Science, Kyusyu University, Fukuoka, Japan
| | - Hyun-Soo Cho
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ken Masuda
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuka Yamane
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masanori Yoshimatsu
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Surgery and Science, Graduate School of Medical Science, Kyusyu University, Fukuoka, Japan
| | - Shinya Hayami
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masashi Takawa
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yukiko Iwai
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yataro Daigo
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Medical Oncology, Shiga University of Medical Science, Seta Tsukinowa-cho, Shiga, Japan
| | - Eiju Tsuchiya
- Department of Pathology, Saitama Cancer Center, Saitama, Japan
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Kanagawa, Japan
| | | | - Helen I Field
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - John D Kelly
- Department of Oncology, Cancer Research UK Cambridge Research Institute, University of Cambridge, Cambridge, UK
- Division of Surgery & Interventional Science, UCL Medical School, University College London, London, UK
| | - David E Neal
- Department of Oncology, Cancer Research UK Cambridge Research Institute, University of Cambridge, Cambridge, UK
| | - Yoshihiko Maehara
- Department of Surgery and Science, Graduate School of Medical Science, Kyusyu University, Fukuoka, Japan
| | - Bruce AJ Ponder
- Department of Oncology, Cancer Research UK Cambridge Research Institute, University of Cambridge, Cambridge, UK
| | - Yusuke Nakamura
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ryuji Hamamoto
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Oncology, Cancer Research UK Cambridge Research Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
23
|
Koch KR, Zhang CO, Kaczmarek P, Barchi J, Guo L, Shahjee HM, Keay S. The effect of a novel frizzled 8-related antiproliferative factor on in vitro carcinoma and melanoma cell proliferation and invasion. Invest New Drugs 2011; 30:1849-64. [PMID: 21931970 DOI: 10.1007/s10637-011-9746-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/13/2011] [Accepted: 08/29/2011] [Indexed: 01/07/2023]
Abstract
Antiproliferative factor (APF) is a potent frizzled protein 8-related sialoglycopeptide inhibitor of bladder epithelial cell proliferation that mediates its activity by binding to cytoskeletal associated protein 4 in the cell membrane. Synthetic asialylated APF (as-APF) (Galβ1-3GalNAcα-O-TVPAAVVVA) was previously shown to inhibit both normal bladder epithelial as well as T24 bladder carcinoma cell proliferation and heparin-binding epidermal growth factor-like growth factor (HB-EGF) production at low nanomolar concentrations, and an L: -pipecolic acid derivative (Galβ1-3GalNAcα-O-TV-pipecolic acid-AAVVVA) was also shown to inhibit normal bladder epithelial cell proliferation. To better determine their spectrum of activity, we measured the effects of these APF derivatives on the proliferation of cells derived from additional urologic carcinomas (bladder and kidney), non-urologic carcinomas (ovary, lung, colon, pancreas, and breast), and melanomas using a (3)H-thymidine incorporation assay. We also measured the effects of as-APF on cell HB-EGF and matrix metalloproteinase (MMP2) secretion plus cell invasion, using qRT-PCR, Western blot and an in vitro invasion assay. L: -pipecolic acid as-APF and/or as-APF significantly inhibited proliferation of each cell line in a dose-dependent manner with IC(50)'s in the nanomolar range, regardless of tissue origin, cell type (carcinoma vs. melanoma), or p53 or ras mutation status. as-APF also inhibited HB-EGF and MMP2 production plus in vitro invasion of tested bladder, kidney, breast, lung, and melanoma tumor cell lines, in a dose-dependent manner (IC(50) = 1-100 nM). Synthetic APF derivatives are potent inhibitors of urologic and non-urologic carcinoma plus melanoma cell proliferation, MMP2 production, and invasion, and may be useful for development as adjunctive antitumor therapy(ies).
Collapse
Affiliation(s)
- Kristopher R Koch
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | | | | | | | | | | | | |
Collapse
|
24
|
Georgopoulos NT, Kirkwood LA, Walker DC, Southgate J. Differential regulation of growth-promoting signalling pathways by E-cadherin. PLoS One 2010; 5:e13621. [PMID: 21049033 PMCID: PMC2964323 DOI: 10.1371/journal.pone.0013621] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/16/2010] [Accepted: 07/13/2010] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Despite the well-documented association between loss of E-cadherin and carcinogenesis, as well as the link between restoration of its expression and suppression of proliferation in carcinoma cells, the ability of E-cadherin to modulate growth-promoting cell signalling in normal epithelial cells is less well understood and frequently contradictory. The potential for E-cadherin to co-ordinate different proliferation-associated signalling pathways has yet to be fully explored. METHODOLOGY/PRINCIPAL FINDINGS Using a normal human urothelial (NHU) cell culture system and following a calcium-switch approach, we demonstrate that the stability of NHU cell-cell contacts differentially regulates the Epidermal Growth Factor Receptor (EGFR)/Extracellular Signal-Regulated Kinase (ERK) and Phosphatidylinositol 3-Kinase (PI3-K)/AKT pathways. We show that stable cell contacts down-modulate the EGFR/ERK pathway, whilst inducing PI3-K/AKT activity, which transiently enhances cell growth at low density. Functional inactivation of E-cadherin interferes with the capacity of NHU cells to form stable calcium-mediated contacts, attenuates E-cadherin-mediated PI3-K/AKT induction and enhances NHU cell proliferation by allowing de-repression of the EGFR/ERK pathway and constitutive activation of β-catenin-TCF signalling. CONCLUSIONS/SIGNIFICANCE Our findings provide evidence that E-cadherin can differentially and concurrently regulate specific growth-related signalling pathways in a context-specific fashion, with direct, functional consequences for cell proliferation and population growth. Our observations not only reveal a novel, complex role for E-cadherin in normal epithelial cell homeostasis and tissue regeneration, but also provide the basis for a more complete understanding of the consequences of E-cadherin loss on malignant transformation.
Collapse
Affiliation(s)
- Nikolaos T. Georgopoulos
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York, United Kingdom
| | - Lisa A. Kirkwood
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York, United Kingdom
| | - Dawn C. Walker
- Department of Computer Science, Kroto Research Institute, University of Sheffield, Sheffield, United Kingdom
| | - Jennifer Southgate
- Jack Birch Unit for Molecular Carcinogenesis, Department of Biology, University of York, York, United Kingdom
- * E-mail:
| |
Collapse
|
25
|
Shi TP, Xu H, Wei JF, Ai X, Ma X, Wang BJ, Ju ZH, Zhang GX, Wang C, Wu ZQ, Zhang X. Association of low expression of notch-1 and jagged-1 in human papillary bladder cancer and shorter survival. J Urol 2008; 180:361-6. [PMID: 18499162 DOI: 10.1016/j.juro.2008.02.037] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/11/2007] [Indexed: 11/27/2022]
Abstract
PURPOSE We investigated the expression of Notch receptors and ligands in normal bladder transitional epithelium and transitional cell carcinoma of the bladder. We also explored its clinical and pathological implications. MATERIALS AND METHODS The expression of Notch-1 to 3, Jagged-1 and Delta-like-1 was detected respectively in 70 cases of bladder carcinoma, 10 of normal urothelium and the 2 cell lines T24 and BIU-87 using immunohistochemistry. Reverse transcriptase-polymerase chain reaction and Western blot were used to assay the expression level of Notch-1 and Jagged-1. The predictive value of this expression for prognosis was investigated by Kaplan-Meier curves and Cox proportional hazards analysis in a multivariate model. RESULTS All 5 kinds of Notch factors were intensively stained in normal bladder transitional epithelium immunohistochemically but expression was significantly decreased in tumor tissues. Moreover, expression of the 5 genes in papillary tumors was lower than in invasive tumors but only Notch-1 and Jagged-1 showed a statistically significant difference. Postoperative disease-free survival time in patients with low Notch-1 plus Jagged-1 expression was significantly shorter than that in patients with other expression patterns in papillary tumors (p = 0.014). Multivariate Cox proportional hazards model analysis identified Jagged-1 expression as an independent prognostic factor for disease-free survival (RR 3.09, p = 0.011). CONCLUSIONS The Notch family expression pattern in papillary bladder transitional cell carcinoma is different from that in invasive bladder transitional cell carcinoma. Low expression of Notch-1 as well as Jagged-1 is potentially a useful marker for survival in patients with papillary bladder transitional cell carcinoma.
Collapse
Affiliation(s)
- Tao-ping Shi
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Gonzalgo ML, Datar RH, Schoenberg MP, Cote RJ. The role of deoxyribonucleic acid methylation in development, diagnosis, and prognosis of bladder cancer. Urol Oncol 2007; 25:228-35. [PMID: 17483020 DOI: 10.1016/j.urolonc.2006.07.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/06/2006] [Revised: 06/20/2006] [Accepted: 07/14/2006] [Indexed: 12/28/2022]
Abstract
Alterations in global levels and regional patterns of deoxyribonucleic acid methylation are among the earliest and most common events known to occur in human cancer. The mutational and epigenetic effects of this covalent deoxyribonucleic acid modification to the development of bladder cancer are well recognized. The contribution of aberrant methylation to mutational hot spots located within genes, transcriptional silencing, and chromosomal instability is reviewed in the context of its relevance to bladder carcinogenesis. Understanding how such processes evolve during the progression of bladder cancer is essential for using these molecular changes in the clinical setting. The recent development of sensitive and specific techniques for quantifying methylation changes in urine specimens and bodily fluids underscores the potential use of this molecular marker for early detection and surveillance of bladder cancer. Further refinement of these molecular biological techniques holds much promise for the use of methylation markers for bladder cancer diagnosis, risk stratification, and disease prognostication.
Collapse
Affiliation(s)
- Mark L Gonzalgo
- Department of Urology, The James Buchanan Brady Urological Institute, The Johns Hopkins Medical Institutions, Baltimore, MD 21287, USA
| | | | | | | |
Collapse
|
27
|
Lim SK, Hoffmann FM. Smad4 cooperates with lymphoid enhancer-binding factor 1/T cell-specific factor to increase c-myc expression in the absence of TGF-beta signaling. Proc Natl Acad Sci U S A 2006; 103:18580-5. [PMID: 17132729 PMCID: PMC1693705 DOI: 10.1073/pnas.0604773103] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/11/2023] Open
Abstract
The c-myc protooncogene is a key regulator of cell proliferation whose expression is reduced in normal epithelial cells in response to the growth inhibitory cytokine TGF-beta. Smad4 mediates this inhibitory effect of TGF-beta by forming a complex with Smad3, E2F4/5, and p107 at the TGF-beta inhibitory element (TIE) element on the c-myc promoter. In contrast, cell proliferation and c-myc expression are increased in response to Wnt ligands; this effect is mediated through the lymphoid enhancer-binding factor 1/T cell-specific factor (LEF/TCF) family of transcription factors on the c-myc promoter LEF/TCF-binding elements (TBE1 and TBE2). We report that a peptide aptamer designed to inhibit the binding between Smad4 and LEF/TCF reduced c-myc expression and the growth rate of HepG2 cells. Further analysis demonstrated that, in the absence of TGF-beta, Smad4 was bound to the positive regulatory element TBE1 from the c-myc promoter and activated c-myc promoter activity. Smad4 binding to the positive TBE1 c-myc element was reduced by TGF-beta, consistent with Smad4's inhibitory role on c-myc expression in response to TGF-beta. Reduction of Smad4 levels by RNAi knockdown also reduced c-myc expression levels and sensitized hepatocytes to cell death by serum deprivation. Two tumor-derived mutant Smad4 proteins that fail to mediate TGF-beta responses were still competent to cooperate with LEF1 to activate the c-myc promoter. These results support a previously unreported TGF-beta-independent function for Smad4 in cooperating with LEF/TCF to activate c-myc expression.
Collapse
Affiliation(s)
| | - F. Michael Hoffmann
- *McArdle Laboratory for Cancer Research and
- Laboratory of Genetics, University of Wisconsin, Madison, WI 53706
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
28
|
Abstract
Urothelial carcinoma (UC), the common histological subtype of bladder cancer, presents as a papillary tumor or as an invasive, often lethal form. To study UC molecular biology, candidate gene and genome-wide approaches have been followed. Here, it is argued that a 'cancer pathway' perspective is useful to integrate findings from both approaches. According to this view, papillary cancers typically exhibit activation of the MAPK pathway, as a consequence of oncogenic mutations in FGFR3 or HRAS, with increased Cyclin D1 expression. In contrast, invasive UC are characterized by severe disturbances in proximate cell cycle regulators, e.g. RB1 and CDKN2A/p16(INK4A), which decrease dependency on mitogenic signaling. In addition, these disturbances permit, promote and are in turn exacerbated by chromosomal instability, which is further enhanced by loss of TP53 function. In another vicious cycle, defective cell cycle regulation interacts with DNA methylation alterations. The transition toward invasive UC may require concomitant and interacting defects in cell cycle regulation and the control of genomic stability. Intriguingly, neither canonical WNT/beta-Catenin nor hedgehog signaling appear to play major roles in UC. This may reflect its origin from more differentiated urothelial cells possessing a high regenerative potential rather than a stem cell population.
Collapse
Affiliation(s)
- Wolfgang A Schulz
- Department of Urology, Heinrich-Heine-University Düsseldorf, Germany.
| |
Collapse
|
29
|
Moyano Calvo JL, Blanco Palenciano E, Beato Moreno A, Gutiérrez González M, Pérez-Lanzac Lorca A, Samaniego Torres A, Montaño JA, Fernández Castiñeiras J. Cadherina E, Catenina Beta, Antígeno Ki-67 y proteína p53 en el pronóstico de la recidiva tumoral en los tumores superficiales de vejiga T1. Actas Urol Esp 2006; 30:871-8. [PMID: 17175927 DOI: 10.1016/s0210-4806(06)73553-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The natural history of the superficial carcinoma of bladder is characterized by his high rate of recurrence and by the aptitude to progress to higher stages. We are going to investigate the capacity of prediction for tumor recurrence of protein p53, antigen Ki-67, E Cadherin and Beta Catenin MATERIAL AND METHOD 88 T1 tumors with a median of free time of disease of 36 months. 58% of the serie has received prophylactic treatment with BCG 81 mg. weekly for six weeks. Cut-oof level for For P53 and Ki-67 is 10 % of stained cells. For E Cadherin and Beta Catenin we have established two groups: one with the values 0-4 (negative), and other one with the values 5-8 (positive). RESULTS Recurrence rate 31%, stage progression 3%. Ki-67 expression is correlated with grade (p .002) and lymphatic permeation (p .028). Multiplicity is correlated with lack( of Cadherin and Catenin's expression. Only Ki-67 expression (p .049) and lack of Beta Catenin expression (p .039) reach statistical significance. In multivariant study only lack of Beta Catenin's expression shows independent recurrence value (p .049; O.R: 2,4-6,9) CONCLUSIONS The most useful prognmostic markers are Ki-67 and Catenina Beta Only Beta Catenin Beta shows independent value for tumour recurrence. Tumors wich lack expression for Catenin B or Cadherin E have lower recurrence free time.
Collapse
|
30
|
Chan VYW, Chan MWY, Leung WK, Leung PS, Sung JJY, Chan FKL. Intestinal trefoil factor promotes invasion in non-tumorigenic Rat-2 fibroblast cell. ACTA ACUST UNITED AC 2005; 127:87-94. [PMID: 15680474 DOI: 10.1016/j.regpep.2004.10.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/11/2004] [Revised: 10/22/2004] [Accepted: 10/27/2004] [Indexed: 12/11/2022]
Abstract
Intestinal trefoil factor (TFF3) is essential in regulating cell migration and maintaining mucosal integrity in gastrointestinal tract. We previously showed that TFF3 was overexpressed in gastric carcinoma. Whether TFF3 possesses malignant potential is not fully elucidated. We sought to investigate the effects of inducting TFF3 expression in a non-malignant rat fibroblast cell line (Rat-2) on the cell proliferation, invasion and the genes regulating cell invasion. Invasiveness and proliferation of transfected Rat-2 cell line were assessed using in vitro invasion chamber assay and colorimetric MTS assay. Differential mRNA expressions of invasion-related genes, namely, metalloproteinases (MMP-9), tissue inhibitors of metalloproteinases (TIMP-1), beta-catenin and E-cadherin, were determined by quantitative real-time polymerase chain reaction (PCR). We showed that TFF3 did not inhibit the proliferation of Rat-2 cells. We also demonstrated that transfection of TFF3 significantly promoted invasion of Rat-2 cells by 1.4- to 2.2-folds. There was an upregulation of beta-catenin (13.1-23.0%) and MMP-9 (43.4-92.2%) mRNA expression levels, and downregulation of E-cadherin (25.6-33.8%) and TIMP-1 (31.5-37.8%) in TFF3-transfected cells compared to controls during 48-h incubation. Our results suggested that TFF3 possesses malignant potential through promotion of cell invasiveness and alteration of invasion-related genes.
Collapse
Affiliation(s)
- Victor Y W Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong
| | | | | | | | | | | |
Collapse
|
31
|
Rieger-Christ KM, Ng L, Hanley RS, Durrani O, Ma H, Yee AS, Libertino JA, Summerhayes IC. Restoration of plakoglobin expression in bladder carcinoma cell lines suppresses cell migration and tumorigenic potential. Br J Cancer 2005; 92:2153-9. [PMID: 15942628 PMCID: PMC2361803 DOI: 10.1038/sj.bjc.6602651] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/15/2023] Open
Abstract
The reduction or loss of plakoglobin expression in late-stage bladder cancer has been correlated with poor survival where upregulation of this catenin member by histone deacetylase inhibitors has been shown to accompany tumour suppression in an in vivo model. In this study, we directly addressed the question of the role of plakoglobin in bladder tumorigenesis following restoration, or knockdown of expression in bladder carcinoma cell lines. Restoration of plakoglobin expression resulted in a reduction in migration and suppression of tumorigenic potential in vivo. Immunocytochemistry revealed cytoplasmic and membranous localisation of plakoglobin in transfectants with <1% of cells displaying detectable nuclear localisation of plakoglobin. siRNA knockdown experiments targeting plakoglobin, revealed enhanced migration in all cell lines in the presence and absence of E-cadherin expression. In bladder cell lines expressing low levels of plakoglobin and desmoglein-2, elevated levels of desmoglein-2 were detected following restoration of plakoglobin expression in transfected cell lines. Analysis of wnt signalling revealed no activation event associated with plakoglobin expression in the bladder model. These results show that plakoglobin acts as a tumour suppressor gene in bladder carcinoma cells and the silencing of plakoglobin gene expression in late-stage bladder cancer is a primary event in tumour progression.
Collapse
Affiliation(s)
- K M Rieger-Christ
- Cell and Molecular Biology Laboratory, Robert E Wise MD Research and Education Institute, 31 Mall Road, Burlington, MA 01805, USA
- Department of Urology, Lahey Clinic, 41 Mall Road, Burlington, MA 01805, USA
| | - L Ng
- Department of Urology, Lahey Clinic, 41 Mall Road, Burlington, MA 01805, USA
| | - R S Hanley
- Department of Urology, Lahey Clinic, 41 Mall Road, Burlington, MA 01805, USA
| | - O Durrani
- Department of Urology, Lahey Clinic, 41 Mall Road, Burlington, MA 01805, USA
| | - H Ma
- Tufts University School of Medicine, 136 Harrison Avenue, Boston, USA
| | - A S Yee
- Tufts University School of Medicine, 136 Harrison Avenue, Boston, USA
| | - J A Libertino
- Department of Urology, Lahey Clinic, 41 Mall Road, Burlington, MA 01805, USA
| | - I C Summerhayes
- Cell and Molecular Biology Laboratory, Robert E Wise MD Research and Education Institute, 31 Mall Road, Burlington, MA 01805, USA
- Department of Urology, Lahey Clinic, 41 Mall Road, Burlington, MA 01805, USA
- Cell and Molecular Biology Laboratory, Robert E Wise MD Research and Education Institute, 31 Mall Road, Burlington, MA 01805, USA. E-mail:
| |
Collapse
|
32
|
Cote RJ, Laird PW, Datar RH. Promoter hypermethylation: a new therapeutic target emerges in urothelial cancer. J Clin Oncol 2005; 23:2879-81. [PMID: 15753457 DOI: 10.1200/jco.2005.11.923] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/31/2022] Open
|
33
|
Thievessen I, Wolter M, Prior A, Seifert HH, Schulz WA. Hedgehog signaling in normal urothelial cells and in urothelial carcinoma cell lines. J Cell Physiol 2005; 203:372-7. [PMID: 15521068 DOI: 10.1002/jcp.20248] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/21/2022]
Abstract
Constitutive activation of hedgehog signaling, often caused by PTCH1 inactivation and leading to inappropriate activation of GLI target genes, is crucial for the development of several human tumors including basal cell carcinoma of the skin and medulloblastoma. The PTCH1 gene at 9q22 is also considered as a candidate tumor suppressor in transitional cell carcinoma (TCC), of which >50% show LOH in this region. However, only rare mutations have been found in PTCH1. We have therefore investigated GLI-dependent promoter activity and expression of hedgehog pathway components in TCC cell lines and proliferating normal urothelial cells. Normal urothelial cells cultured in serum-free medium, but not TCC lines exhibited low, but significant promoter activity under standard growth conditions. Accordingly, GLI1-3 and PTCH1 mRNAs were expressed at moderate levels, and sonic hedgehog (SHH) mRNA expression was low to undetectable. In co-transfection experiments GLI1 increased promoter activity significantly in one TCC line and further in normal urothelial cells, but less strongly in other TCC lines. Expression patterns of GLI factor mRNAs did not correlate with inducibility. No significant effects of SHH or cyclopamine on proliferation were observed, ruling out autocrine effects. However, SHH induced GLI-dependent promoter activity in normal urothelial cells. Taken together, our data suggest that the hedgehog pathway is weakly active in normal adult urothelial cells and of limited importance in TCC.
Collapse
MESH Headings
- Carcinoma, Transitional Cell/metabolism
- Carcinoma, Transitional Cell/physiopathology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cell Transformation, Neoplastic/metabolism
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/physiology
- Genes, Tumor Suppressor
- Hedgehog Proteins
- Humans
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Patched Receptors
- Patched-1 Receptor
- Promoter Regions, Genetic/genetics
- RNA, Messenger/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Trans-Activators/pharmacology
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Urinary Bladder Neoplasms/metabolism
- Urinary Bladder Neoplasms/physiopathology
- Urothelium/drug effects
- Urothelium/metabolism
- Urothelium/physiopathology
- Veratrum Alkaloids/pharmacology
- Zinc Finger Protein GLI1
Collapse
Affiliation(s)
- Ingo Thievessen
- Urologische Klinik, Heinrich-Heine-Universität, Düsseldorf, Germany
| | | | | | | | | |
Collapse
|
34
|
Keay SK, Szekely Z, Conrads TP, Veenstra TD, Barchi JJ, Zhang CO, Koch KR, Michejda CJ. An antiproliferative factor from interstitial cystitis patients is a frizzled 8 protein-related sialoglycopeptide. Proc Natl Acad Sci U S A 2004; 101:11803-8. [PMID: 15282374 PMCID: PMC511055 DOI: 10.1073/pnas.0404509101] [Citation(s) in RCA: 154] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/20/2023] Open
Abstract
Approximately 1 million people in the United States suffer from interstitial cystitis, a chronic painful urinary bladder disorder characterized by thinning or ulceration of the bladder epithelial lining; its etiology is unknown. We have identified a glycosylated frizzled-related peptide inhibitor of cell proliferation that is secreted specifically by bladder epithelial cells from patients with this disorder. This antiproliferative factor (APF) profoundly inhibits bladder cell proliferation by means of regulation of cell adhesion protein and growth factor production. The structure of APF was deduced by using ion trap mass spectrometry (MS), enzymatic digestion, lectin affinity chromatography, and total synthesis, and confirmed by coelution of native and synthetic APF derivatives on microcapillary reversed-phase liquid chromatography (microRPLC)/MS. APF was determined to be an acidic, heat-stable sialoglycopeptide whose peptide chain has 100% homology to the putative sixth transmembrane domain of frizzled 8. Both synthetic and native APF had identical biological activity in normal bladder epithelial cells and T24 bladder cancer cells. Northern blot analysis indicated binding of a probe containing the sequence for the frizzled 8 segment with mRNA extracted from cells of patients with interstitial cystitis but not controls. APF is therefore a frizzled-related peptide growth inhibitor shown to contain exclusively a transmembrane segment of a frizzled protein and is a potential biomarker for interstitial cystitis.
Collapse
Affiliation(s)
- Susan K Keay
- Division of Infectious Diseases, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | | | | | | | | | | | | | | |
Collapse
|