1
|
Parsons MJ, Tammela T, Dow LE. WNT as a Driver and Dependency in Cancer. Cancer Discov 2021; 11:2413-2429. [PMID: 34518209 DOI: 10.1158/2159-8290.cd-21-0190] [Citation(s) in RCA: 118] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/30/2021] [Accepted: 06/11/2021] [Indexed: 12/15/2022]
Abstract
The WNT signaling pathway is a critical regulator of development and adult tissue homeostasis and becomes dysregulated in many cancer types. Although hyperactivation of WNT signaling is common, the type and frequency of genetic WNT pathway alterations can vary dramatically between different cancers, highlighting possible cancer-specific mechanisms for WNT-driven disease. In this review, we discuss how WNT pathway disruption contributes to tumorigenesis in different organs and how WNT affects the tumor cell and immune microenvironment. Finally, we describe recent and ongoing efforts to target oncogenic WNT signaling as a therapeutic strategy. SIGNIFICANCE: WNT signaling is a fundamental regulator of tissue homeostasis and oncogenic driver in many cancer types. In this review, we highlight recent advances in our understanding of WNT signaling in cancer, particularly the complexities of WNT activation in distinct cancer types, its role in immune evasion, and the challenge of targeting the WNT pathway as a therapeutic strategy.
Collapse
Affiliation(s)
- Marie J Parsons
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York
| | - Tuomas Tammela
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lukas E Dow
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York. .,Department of Medicine, Weill Cornell Medicine, New York, New York
| |
Collapse
|
2
|
Johari B, Rezaeejam H, Moradi M, Taghipour Z, Saltanatpour Z, Mortazavi Y, Nasehi L. Increasing the colon cancer cells sensitivity toward radiation therapy via application of Oct4-Sox2 complex decoy oligodeoxynucleotides. Mol Biol Rep 2020; 47:6793-6805. [PMID: 32865703 DOI: 10.1007/s11033-020-05737-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/24/2020] [Indexed: 02/07/2023]
Abstract
Low sensitivity of cancer stem cells toward regular cancer therapy strategies is an important issue in the field of cancer remedy. The concept of cancer stem cell elimination has been a topic of interest in the field of molecular medicine for a long time. At the current study, it was aimed to elevate the sensitivity of cancer stem-like cells toward radiotherapy by treating with Oct4-Sox2 complex decoy oligodeoxynucleotides (ODNs). After treating HT29 and HT29-ShE cells with Oct4-Sox2 complex decoy ODNs, and analyzing the cellular uptake and localization of decoys, treated cells and control groups were subjected to irradiation by fractionated 6MV X-ray with a final dose of 2 Gy. Thereafter, the influence of radiotherapy on ODNs treated groups and control group was investigated on cell viability, cell cycle, apoptosis, colonosphere formation and scratch assay. Cellular uptake and localization assays demonstrated that decoy ODNs can efficiently be transfected to the cells and reside in subcellular compartment, where they pose their action on gene regulation. Post radiotherapy analysis indicated statistical significance in decoy ODNs treated cells by means of lower cell viability, cell cycle arrest in G2/M phase, increased cellular apoptosis, and reduced cell motility. Also, formed colonospheres were smaller in size and fewer in numbers. Considering the role of Oct4, and Sox2 transcription factors in signaling pathways of preserving stemness and inducing reverse EMT, application of decoy strategy could increase the sensitivity of cancer cells toward irradiation, which has a potential to eliminate the cancerous cells from tumors and support cancer treatment.
Collapse
Affiliation(s)
- Behrooz Johari
- Student Research Committee, Zanjan University of Medical Sciences, Zanjan, Iran.,Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.,Zanjan Pharmaceutical Biotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hamed Rezaeejam
- Department of Radiation Oncology, Vali-e-Asr Hospital, Zanjan University of Medical Sciences, Zanjan, Iran.,Department of Radiology, School of Paramedical Sciences, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohammad Moradi
- Department of Biotechnology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Zahraa Taghipour
- Student Research Committee, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Zohreh Saltanatpour
- Stem Cell and Regenerative Medicine Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Yousef Mortazavi
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.,Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Leila Nasehi
- Student Research Committee, Zanjan University of Medical Sciences, Zanjan, Iran. .,Department of Medical Laboratory, School of Paramedical Sciences, Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
3
|
Pádua D, Figueira P, Ribeiro I, Almeida R, Mesquita P. The Relevance of Transcription Factors in Gastric and Colorectal Cancer Stem Cells Identification and Eradication. Front Cell Dev Biol 2020; 8:442. [PMID: 32626705 PMCID: PMC7314965 DOI: 10.3389/fcell.2020.00442] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/11/2020] [Indexed: 12/12/2022] Open
Abstract
Gastric and colorectal cancers have a high incidence and mortality worldwide. The presence of cancer stem cells (CSCs) within the tumor mass has been indicated as the main reason for tumor relapse, metastasis and therapy resistance, leading to poor overall survival. Thus, the elimination of CSCs became a crucial goal for cancer treatment. The identification of these cells has been performed by using cell-surface markers, a reliable approach, however it lacks specificity and usually differs among tumor type and in some cases even within the same type. In theory, the ideal CSC markers are those that are required to maintain their stemness features. The knowledge that CSCs exhibit characteristics comparable to normal stem cells that could be associated with the expression of similar transcription factors (TFs) including SOX2, OCT4, NANOG, KLF4 and c-Myc, and signaling pathways such as the Wnt/β-catenin, Hedgehog (Hh), Notch and PI3K/AKT/mTOR directed the attention to the use of these similarities to identify and target CSCs in different tumor types. Several studies have demonstrated that the abnormal expression of some TFs and the dysregulation of signaling pathways are associated with tumorigenesis and CSC phenotype. The disclosure of common and appropriate biomarkers for CSCs will provide an incredible tool for cancer prognosis and treatment. Therefore, this review aims to gather the new insights in gastric and colorectal CSC identification specially by using TFs as biomarkers and divulge promising drugs that have been found and tested for targeting these cells.
Collapse
Affiliation(s)
- Diana Pádua
- i3S – Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Paula Figueira
- i3S – Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Inês Ribeiro
- i3S – Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Raquel Almeida
- i3S – Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
- Department of Biology, Faculty of Sciences, University of Porto, Porto, Portugal
| | - Patrícia Mesquita
- i3S – Institute for Research and Innovation in Health, University of Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| |
Collapse
|
4
|
CDCP1 enhances Wnt signaling in colorectal cancer promoting nuclear localization of β-catenin and E-cadherin. Oncogene 2019; 39:219-233. [DOI: 10.1038/s41388-019-0983-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 04/02/2019] [Accepted: 04/04/2019] [Indexed: 11/08/2022]
|
5
|
He JH, Han ZP, Zou MX, He ML, Li YG, Zheng L. CDX2/mir-145-5p/SENP1 Pathways Affect LNCaP Cells Invasion and Migration. Front Oncol 2019; 9:477. [PMID: 31249806 PMCID: PMC6582308 DOI: 10.3389/fonc.2019.00477] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 05/20/2019] [Indexed: 01/01/2023] Open
Abstract
Background/Aims: Recently, rapidly accumulating evidence has shown that microRNAs (miRNAs) are involved in human tumorigenesis, and the dysregulation of miRNAs has been observed in many cancers, including prostate cancer. miR-145-5p, an miRNA with reduced expression in prostate cancer cells, has been shown to have a tumor suppressive role in a variety of tumors. However, its underlying mechanism requires further elucidation. Methods: A lentiviral expression vector for miR-145-5p was constructed and used to establish a stable cell line (LNCaP) expressing miR-145-5p. The cells were cultured normally and divided into the control group (control), negative control group (negative control), and test group (miR-145-5p). Inhibition of proliferation was measured by a WST-8 assay. The early apoptosis rate of cells was detected by flow cytometry. Clone formation ability was detected by a clone formation inhibition test. Cell invasion and migration capacity was detected by a Transwell assay. The relative expression levels of proteins were detected by western blotting. We constructed a nude mouse model of prostate cancer to observe the effect of miR-145-5p on the growth of transplanted tumors. TargetScan bioinformatics software was used to predict target genes regulated by miR-14-5p. ChIPBase was used to predict transcription factors with binding sites in the upstream promoter region of miR-145-5p. Quantitative reverse transcription PCR was used to detect the relative expression level of genes. A bifluorescence-reporter gene vector was constructed to confirm the regulation of target genes by miR-145-5p. We used 5′ rapid amplification of cDNA ends to confirm the transcription start site of miR-145-5p.Chromatin immunoprecipitation technology was used to detect the effect of transcription factors binding to miR-145-5p. Results: The overexpression of miR-145-5p not only inhibited the proliferation, invasion, and migration of LNCaP cells but also promoted their early apoptosis. After overexpressing miR-145-5p, the expression of small ubiquitin-like modifier protein-specific protease 1 (SENP1), and caudal-related homeobox 2 (CDX2) protein was decreased in LNCaP cells. The transcription factor CDX2 bound to the miR-145-5p promoter region and inhibited its transcription. The transcription start site of miR-145-5p was located at a guanine residue 1,408 bp upstream of the stem-loop sequence. Upon overexpression, miR-145-5p could bind to the 3′-untranslated region of SENP1 to inhibit its translation. Conclusion: These results suggested that CDX2 inhibits the expression of miR-145-5p, thereby relieving the inhibitory effect of miR-145-5p on the translation of SENP1 and affecting the invasion and migration of prostate cancer cells.
Collapse
Affiliation(s)
- Jin-Hua He
- Department of Laboratory Medicine, Central Hospital of Panyu District, Guangzhou, China
| | - Ze-Ping Han
- Department of Laboratory Medicine, Central Hospital of Panyu District, Guangzhou, China
| | - Mao-Xian Zou
- Department of Laboratory Medicine, Central Hospital of Panyu District, Guangzhou, China
| | - Meng-Ling He
- Department of Laboratory Medicine, Central Hospital of Panyu District, Guangzhou, China
| | - Yu-Guang Li
- Department of Laboratory Medicine, Central Hospital of Panyu District, Guangzhou, China
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
6
|
Javan B, Atyabi F, Shahbazi M. Hypoxia-inducible bidirectional shRNA expression vector delivery using PEI/chitosan-TBA copolymers for colorectal Cancer gene therapy. Life Sci 2018; 202:140-151. [PMID: 29656061 DOI: 10.1016/j.lfs.2018.04.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 04/07/2018] [Accepted: 04/11/2018] [Indexed: 10/17/2022]
Abstract
AIMS This investigation was conducted to construct a hypoxia/colorectal dual-specific bidirectional short hairpin RNA (shRNA) expression vector and to transfect it into the colon cancer cell line HT-29 with PEI/chitosan-TBA nanoparticles for the simultaneous knock down of β-catenin and Bcl-2 under hypoxia. MAIN METHODS To construct a pRNA-bipHRE-CEA vector, the carcinoma embryonic antigen (CEA) promoter designed in two directions and the vascular endothelial growth factor (VEGF) enhancer were inserted between two promoters for hypoxic cancer specific gene expression. To confirm the therapeutic effect of the dual-specific vector, β-catenin and Bcl-2 shRNAs were inserted downstream of each promoter. The physicochemical properties, the cytotoxicity, and the transfection efficiency of these PEI/chitosan-TBA nanoparticles were investigated. In addition, the antitumor effects of the designed vector on the expression of β-catenin and Bcl-2, cell cycle distribution, and apoptosis were investigated in vitro. KEY FINDINGS The silencing effect of the hypoxia-response shRNA expression vector was relatively low (18%-25%) under normoxia, whereas it was significantly increased to approximately 50%-60% in the HT-29 cell line. Moreover, the cancer cells showed significant G0/G1 arrest and increased apoptosis due to gene silencing under hypoxia. Furthermore, MTS assay, fluorescence microscopy images, and flow cytometry analyses confirmed that the PEI/chitosan-TBA blend system provided effective transfection with low cytotoxicity. SIGNIFICANCE This novel hypoxia-responsive shRNA expression vector may be useful for RNA interference (RNAi)-based cancer gene therapy in hypoxic colorectal tumors. Moreover, the PEI/chitosan-TBA copolymer might be a promising gene carrier for use in gene transfer in vivo.
Collapse
Affiliation(s)
- Bita Javan
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran; Medical Cellular & Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran.
| | - Fatemeh Atyabi
- Department of Pharmaceutics, Faculty of Pharmacy, Nanotechnology Research Centre, Tehran University of Medical Sciences, Tehran, Iran.
| | - Majid Shahbazi
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran; Medical Cellular & Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran; Arya Tina Gene (ATG), Biopharmaceutical Company, Gorgan, Iran.
| |
Collapse
|
7
|
Lungova V, Verheyden JM, Sun X, Thibeault SL. β-Catenin signaling is essential for mammalian larynx recanalization and the establishment of vocal fold progenitor cells. Development 2018; 145:dev.157677. [PMID: 29386246 DOI: 10.1242/dev.157677] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 01/18/2018] [Indexed: 02/01/2023]
Abstract
Congenital laryngeal webs result from failure of vocal fold separation during development in utero Infants present with life-threatening respiratory problems at birth, and extensive lifelong difficulties in breathing and voicing. The molecular mechanisms that instruct vocal fold formation are rarely studied. Here, we show, for the first time, that conditional inactivation of the gene encoding β-catenin in the primitive laryngopharyngeal epithelium leads to failure in separation of the vocal folds, which approximates the gross phenotype of laryngeal webbing. These defects can be traced to a series of morphogenesis defects, including delayed fusion of the epithelial lamina and formation of the laryngeal cecum, failed separation of the larynx and esophagus with reduced and disorganized cartilages and muscles. Parallel to these morphogenesis defects, inactivation of β-catenin disrupts stratification of epithelial cells and establishment of p63+ basal progenitors. These findings provide the first line of evidence that links β-catenin function to the cell proliferation and progenitor establishment during larynx and vocal fold development.
Collapse
Affiliation(s)
- Vlasta Lungova
- Department of Surgery, University of Wisconsin-Madison, 5107 WIMR, 1111 Highland Avenue, Madison, WI 53705, USA
| | - Jamie M Verheyden
- Laboratory of Genetics, Biotechnology Center, University of Wisconsin-Madison, 425G Henry Mall, Madison, WI 53706, USA
| | - Xin Sun
- Laboratory of Genetics, Biotechnology Center, University of Wisconsin-Madison, 425G Henry Mall, Madison, WI 53706, USA
| | - Susan L Thibeault
- Department of Surgery, University of Wisconsin-Madison, 5107 WIMR, 1111 Highland Avenue, Madison, WI 53705, USA
| |
Collapse
|
8
|
Tate SC, Burke TF, Hartman D, Kulanthaivel P, Beckmann RP, Cronier DM. Optimising the combination dosing strategy of abemaciclib and vemurafenib in BRAF-mutated melanoma xenograft tumours. Br J Cancer 2016; 114:669-79. [PMID: 26978007 PMCID: PMC4800303 DOI: 10.1038/bjc.2016.40] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 01/15/2016] [Accepted: 01/28/2016] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Resistance to BRAF inhibition is a major cause of treatment failure for BRAF-mutated metastatic melanoma patients. Abemaciclib, a cyclin-dependent kinase 4 and 6 inhibitor, overcomes this resistance in xenograft tumours and offers a promising drug combination. The present work aims to characterise the quantitative pharmacology of the abemaciclib/vemurafenib combination using a semimechanistic pharmacokinetic/pharmacodynamic modelling approach and to identify an optimum dosing regimen for potential clinical evaluation. METHODS A PK/biomarker model was developed to connect abemaciclib/vemurafenib concentrations to changes in MAPK and cell cycle pathway biomarkers in A375 BRAF-mutated melanoma xenografts. Resultant tumour growth inhibition was described by relating (i) MAPK pathway inhibition to apoptosis, (ii) mitotic cell density to tumour growth and, under resistant conditions, (iii) retinoblastoma protein inhibition to cell survival. RESULTS The model successfully described vemurafenib/abemaciclib-mediated changes in MAPK pathway and cell cycle biomarkers. Initial tumour shrinkage by vemurafenib, acquisition of resistance and subsequent abemaciclib-mediated efficacy were successfully captured and externally validated. Model simulations illustrate the benefit of intermittent vemurafenib therapy over continuous treatment, and indicate that continuous abemaciclib in combination with intermittent vemurafenib offers the potential for considerable tumour regression. CONCLUSIONS The quantitative pharmacology of the abemaciclib/vemurafenib combination was successfully characterised and an optimised, clinically-relevant dosing strategy was identified.
Collapse
Affiliation(s)
- Sonya C Tate
- Global PK/PD, Eli Lilly and Company, Erl Wood Manor, Windlesham, Surrey GU20 6PH, UK
| | - Teresa F Burke
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | - Daisy Hartman
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | | | - Richard P Beckmann
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | - Damien M Cronier
- Global PK/PD, Eli Lilly and Company, Erl Wood Manor, Windlesham, Surrey GU20 6PH, UK
| |
Collapse
|
9
|
Jiang R, Niu X, Huang Y, Wang X. β-Catenin is important for cancer stem cell generation and tumorigenic activity in nasopharyngeal carcinoma. Acta Biochim Biophys Sin (Shanghai) 2016; 48:229-37. [PMID: 26849897 DOI: 10.1093/abbs/gmv134] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 11/28/2015] [Indexed: 12/22/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is one of the most common malignant tumors with poor prognosis and recurrence in South China. The hard eradication of NPC in clinic is predominantly due to cancer stem cells (CSCs). Increasing evidence revealed that the aberrant activation of Wnt/β-catenin was positively correlated with the produce of CSCs. To further investigate the effect of β-catenin on CSCs and tumorigenesis in NPC, a CNE2 cell line (pLKO.1-sh-β-catenin-CNE2) with stably suppressed expression of β-catenin was used in this study. The expressions of biomarkers in CSCs including c-myc, Nanog, Oct3/4, Sox2, EpCAM as well as adhesion-related proteins like E-cadherin and vimentin were analyzed by western blot analysis and immunofluorescent staining. The proliferation and migration abilities were investigated by MTT assay and Transwell assay, respectively. Cell cycle was analyzed by flow cytometry. Finally, xenograft was performed to determine the effect of β-catenin on oncogenesis in vivo. Results showed that the expressions of c-myc, Nanog, Oct3/4, Sox2, and EpCAM were all decreased in pLKO.1-sh-β-catenin-CNE2 cells. It was also found that vimentin was downregulated, while E-cadherin was upregulated. Results of MTT and Transwell assays suggested that the proliferation and migration abilities were impaired by silencing of β-catenin, and more cells were arrested in G1 phase when compared with the control. In vivo study indicated that the tumor growth was markedly suppressed in experimental group. Based on current findings, β-catenin may function as an essential protein for the maintenance of migration and proliferation abilities of NPC cells, and a complicated network consisting of c-myc, Nanog, Oct3/4, Sox2, EpCAM, E-cadherin, vimentin, and β-catenin may be involved in the inherent regulation mechanisms.
Collapse
Affiliation(s)
- Rui Jiang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 2000031, China
| | - Xiaoshuang Niu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 2000031, China
| | - Yuxiang Huang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 2000031, China
| | - Xiaosheng Wang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai 2000031, China
| |
Collapse
|
10
|
TCF4 silencing sensitizes the colon cancer cell line to oxaliplatin as a common chemotherapeutic drug. Anticancer Drugs 2014; 25:908-16. [DOI: 10.1097/cad.0000000000000118] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
11
|
Tate SC, Cai S, Ajamie RT, Burke T, Beckmann RP, Chan EM, De Dios A, Wishart GN, Gelbert LM, Cronier DM. Semi-mechanistic pharmacokinetic/pharmacodynamic modeling of the antitumor activity of LY2835219, a new cyclin-dependent kinase 4/6 inhibitor, in mice bearing human tumor xenografts. Clin Cancer Res 2014; 20:3763-74. [PMID: 24850847 DOI: 10.1158/1078-0432.ccr-13-2846] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Selective inhibition of cyclin-dependent kinases 4 and 6 (CDK4/6) represents a promising therapeutic strategy. However, despite documented evidence of clinical activity, limited information is available on the optimal dosing strategy of CDK4/6 inhibitors. Here, we present an integrated semi-mechanistic pharmacokinetic/pharmacodynamic model to characterize the quantitative pharmacology of LY2835219, a CDK4/6 inhibitor, in xenograft tumors. EXPERIMENTAL DESIGN LY2835219 plasma concentrations were connected to CDK4/6 inhibition and cell-cycle arrest in colo-205 human colorectal xenografts by incorporating the biomarkers, phospho-(ser780)-Rb, topoisomerase II α, and phosphohistone H3, into a precursor-dependent transit compartment model. This biomarker model was then connected to tumor growth inhibition (TGI) by: (i) relating the rate of tumor growth to mitotic cell density, and (ii) incorporating a concentration-dependent mixed cytostatic/cytotoxic effect driving quiescence and cell death at high doses. Model validation was evaluated by predicting LY2835219-mediated antitumor effect in A375 human melanoma xenografts. RESULTS The model successfully described LY2835219-mediated CDK4/6 inhibition, cell-cycle arrest, and TGI in colo-205, and was validated in A375. The model also demonstrated that a chronic dosing strategy achieving minimum steady-state trough plasma concentrations of 200 ng/mL is required to maintain durable cell-cycle arrest. Quiescence and cell death can be induced by further increasing LY2835219 plasma concentrations. CONCLUSIONS Our model provides mechanistic insight into the quantitative pharmacology of LY2835219 and supports the therapeutic dose and chronic dosing strategy currently adopted in clinical studies.
Collapse
Affiliation(s)
- Sonya C Tate
- Authors' Affiliations: Eli Lilly and Company, Windlesham, United Kingdom; and
| | - Shufen Cai
- Eli Lilly and Company, Indianapolis, Indiana
| | | | | | | | | | | | - Graham N Wishart
- Authors' Affiliations: Eli Lilly and Company, Windlesham, United Kingdom; and
| | | | - Damien M Cronier
- Authors' Affiliations: Eli Lilly and Company, Windlesham, United Kingdom; and
| |
Collapse
|
12
|
Seo JH, Jeong ES, Choi YK. Therapeutic effects of lentivirus-mediated shRNA targeting of cyclin D1 in human gastric cancer. BMC Cancer 2014; 14:175. [PMID: 24618206 PMCID: PMC3975285 DOI: 10.1186/1471-2407-14-175] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Accepted: 03/03/2014] [Indexed: 12/11/2022] Open
Abstract
Background Gastric cancer is the second most common cause of cancer-related death in males and the fourth in females. Traditional treatment has poor prognosis because of recurrence and systemic side effects. Therefore, the development of new therapeutic strategies is an important issue. Lentivirus-mediated shRNA stably inhibits target genes and can efficiently transduce most cells. Since overexpressed cyclin D1 is closely related to human gastric cancer progression, inhibition of cyclin D1 using specific targeting could be an effective treatment method of human gastric cancer. Methods The therapeutic effect of lentivirus-mediated shRNA targeting of cyclin D1 (ShCCND1) was analyzed both in vitro and in vivo experiments. Results In vitro, NCI-N87 cells with downregulation of cyclin D1 by ShCCND1 showed significant inhibition of cell proliferation, cell motility, and clonogenicity. Downregulation of cyclin D1 in NCI-N87 cells also resulted in significantly increased G1 arrest and apoptosis. In vivo, stable NCI-N87 cells expressing ShCCND1 were engrafted into nude mice. Then, the cancer-growth inhibition effect of lentivirus was confirmed. To assess lentivirus including ShCCND1 as a therapeutic agent, intratumoral injection was conducted. Tumor growth of the lentivirus-treated group was significantly inhibited compared to growth of the control group. These results are in accordance with the in vitro data and lend support to the mitotic figure count and apoptosis analysis of the tumor mass. Conclusion The lentivirus-mediated ShCCND1 was constructed, which effectively inhibited growth of NCI-N87-derived cancer both in vitro and in vivo. The efficiency of shRNA knockdown and variation in the degree of inhibition is mediated by different shRNA sequences and cancer cell lines. These experimental results suggest the possibility of developing new gastric cancer therapies using lentivirus-mediated shRNA.
Collapse
Affiliation(s)
| | | | - Yang-Kyu Choi
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Neungdong-ro, Gwangjin-gu, Seoul 143-701, Republic of Korea.
| |
Collapse
|
13
|
Rosenbluh J, Wang X, Hahn WC. Genomic insights into WNT/β-catenin signaling. Trends Pharmacol Sci 2013; 35:103-9. [PMID: 24365576 DOI: 10.1016/j.tips.2013.11.007] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 11/21/2013] [Accepted: 11/26/2013] [Indexed: 01/23/2023]
Abstract
The canonical WNT pathway regulates the stability of the proto-oncogene β-catenin and is aberrantly activated in many cancer types. Studies in a wide range of experimental models confirm that β-catenin activity is required for tumor initiation in cancers where this pathway is deregulated. However, to date this pathway has proven to be challenging to target therapeutically. Moreover, several lines of evidence suggest that other components and regulators of β-catenin exist. Here we will describe recent structural and functional studies describing genomic alterations and new regulators of β-catenin that lead to aberrant activation of the WNT/β-catenin pathway. These findings provide new insights into the biology of WNT/β-catenin signaling and suggest potential therapeutic opportunities.
Collapse
Affiliation(s)
- Joseph Rosenbluh
- Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, MA 02142 USA; Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215 USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115 USA
| | - Xiaoxing Wang
- Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, MA 02142 USA; Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215 USA
| | - William C Hahn
- Broad Institute of Harvard and MIT, 7 Cambridge Center, Cambridge, MA 02142 USA; Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215 USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 75 Francis Street, Boston, MA 02115 USA.
| |
Collapse
|
14
|
Seo JH, Jeong ES, Lee KS, Heo SH, Jeong DG, Choi YK. Lentivirus-mediated shRNA targeting of cyclin D1 enhances the chemosensitivity of human gastric cancer to 5-fluorouracil. Int J Oncol 2013; 43:2007-14. [PMID: 24100731 DOI: 10.3892/ijo.2013.2119] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 08/05/2013] [Indexed: 11/06/2022] Open
Abstract
Gastric cancer is one of the major public health problems. Despite new chemotherapeutic treatments, the prognosis of gastric cancer remains poor. 5-Fluorouracil (5-FU) is used as a standard chemotherapy drug in gastric cancer. However, 5-FU resistance develops frequently and is a main cause of chemotherapy failure in human gastric cancer. Overexpression of cyclin D1 is related to rapid cell growth, a poor prognosis and increased chemoresistance in several types of cancers. In this study, we investigated whether treatment of gastric cancer cells with shRNA targeting cyclin D1 (ShCCND1) or 5-FU, alone or in combination, influences the activation of phosphorylated AKT (pAKT) and pNFκB, which are markers that are increased in 5-FU chemoresistance. We also investigated the effect of combined treatment with ShCCND1 and 5-FU on cell growth and chemosensitivity to 5-FU in the gastric cancer cell line AGS. The data showed that ShCCND1-mediated cyclin D1 downregulation in AGS cells significantly inhibited cell proliferation, cell mobility and clonogenicity. In addition, combined treatment with ShCCND1 and 5-FU significantly decreased the survival rate of AGS cells, compared to single-treatment with either agent. These results demonstrated that ShCCND1 increases 5-FU chemosensitivity, a conclusion that is also supported by the concomitant reduction in expression of pAKT and pNFκB, increase of G1 arrest and induction of apoptosis. Taken together, these data provide further evidence that therapeutic strategies targeting cyclin D1 may have the dual advantage of suppressing the growth of cancer cells, while enhancing their chemosensitivity.
Collapse
Affiliation(s)
- Jin-Hee Seo
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 143-701, Republic of Korea
| | | | | | | | | | | |
Collapse
|
15
|
γ-Catenin at adherens junctions: mechanism and biologic implications in hepatocellular cancer after β-catenin knockdown. Neoplasia 2013; 15:421-34. [PMID: 23555187 DOI: 10.1593/neo.122098] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 01/29/2013] [Accepted: 01/30/2013] [Indexed: 12/22/2022] Open
Abstract
β-Catenin is important in liver homeostasis as a part of Wnt signaling and adherens junctions (AJs), while its aberrant activation is observed in hepatocellular carcinoma (HCC). We have reported hepatocyte-specific β-catenin knockout (KO) mice to lack adhesive defects as γ-catenin compensated at AJ. Because γ-catenin is a desmosomal protein, we asked if its increase in KO might deregulate desmosomes. No changes in desmosomal proteins or ultrastructure other than increased plakophilin-3 were observed. To further elucidate the role and regulation of γ-catenin, we contemplate an in vitro model and show γ-catenin increase in HCC cells upon β-catenin knockdown (KD). Here, γ-catenin is unable to rescue β-catenin/T cell factor (TCF) reporter activity; however, it sufficiently compensates at AJs as assessed by scratch wound assay, centrifugal assay for cell adhesion (CAFCA), and hanging drop assays. γ-Catenin increase is observed only after β-catenin protein decrease and not after blockade of its transactivation. γ-Catenin increase is associated with enhanced serine/threonine phosphorylation and abrogated by protein kinase A (PKA) inhibition. In fact, several PKA-binding sites were detected in γ-catenin by in silico analysis. Intriguingly γ-catenin KD led to increased β-catenin levels and transactivation. Thus, γ-catenin compensates for β-catenin loss at AJ without affecting desmosomes but is unable to fulfill functions in Wnt signaling. γ-Catenin stabilization after β-catenin loss is brought about by PKA. Catenin-sensing mechanism may depend on absolute β-catenin levels and not its activity. Anti-β-catenin therapies for HCC affecting total β-catenin may target aberrant Wnt signaling without negatively impacting intercellular adhesion, provided mechanisms leading to γ-catenin stabilization are spared.
Collapse
|
16
|
Li M, Tan SY, Zhang J, You HX. Effects of paeonol on intracellular calcium concentration and expression of RUNX3 in LoVo human colon cancer cells. Mol Med Rep 2013; 7:1425-30. [PMID: 23504038 DOI: 10.3892/mmr.2013.1372] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2012] [Accepted: 03/12/2013] [Indexed: 11/06/2022] Open
Abstract
Paeonol, a major phenolic component of the root bark of Paeonia moutan, is known to exhibit antitumor effects. However, the underlying mechanisms remain unknown. In the present study, the effects of paeonol on cell viability, intracellular calcium concentration and the expression of runt‑related transcription factor 3 (RUNX3) were analyzed in LoVo human colon cancer cells. Results revealed that paeonol markedly reduced LoVo cell viability in a time‑ and dose‑dependent manner. Flow cytometry assays demonstrated that paeonol blocked the cell cycle at the G1 to S transition and significantly induced apoptosis in LoVo cells. Intracellular calcium accumulation occurred following a 48 h treatment with paeonol. Furthermore, RUNX3 gene expression was increased in paeonol‑treated cells. These observations indicate that paeonol possesses antiproliferative properties and apoptosis‑inducing activity. One of the antitumor mechanisms of paeonol may be its apoptosis‑inducing activity through an increased intracellular calcium concentration and the upregulation of RUNX3 expression. Paeonol may be a promising antitumor agent for colon carcinoma treatment.
Collapse
Affiliation(s)
- Ming Li
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, PR China
| | | | | | | |
Collapse
|
17
|
Xie J, Xiang DB, Wang H, Zhao C, Chen J, Xiong F, Li TY, Wang XL. Inhibition of Tcf-4 induces apoptosis and enhances chemosensitivity of colon cancer cells. PLoS One 2012; 7:e45617. [PMID: 23029137 PMCID: PMC3454396 DOI: 10.1371/journal.pone.0045617] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 08/23/2012] [Indexed: 12/30/2022] Open
Abstract
Aberrant activation of β-catenin/Tcf-4 signaling has been implicated in human carcinogenesis, including colorectal cancer. In this study, we compared the effects of Tcf-4 knockdown with β-catenin knockdown on cell proliferation, apoptosis, and chemosensitivity in SW480 and HCT116 colon cancer cells using adenoviral vector-mediated short hairpin RNA (shRNA). Our results show that, compared to β-catenin knockdown, Tcf-4 knockdown more effectively inhibited colony formation, induced apoptosis, and increased 5-FU and oxaliplatin-mediated cytotoxicity in colon cancer cells. We further investigated the mechanisms involved in the different efficacies observed with β-catenin and Tcf-4 knockdown in colon cancer cells. FOXO4 is a member of the subfamily of mammalian FOXO forkhead transcription factors and plays a major role in controlling cellular proliferation, apoptosis, and DNA repair. Our data showed that the protein level of FOXO4 did not change after treatment with both β-catenin and Tcf-4 shRNA. However, β-catenin shRNA was found to increase the accumulation of phosphorylated FOXO4 S193 and decrease the expression of FOXO target genes p27Kip1 and MnSOD, whereas Tcf-4 shRNA showed the opposite effect. Therefore, compared to β-catenin knockdown, Tcf-4 knockdown shows better efficacy for inhibiting proliferation and inducing apoptosis of colorectal cancer cells, which may be related to increased FOXO4 transcriptional activity. These results suggest that Tcf-4 is an attractive potential therapeutic target for colorectal cancer therapy.
Collapse
Affiliation(s)
- Jiang Xie
- Children’s Hospital, Chongqing Medical University, Chongqing, China
- The Third People’s Hospital of Chengdu, Sichuan, China
- * E-mail: (JX); (TYL)
| | - De-Bing Xiang
- Cancer Center, Jiangjin Central Hospital, Chongqing, China
| | - Hong Wang
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Cong Zhao
- The Third People’s Hospital of Chengdu, Sichuan, China
| | - Jie Chen
- Children’s Hospital, Chongqing Medical University, Chongqing, China
| | - Feng Xiong
- Children’s Hospital, Chongqing Medical University, Chongqing, China
| | - Ting-Yu Li
- Children’s Hospital, Chongqing Medical University, Chongqing, China
- * E-mail: (JX); (TYL)
| | - Xiao-Lei Wang
- The Third People’s Hospital of Chengdu, Sichuan, China
| |
Collapse
|
18
|
Huang WS, Wang TB, He Y, Chen YJ, Zhong SL, Tan M. Phosphoinositide-3-kinase, catalytic, alpha polypeptide RNA interference inhibits growth of colon cancer cell SW948. World J Gastroenterol 2012; 18:3458-64. [PMID: 22807617 PMCID: PMC3396200 DOI: 10.3748/wjg.v18.i26.3458] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Revised: 05/09/2012] [Accepted: 05/13/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the gene knock-down effect by the phosphoinositide-3-kinase, catalytic, alpha polypeptide (PIK3CA)-targeted double-stranded RNA (dsRNA) and its effect on cell proliferation and cycle distribution in SW948.
METHODS: Two PIK3CA-targeted dsRNAs were constructed and transfected into SW948 cells. Transfections were performed using lipofectamineTM 2000. The transfection effectiveness was calculated basing on the rate of fluorescence cell of SW948 at 6 h after transfection. Total messenger RNA was extracted from these cells using the RNeasy kit, and semiquantitative reverse transcription polymerase chain reaction was performed to detect the down-regulation of PIK3CA, AKT1, MYC, and CCND1 gene expression. Cells were harvested, proteins were resolved, and western blot was employed to detect the expression levels of PIK3CA, AKT1, MYC, and CCND1 gene. Cell proliferation was assessed by 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide assay and the inhibition rate was calculated. Soft agar colony formation assay was performed basing on colonies greater than 60 μm in diameter at ×100 magnification. The effect on cell cycle distribution and apoptosis was assessed by flow cytometry. All experiments were performed in triplicate.
RESULTS: Green fluorescence was observed in SW948 cell transfected with plasmid Pgenesil-1, and the transfection effectiveness was about 65%. Forty-eight hours post-transfection, mRNA expression of PIK3CA in SW948 cells was 0.51 ± 0.04 vs 0.49 ± 0.03 vs 0.92 ± 0.01 vs 0.93 ± 0.03 (P = 0.001 ) in Pgenesil-CA1, Pgenesil-CA2, negative and blank group respectively. mRNA expression of AKT1 was 0.50 ± 0.03 vs 0.48 ± 0.01 vs 0.93 ± 0.04 vs 0.92 ± 0.02 (P = 0.000) in Pgenesil-CA1, Pgenesil-CA2, negative and blank group respectively. mRNA expression of MYC was 0.49 ± 0.01 vs 0.50 ± 0.04 vs 0.90 ± 0.02 vs 0.91 ± 0.03 (P = 0.001) in the four groups respectively. mRNA expression of CCND1 was 0.45 ± 0.02 vs 0.51 ± 0.01 vs 0.96 ± 0.03 vs 0.98 ± 0.01 (P = 0.001) in the four groups respectively. The protein level of PIK3CA was 0.53 ± 0.01 vs 0.54 ± 0.02 vs 0.92 ± 0.03 vs 0.91 ± 0.02 (P = 0.001) in Pgenesil-CA1, Pgenesil-CA2, negative and blank group respectively. The protein level of AKT1 in the four groups was 0.49 ± 0.02 vs 0.55 ± 0.03 vs 0.94 ± 0.03 vs 0.95 ± 0.04, P = 0.000). The protein level of MYC in the four groups was 0.51 ± 0.03 vs 0.52 ± 0.04 vs 0.92 ± 0.02 vs 0.95 ± 0.01 (P = 0.000). The protein level of CCND1 in the four groups was 0.54 ± 0.04 vs 0.56 ± 0.03 vs 0.93 ± 0.01 vs 0.93 ± 0.03 (P = 0.000). Both Pgenesil-CA1 and Pgenesil-CA2 plasmids significantly suppressed the growth of SW948 cells when compared with the negative or blank group at 48 h after transfection (29% vs 25% vs 17% vs 14%, P = 0.001), 60 h after transfection (38% vs 34% vs 19% vs 16%, P = 0.001), and 72 h after transfection (53% vs 48% vs 20% vs 17%, P = 0.000). Numbers of colonies in negative, blank, CA1, and CA2 groups were 42 ± 4, 45 ± 5, 8 ± 2, and 10 ± 3, respectively (P = 0.000). There were more than 4.5 times colonies in the blank and negative control groups as there were in the CA1 and CA2 groups. In addition, the colonies in blank and negative control groups were also larger than those in the CA1 and CA2 groups. The percentage of cells in the CA1 and CA2 groups was significantly higher in G0/G1 phase, but lower in S and G2/M phase when compared with the negative and control groups. Moreover, cell apoptosis rates in the CA1 and CA2 groups were 5.11 ± 0.32 and 4.73 ± 0.32, which were significantly higher than those in negative (0.95 ± 0.11, P = 0.000) and blank groups (0.86 ± 0.13, P = 0.001). No significant difference was found between CA1 and CA2 groups in cell cycle distribution and apoptosis.
CONCLUSION: PIK3CA-targeted short hairpin RNAs can block the phosphoinositide 3-kinase-Akt signaling pathway and inhibit cell growth, increase apoptosis, and induce cell cycle arrest in the PIK3CA-mutant colon cancer SW948 cells.
Collapse
|
19
|
Lin YY, Hsu YH, Huang HY, Shann YJ, Huang CYF, Wei SC, Chen CL, Jou TS. Aberrant nuclear localization of EBP50 promotes colorectal carcinogenesis in xenotransplanted mice by modulating TCF-1 and β-catenin interactions. J Clin Invest 2012; 122:1881-94. [PMID: 22466651 DOI: 10.1172/jci45661] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Dysregulation of canonical Wnt signaling is thought to play a role in colon carcinogenesis. β-Catenin, a key mediator of the pathway, is stabilized upon Wnt activation and accumulates in the nucleus, where it can interact with the transcription factor T cell factor (TCF) to transactivate gene expression. Normal colonic epithelia express a truncated TCF-1 form, called dnTCF-1, that lacks the critical β-catenin-binding domain and behaves as a transcriptional suppressor. How the cell maintains a balance between the two forms of TCF-1 is unclear. Here, we show that ERM-binding phosphoprotein 50 (EBP50) modulates the interaction between β-catenin and TCF-1. We observed EBP50 localization to the nucleus of human colorectal carcinoma cell lines at low cell culture densities and human primary colorectal tumors that manifested a poor clinical outcome. In contrast, EBP50 was primarily membranous in confluent cell lines. Aberrantly located EBP50 stabilized conventional β-catenin/TCF-1 complexes and connected β-catenin to dnTCF-1 to form a ternary molecular complex that enhanced Wnt/β-catenin signaling events, including the transcription of downstream oncogenes such as c-Myc and cyclin D1. Genome-wide analysis of the EBP50 occupancy pattern revealed consensus binding motifs bearing similarity to Wnt-responsive element. Conventional chromatin immunoprecipitation assays confirmed that EBP50 bound to genomic regions highly enriched with TCF/LEF binding motifs. Knockdown of EBP50 in human colorectal carcinoma cell lines compromised cell cycle progression, anchorage-independent growth, and tumorigenesis in nude mice. We therefore suggest that nuclear EBP50 facilitates colon tumorigenesis by modulating the interaction between β-catenin and TCF-1.
Collapse
Affiliation(s)
- Yu-Yu Lin
- Graduate Institute of Molecular Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Yao H, Ashihara E, Strovel JW, Nakagawa Y, Kuroda J, Nagao R, Tanaka R, Yokota A, Takeuchi M, Hayashi Y, Shimazaki C, Taniwaki M, Strand K, Padia J, Hirai H, Kimura S, Maekawa T. AV-65, a novel Wnt/β-catenin signal inhibitor, successfully suppresses progression of multiple myeloma in a mouse model. Blood Cancer J 2011; 1:e43. [PMID: 22829079 PMCID: PMC3256754 DOI: 10.1038/bcj.2011.41] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Revised: 07/16/2011] [Accepted: 09/09/2011] [Indexed: 01/14/2023] Open
Abstract
Multiple myeloma (MM) is a malignant neoplasm of plasma cells. Although new molecular targeting agents against MM have been developed based on the better understanding of the underlying pathogenesis, MM still remains an incurable disease. We previously demonstrated that β-catenin, a downstream effector in the Wnt pathway, is a potential target in MM using RNA interference in an in vivo experimental mouse model. In this study, we have screened a library of more than 100 000 small-molecule chemical compounds for novel Wnt/β-catenin signaling inhibitors using a high-throughput transcriptional screening technology. We identified AV-65, which diminished β-catenin protein levels and T-cell factor transcriptional activity. AV-65 then decreased c-myc, cyclin D1 and survivin expression, resulting in the inhibition of MM cell proliferation through the apoptotic pathway. AV-65 treatment prolonged the survival of MM-bearing mice. These findings indicate that this compound represents a novel and attractive therapeutic agent against MM. This study also illustrates the potential of high-throughput transcriptional screening to identify candidates for anticancer drug discovery.
Collapse
|
21
|
Sack U, Walther W, Scudiero D, Selby M, Aumann J, Lemos C, Fichtner I, Schlag PM, Shoemaker RH, Stein U. S100A4-induced cell motility and metastasis is restricted by the Wnt/β-catenin pathway inhibitor calcimycin in colon cancer cells. Mol Biol Cell 2011; 22:3344-54. [PMID: 21795396 PMCID: PMC3172260 DOI: 10.1091/mbc.e10-09-0739] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Calcimycin restricts Wnt/β-catenin–regulated S100A4 expression, leading to reduced S100A4-mediated cell migration and invasion in colon cancer cells, as well as to inhibition of metastasis formation in xenografted mice. The calcium-binding protein S100A4 is a central mediator of metastasis formation in colon cancer. S100A4 is a target gene of the Wnt/β-catenin pathway, which is constitutively active in the majority of colon cancers. In this study a high-throughput screen was performed to identify small-molecule compounds targeting the S100A4-promoter activity. In this screen calcimycin was identified as a transcriptional inhibitor of S100A4. In colon cancer cells calcimycin treatment reduced S100A4 mRNA and protein expression in a dose- and time-dependent manner. S100A4-induced cellular processes associated with metastasis formation, such as cell migration and invasion, were inhibited by calcimycin in an S100A4-specific manner. Calcimycin reduced β-catenin mRNA and protein levels despite the expression of Δ45-mutated β-catenin. Consequently, calcimycin inhibited Wnt/β-catenin pathway activity and the expression of prominent β-catenin target genes such as S100A4, cyclin D1, c-myc, and dickkopf-1. Finally, calcimycin treatment of human colon cancer cells inhibited metastasis formation in xenografted immunodeficient mice. Our results demonstrate that targeting the expression of S100A4 with calcimycin provides a functional strategy to restrict cell motility in colon cancer cells. Therefore calcimycin may be useful for studying S100A4 biology, and these studies may serve as a lead for the development of treatments for colon cancer metastasis.
Collapse
Affiliation(s)
- Ulrike Sack
- Max-Delbrück-Center for Molecular Medicine, 13125 Berlin, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Mao Y, Li Z, Lou C, Zhang Y. Expression of phosphorylated Stat5 predicts expression of cyclin D1 and correlates with poor prognosis of colonic adenocarcinoma. Int J Colorectal Dis 2011; 26:29-35. [PMID: 21079973 DOI: 10.1007/s00384-010-1090-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/27/2010] [Indexed: 02/04/2023]
Abstract
PURPOSE Constitutive activation of signal transducer and activator of transcription-5 (Stat5) was recently found to be associated with tumor progression through stimulating cell proliferation and preventing apoptosis. However, it is not clear how activated Stat5 is expressed in colon cancer. We aimed to investigate the correlation between phosphorylated Stat5 (p-Stat5) expression and cell cycle regulators (cyclin D1) expression in colonic adenocarcinoma and the relationship between expression of these two proteins and various clinicopathological parameters, including overall survival. METHODS P-Stat5 and cyclin D1 expression were determined by immunohistochemical staining from 169 cases of resected colonic adenocarcinoma specimens. RESULTS P-Stat5 expression correlated with cyclin D1 expression (r = 0.250, P = 0.001). P-Stat5-positive staining was associated with the depth of tumor invasion (P = 0.002). Univariate survival analysis showed that lymph node metastasis, distant metastasis, TNM stage (all P < 0.0001), T stage (P = 0.024), p-Stat5-positive expression (P = 0.002), and cyclin D1-positive expression (P = 0.039) were associated with shorter survival in patients with colonic adenocarcinoma. Multivariate survival analysis showed that only distant metastasis (P < 0.001; hazard ratio [HR] = 4.96), TNM stage (P < 0.001; HR = 9.80), and p-Stat5 overexpression (P = 0.020; HR = 1.84) were independent predictors of poor prognosis. CONCLUSIONS Our findings provide the first evidence that p-Stat5 may play an important role in cyclin D1 overexpression and contribute to colonic adenocarcinoma progression.
Collapse
Affiliation(s)
- Yinling Mao
- Department of Gastrointestinal Medical Oncology, The Affiliated Tumor Hospital of Harbin Medical University, No. 150, Haping Road, Harbin, Heilongjiang Province 150040, China
| | | | | | | |
Collapse
|
23
|
Tentler JJ, Nallapareddy S, Tan AC, Spreafico A, Pitts TM, Morelli MP, Selby HM, Kachaeva MI, Flanigan SA, Kulikowski GN, Leong S, Arcaroli JJ, Messersmith WA, Eckhardt SG. Identification of predictive markers of response to the MEK1/2 inhibitor selumetinib (AZD6244) in K-ras-mutated colorectal cancer. Mol Cancer Ther 2010; 9:3351-62. [PMID: 20923857 DOI: 10.1158/1535-7163.mct-10-0376] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mutant K-ras activity leads to the activation of the RAS/RAF/MEK/ERK pathway in approximately 44% of colorectal cancer (CRC) tumors. Accordingly, several inhibitors of the MEK pathway are under clinical evaluation in several malignancies including CRC. The aim of this study was to develop and characterize predictive biomarkers of response to the MEK1/2 inhibitor AZD6244 in CRC in order to maximize the clinical utility of this agent. Twenty-seven human CRC cell lines were exposed to AZD6244 and classified according to the IC(50) value as sensitive (≤ 0.1 μmol/L) or resistant (>1 μmol/L). All cell lines were subjected to immunoblotting for effector proteins, K-ras/BRAF mutation status, and baseline gene array analysis. Further testing was done in cell line xenografts and K-ras mutant CRC human explants models to develop a predictive genomic classifier for AZD6244. The most sensitive and resistant cell lines were subjected to differential gene array and pathway analyses. Members of the Wnt signaling pathway were highly overexpressed in cell lines resistant to AZD6244 and seem to be functionally involved in mediating resistance by shRNA knockdown studies. Baseline gene array data from CRC cell lines and xenografts were used to develop a k-top scoring pair (k-TSP) classifier, which predicted with 71% accuracy which of a test set of patient-derived K-ras mutant CRC explants would respond to AZD6244, providing the basis for a patient-selective clinical trial. These results also indicate that resistance to AZD6244 may be mediated, in part, by the upregulation of the Wnt pathway, suggesting potential rational combination partners for AZD6244 in CRC.
Collapse
Affiliation(s)
- John J Tentler
- Division of Medical Oncology, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado 80045, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Armanious H, Gelebart P, Mackey J, Ma Y, Lai R. STAT3 upregulates the protein expression and transcriptional activity of β-catenin in breast cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2010; 3:654-664. [PMID: 20830236 PMCID: PMC2933385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Revised: 07/25/2010] [Accepted: 07/23/2010] [Indexed: 05/29/2023]
Abstract
The expression of β-catenin detectable by immunohistochemistry has been reported to be prognostically important in breast cancer. In this study, we investigated the mechanism by which β-catenin is regulated in breast cancer cells. Our analysis of the gene promoter of β-catenin revealed multiple putative STAT3 binding sites. In support of the concept that STAT3 is a transcriptional regulator for β-catenin, results from our chromatin immunoprecipitation studies showed that STAT3 binds to two of the three potential STAT3-binding sites in the gene promoter of β-catenin (-856 and -938). Using our generated MCF-7 cell clones that carry an inducible STAT3C construct, we found that the expression levels of STAT3C significantly correlated with the transcriptional activity of β-catenin. Similar observations were made when we subjected two breast cancer cell lines (MCF-7 and BT-474) to STAT3 knock-down or transient gene transfection of STAT3C. Using immunohistochemistry, we found that pSTAT3 and β-catenin significantly correlated with each other (p=0.003, Fisher's exact test) in a cohort of primary breast tumors (n=129). To conclude, our results support the concept that STAT3 upregulates the protein expression and transcriptional activity of β-catenin in breast cancer, and these two proteins may cooperate with each other in exerting their oncogenic effects in these tumors.
Collapse
Affiliation(s)
- Hanan Armanious
- Department of Laboratory Medicine and Pathology, Cross Cancer Institute and University of Alberta Edmonton, Alberta, Canada
| | | | | | | | | |
Collapse
|
25
|
Fan Y, Jiang WZ, Wen JJ, Hao WL, Du JN, Liu X, Qian M. B7-DC-silenced dendritic cells induce stronger anti-HBV immunity in transgenic mice. Arch Virol 2009; 154:1813-21. [PMID: 19838621 DOI: 10.1007/s00705-009-0521-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Accepted: 09/15/2009] [Indexed: 01/22/2023]
Abstract
Hepatitis B virus (HBV) is a noncytopathic DNA virus and is the pathogen of acute and chronic hepatitis. Interferon and nucleotide analogues such as lamivudine and adefovir are the current treatment strategies of HBV infection; however, it is still a serious disease. Therefore, the development of new therapeutic options against HBV is needed. In the present study, we have investigated whether the vectors carrying short hairpin RNA (shRNA) targeting the murine B7-DC gene could silence the expression of B7-DC and analyzed the function of gene-modified dendritic cells (DCs) by mixed lymphocyte reaction. The results demonstrated that two shRNA vectors efficiently suppressed the expression of B7-DC. The MLR assay showed that shRNA-B7-DC-transfected DCs induced markedly higher allogeneic lymphocyte proliferation than transfected DCs with the vector plasmid pAS and untreated DCs at all dilutions. The most efficient shRNA plasmid vector against B7-DC was then used to silence the expression of B7-DC on DCs, the gene-modified DCs were pulsed with HBV-specific peptides, and HBV transgenic mice were immunized. After three rounds of immunization, the splenocytes were stimulated in vitro and tested for cytotoxicitic T lymphocyte activity, while the sera were used to detect the level of HBsAg and HBV DNA. The data demonstrated that blockade of B7-DC on DCs augmented the cytolytic activity induced by immunization with peptide-pulsed DCs and significantly reduced the concentration of serum HBsAg and HBV DNA, suggesting that silencing of B7-DC is of potential value in DC-based therapy of HBV infection.
Collapse
Affiliation(s)
- Yan Fan
- School of Life Science, East China Normal University, Shanghai, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
26
|
Nonviral vector-mediated RNA interference: its gene silencing characteristics and important factors to achieve RNAi-based gene therapy. Adv Drug Deliv Rev 2009; 61:760-6. [PMID: 19386274 DOI: 10.1016/j.addr.2009.04.006] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2009] [Accepted: 04/05/2009] [Indexed: 01/03/2023]
Abstract
RNA interference (RNAi) is a potent and specific gene silencing event in which small interfering RNA (siRNA) degrades target mRNA. Therefore, RNAi is of potential use as a therapeutic approach for the treatment of a variety of diseases in which aberrant expression of mRNA causes a problem. RNAi can be achieved by delivering siRNA or vectors that transcribe siRNA or short-hairpin RNA (shRNA). The aim of this review is to examine the potential of nonviral vector-mediated RNAi technology in treating diseases. The characteristics of plasmid DNA expressing shRNA were compared with those of siRNA, focusing on the duration of gene silencing, delivery to target cells and target specificity. Recent progresses in prolonging the RNAi effect, improving the delivery to target cells and increasing the specificity of RNAi in vivo are also reviewed.
Collapse
|
27
|
Dominguez I, Sonenshein GE, Seldin DC. Protein kinase CK2 in health and disease: CK2 and its role in Wnt and NF-kappaB signaling: linking development and cancer. Cell Mol Life Sci 2009; 66:1850-7. [PMID: 19387549 PMCID: PMC3905806 DOI: 10.1007/s00018-009-9153-z] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
CK2 is a highly conserved tetrameric serine/ threonine kinase present in all eukaryotic organisms. It is constitutively active, and appears to be regulated by level of expression and activity, and subcellular localization. In turn, it has been postulated to control the function of many proteins through changes in phosphorylation that affect protein stability, protein-protein interactions, and subcellular localization. Through these mechanisms, CK2 regulates many fundamental cellular properties. An enzyme that carries out such a master regulatory function is likely to be important in organismic development and in cancer. We have shown that overexpression of CK2 catalytic subunits is capable of promoting tumorigenesis, and that loss of CK2 catalytic subunits in development can be lethal. Through studies in cells, mice, and frogs, we and others have identified the Wnt and NF-kappaB pathways as two key signal transduction pathways that are regulated by CK2 activity, in embryonic development and in cancer. These results suggest that inhibiting CK2 could be useful in treating cancer, but dangerous to developing organisms.
Collapse
Affiliation(s)
- I. Dominguez
- Section of Hematology-Oncology, Department of Medicine, Boston University School of Medicine and Boston Medical Center, 650 Albany Street, Boston, Massachusetts 02118 USA
| | - G. E. Sonenshein
- Section of Hematology-Oncology, Department of Medicine, Boston University School of Medicine and Boston Medical Center, 650 Albany Street, Boston, Massachusetts 02118 USA
| | - D. C. Seldin
- Section of Hematology-Oncology, Department of Medicine, Boston University School of Medicine and Boston Medical Center, 650 Albany Street, Boston, Massachusetts 02118 USA
| |
Collapse
|
28
|
Min HJ, Cho IR, Srisuttee R, Park EH, Cho DH, Ahn JH, Lee IS, Johnston RN, Oh S, Chung YH. Hexachlorophene suppresses beta-catenin expression by up-regulation of Siah-1 in EBV-infected B lymphoma cells. Cancer Lett 2008; 276:136-42. [PMID: 19091460 PMCID: PMC7116996 DOI: 10.1016/j.canlet.2008.10.041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Revised: 10/29/2008] [Accepted: 10/29/2008] [Indexed: 01/27/2023]
Abstract
Many studies have shown that the activation of β-catenin signaling can promote oncogenesis, and it is therefore of interest to find agents that modulate this pathway. Recent work has shown using B lymphoma cells that infection by Epstein–Barr virus (EBV) and expression of its latent membrane protein (LMP)-1, cause increases in the expression of β-catenin and cellular transformation. Conversely, results from cell-based small molecule screening studies have shown that the antibiotic hexachlorophene can down-regulate β-catenin in colon cancer cells. Here we report that hexachlorophene also counteracts the elevated β-catenin levels in EBV-infected B lymphomas. This is associated with restoration in levels of Siah-1 (an E3 ubiquitin ligase that is active in β-catenin regulation) which had been diminished by LMP-1. Our results suggest that Siah-1 is targeted by both LMP-1 and hexachlorophene with opposite effects. The hexachlorophene modulation of Siah-1 and β-catenin is independent of p53 and results in reduced expression of cyclin-D1 and c-Myc (target genes of β-catenin), leading to the growth arrest of B lymphoma cells. From these results we propose that hexachlorophene may provide a novel therapeutic strategy for EBV-infected B lymphoma cells by reducing β-catenin levels via the restoration of Siah-1.
Collapse
Affiliation(s)
- Hye-Jin Min
- Department of Nanomedical Engineering, BK21 Nanofusion Technology Team, Pusan National University, Miryang, Gyeongnam, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Wang JS, Zheng CL, Wang YJ, Wen JF, Ren HZ, Liu Y, Jiang HY. Gene silencing of beta-catenin by RNAi inhibits cell proliferation in human esophageal cancer cells in vitro and in nude mice. Dis Esophagus 2008; 22:151-62. [PMID: 19021690 DOI: 10.1111/j.1442-2050.2008.00875.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
beta-Catenin, which is frequently overexpressed in a variety of human cancers including esophageal cancer, mediates cancer cell proliferation and tumor growth. In the present study, we used a human U6 promoter-driven DNA-template approach to induce short hairpin RNA (shRNA)-triggered RNA interference to silence beta-catenin gene expression in human esophageal squamous cell carcinoma cell line Eca-109, and then evaluated its effects on the proliferation and growth of tumor cells in vitro and in nude mice. beta-Catenin expression levels decreased markedly in Eca-109 cells transfected with a plasmid expressing shRNA for beta-catenin. Downregulation of beta-catenin was concomitantly accompanied by reduction of cyclin D1, colony formation, and growth inhibition of Eca-109 cells in vitro. The mechanism appears to be the G0/G1 phase arrest but not induction of apoptosis. In vivo, treatment of Eca-109 cells with beta-catenin shRNA greatly impeded tumor growth in nude mice. We conclude that plasmid vector-mediated beta-catenin RNA interference holds great promise as a novel treatment on human esophageal cancer with beta-catenin overexpression.
Collapse
Affiliation(s)
- Jin-Sheng Wang
- Department of Pathology, Xiangya Medical College, Central South University, Changsha, China
| | | | | | | | | | | | | |
Collapse
|