1
|
Sharma V, Chaudhary AA, Bawari S, Gupta S, Mishra R, Khan SUD, Ali MAM, Shahid M, Srivastava S, Verma D, Gupta A, Kumar S, Kumar S. Unraveling cancer progression pathways and phytochemical therapeutic strategies for its management. Front Pharmacol 2024; 15:1414790. [PMID: 39246660 PMCID: PMC11377287 DOI: 10.3389/fphar.2024.1414790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 05/09/2024] [Indexed: 09/10/2024] Open
Abstract
Cancer prevention is currently envisioned as a molecular-based approach to prevent carcinogenesis in pre-cancerous stages, i.e., dysplasia and carcinoma in situ. Cancer is the second-leading cause of mortality worldwide, and a more than 61% increase is expected by 2040. A detailed exploration of cancer progression pathways, including the NF-kβ signaling pathway, Wnt-B catenin signaling pathway, JAK-STAT pathway, TNF-α-mediated pathway, MAPK/mTOR pathway, and apoptotic and angiogenic pathways and effector molecules involved in cancer development, has been discussed in the manuscript. Critical evaluation of these effector molecules through molecular approaches using phytomolecules can intersect cancer formation and its metastasis. Manipulation of effector molecules like NF-kβ, SOCS, β-catenin, BAX, BAK, VEGF, STAT, Bcl2, p53, caspases, and CDKs has played an important role in inhibiting tumor growth and its spread. Plant-derived secondary metabolites obtained from natural sources have been extensively studied for their cancer-preventing potential in the last few decades. Eugenol, anethole, capsaicin, sanguinarine, EGCG, 6-gingerol, and resveratrol are some examples of such interesting lead molecules and are mentioned in the manuscript. This work is an attempt to put forward a comprehensive approach to understanding cancer progression pathways and their management using effector herbal molecules. The role of different plant metabolites and their chronic toxicity profiling in modulating cancer development pathways has also been highlighted.
Collapse
Affiliation(s)
- Vikas Sharma
- Metro College of Health Sciences and Research, Greater Noida, India
- School of Pharmacy, Sharda University, Greater Noida, India
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Sweta Bawari
- Amity Institute of Pharmacy, Amity University, Noida, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, India
| | - Richa Mishra
- Department of Computer Engineering, Parul University, Vadodara, India
| | - Salah-Ud-Din Khan
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Mohamed A M Ali
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh, Saudi Arabia
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Mohammad Shahid
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | | | - Devvrat Verma
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, Uttarakhand, India
| | - Arti Gupta
- Lloyd School of Pharmacy, Greater Noida, India
| | - Sanjay Kumar
- Biological and Bio-computational Laboratory, Department of Life Science, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, India
| | - Sandeep Kumar
- School of Pharmacy, Sharda University, Greater Noida, India
- DST-FIST Laboratory, Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, India
| |
Collapse
|
2
|
Gad HA, Diab AM, Elsaied BE, Tayel AA. Biopolymer-based formulations for curcumin delivery toward cancer management. CURCUMIN-BASED NANOMEDICINES AS CANCER THERAPEUTICS 2024:309-338. [DOI: 10.1016/b978-0-443-15412-6.00009-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
3
|
Ries A, Flehberger D, Slany A, Pirker C, Mader JC, Mohr T, Schelch K, Sinn K, Mosleh B, Hoda MA, Dome B, Dolznig H, Krupitza G, Müllauer L, Gerner C, Berger W, Grusch M. Mesothelioma-associated fibroblasts enhance proliferation and migration of pleural mesothelioma cells via c-Met/PI3K and WNT signaling but do not protect against cisplatin. J Exp Clin Cancer Res 2023; 42:27. [PMID: 36683050 PMCID: PMC9869633 DOI: 10.1186/s13046-022-02582-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 12/24/2022] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Pleural mesothelioma (PM) is an aggressive malignancy with poor prognosis. Unlike many other cancers, PM is mostly characterized by inactivation of tumor suppressor genes. Its highly malignant nature in absence of tumor driving oncogene mutations indicates an extrinsic supply of stimulating signals by cells of the tumor microenvironment (TME). Cancer-associated fibroblasts (CAFs) are an abundant cell type of the TME and have been shown to drive the progression of several malignancies. The aim of the current study was to isolate and characterize patient-derived mesothelioma-associated fibroblasts (Meso-CAFs), and evaluate their impact on PM cells. METHODS Meso-CAFs were isolated from surgical specimens of PM patients and analyzed by array comparative genomic hybridization, next generation sequencing, transcriptomics and proteomics. Human PM cell lines were retrovirally transduced with GFP. The impact of Meso-CAFs on tumor cell growth, migration, as well as the response to small molecule inhibitors, cisplatin and pemetrexed treatment was investigated in 2D and 3D co-culture models by videomicroscopy and automated image analysis. RESULTS Meso-CAFs show a normal diploid genotype without gene copy number aberrations typical for PM cells. They express CAF markers and lack PM marker expression. Their proteome and secretome profiles clearly differ from normal lung fibroblasts with particularly strong differences in actively secreted proteins. The presence of Meso-CAFs in co-culture resulted in significantly increased proliferation and migration of PM cells. A similar effect on PM cell growth and migration was induced by Meso-CAF-conditioned medium. Inhibition of c-Met with crizotinib, PI3K with LY-2940002 or WNT signaling with WNT-C59 significantly impaired the Meso-CAF-mediated growth stimulation of PM cells in co-culture at concentrations not affecting the PM cells alone. Meso-CAFs did not provide protection of PM cells against cisplatin but showed significant protection against the EGFR inhibitor erlotinib. CONCLUSIONS Our study provides the first characterization of human patient-derived Meso-CAFs and demonstrates a strong impact of Meso-CAFs on PM cell growth and migration, two key characteristics of PM aggressiveness, indicating a major role of Meso-CAFs in driving PM progression. Moreover, we identify signaling pathways required for Meso-CAF-mediated growth stimulation. These data could be relevant for novel therapeutic strategies against PM.
Collapse
Affiliation(s)
- Alexander Ries
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090, Vienna, Austria
| | - Daniela Flehberger
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090, Vienna, Austria
| | - Astrid Slany
- Department of Analytical Chemistry, University of Vienna, Waehringer Straße 38, 1090, Vienna, Austria
| | - Christine Pirker
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090, Vienna, Austria
| | - Johanna C Mader
- Department of Analytical Chemistry, University of Vienna, Waehringer Straße 38, 1090, Vienna, Austria
| | - Thomas Mohr
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090, Vienna, Austria
- Department of Analytical Chemistry, University of Vienna, Waehringer Straße 38, 1090, Vienna, Austria
- Joint Metabolome Facility, University of Vienna and Medical University of Vienna, Waehringer Guertel 38, 1090, Vienna, Austria
- ScienceConsult - DI Thomas Mohr KG, Enzianweg 10a, 2353, Guntramsdorf, Austria
| | - Karin Schelch
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090, Vienna, Austria
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Katharina Sinn
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Berta Mosleh
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Mir Alireza Hoda
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
| | - Balazs Dome
- Department of Thoracic Surgery, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, 1090, Austria
- National Korányi Institute of Pulmonology, Korányi Frigyes u. 1, Budapest, 1122, Hungary
- Department of Thoracic Surgery, National Institute of Oncology, Semmelweis University, Rath Gyorgy u. 7-9, Budapest, 1122, Hungary
| | - Helmut Dolznig
- Institute of Medical Genetics, Medical University of Vienna, Waehringer Straße 10, 1090, Vienna, Austria
| | - Georg Krupitza
- Department of Clinical Pathology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Leonhard Müllauer
- Department of Clinical Pathology, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Christopher Gerner
- Department of Analytical Chemistry, University of Vienna, Waehringer Straße 38, 1090, Vienna, Austria
| | - Walter Berger
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090, Vienna, Austria
| | - Michael Grusch
- Center for Cancer Research and Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, 1090, Vienna, Austria.
| |
Collapse
|
4
|
Gasper W, Rossi F, Ligorio M, Ghersi D. Variant calling enhances the identification of cancer cells in single-cell RNA sequencing data. PLoS Comput Biol 2022; 18:e1010576. [PMID: 36191033 PMCID: PMC9560611 DOI: 10.1371/journal.pcbi.1010576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/13/2022] [Accepted: 09/15/2022] [Indexed: 12/14/2022] Open
Abstract
Single-cell RNA-sequencing is an invaluable research tool that allows for the investigation of gene expression in heterogeneous cancer cell populations in ways that bulk RNA-seq cannot. However, normal (i.e., non tumor) cells in cancer samples have the potential to confound the downstream analysis of single-cell RNA-seq data. Existing methods for identifying cancer and normal cells include copy number variation inference, marker-gene expression analysis, and expression-based clustering. This work aims to extend the existing approaches for identifying cancer cells in single-cell RNA-seq samples by incorporating variant calling and the identification of putative driver alterations. We found that putative driver alterations can be detected in single-cell RNA-seq data obtained with full-length transcript technologies and noticed that a subset of cells in tumor samples are enriched for putative driver alterations as compared to normal cells. Furthermore, we show that the number of putative driver alterations and inferred copy number variation are not correlated in all samples. Taken together, our findings suggest that augmenting existing cancer-cell filtering methods with variant calling and analysis can increase the number of tumor cells that can be confidently included in downstream analyses of single-cell full-length transcript RNA-seq datasets.
Collapse
Affiliation(s)
- William Gasper
- School of Interdisciplinary Informatics, University of Nebraska at Omaha, Omaha, Nebraska, United States of America
| | - Francesca Rossi
- Department of Surgery, University of Texas Southwestern, Dallas, Texas, United States of America
| | - Matteo Ligorio
- Department of Surgery, University of Texas Southwestern, Dallas, Texas, United States of America
| | - Dario Ghersi
- School of Interdisciplinary Informatics, University of Nebraska at Omaha, Omaha, Nebraska, United States of America
| |
Collapse
|
5
|
Boligala GP, Yang MV, van Wunnik JC, Pruitt K. Nuclear Dishevelled: An enigmatic role in governing cell fate and Wnt signaling. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119305. [PMID: 35688346 DOI: 10.1016/j.bbamcr.2022.119305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/26/2022] [Accepted: 05/29/2022] [Indexed: 06/15/2023]
Abstract
The Dishevelled gene was first identified in Drosophila mutants with disoriented hair and bristle polarity and subsequent work has now demonstrated its importance in critical and diverse aspects of biology. Since those early discoveries, Dishevelled has been shown to coordinate a plethora of developmental and cellular processes that range from controlling cell polarity during gastrulation to partnering with chromatin modifying enzymes to regulate histone methylation at genomic loci. While the role of DVL in development is well-respected and the cytosolic function of DVL has been studied more extensively, its nuclear role continues to remain murky. In this review we highlight some of the seminal discoveries that have contributed to the field, but the primary focus is to discuss recent advances with respect to the nuclear role of Dishevelled. This nuclear function of Dishevelled is a dimension which is proving to be increasingly important yet remains enigmatic.
Collapse
Affiliation(s)
- Geetha Priya Boligala
- Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Mingxiao V Yang
- Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Jenna C van Wunnik
- Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Kevin Pruitt
- Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
6
|
Han L, Shi J, Zhao L, Deng J, Li Y, Zhao H, Wang H, Yan Y, Zou F. BCAP31 is involved in modulating colorectal cancer cell proliferation via the Emerin/β-catenin axis. Exp Cell Res 2022; 418:113265. [PMID: 35716785 DOI: 10.1016/j.yexcr.2022.113265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/12/2022] [Accepted: 06/13/2022] [Indexed: 11/15/2022]
Abstract
Understanding the mechanisms of colorectal cancer (CRC) progression is critical for developing innovative treatment strategies. As an endoplasmic reticulum-located protein, B cell receptor-associated protein 31 (BCAP31) has been identified to be highly expressed in multiple cancers. However, its function and molecular mechanism in CRC remain not fully understood. In the present study, BCAP31 expression and its correlation with the clinical stage were analyzed based on TCGA database. We demonstrated that loss of BCAP31 suppressed CRC cell proliferation in vitro and tumor growth in vivo. Mechanistically, we demonstrated that Emerin was an interaction partner and downstream molecule of BCAP31. Knockdown of BCAP31 promoted the nuclear envelope localization of Emerin, leading to a reduction of β-catenin accumulation in the nucleus, which resulted in downregulation of Wnt/β-catenin downstream target genes, including c-Myc, cyclin D1, Survivin, and Mcl-1. Moreover, downregulation of Emerin partially restored the BCAP31 depletion-mediated β-catenin protein level and tumor suppressive effects in CRC cells.Our data highlights the pivotal role of BCAP31 depletion in inhibiting cell proliferation in CRC cells, and mechanistically via Emerin/β-catenin signaling, which may serve as a promising target for CRC treatment.
Collapse
Affiliation(s)
- Liping Han
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China
| | - Junyang Shi
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China
| | - Lili Zhao
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China
| | - Jiaqiang Deng
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China
| | - Yan Li
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China
| | - Hong Zhao
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China
| | - Huani Wang
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China
| | - Yan Yan
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China
| | - Fangdong Zou
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China.
| |
Collapse
|
7
|
Kim JC, Bodmer WF. Genomic landscape of colorectal carcinogenesis. J Cancer Res Clin Oncol 2022; 148:533-545. [PMID: 35048197 DOI: 10.1007/s00432-021-03888-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/11/2021] [Indexed: 12/19/2022]
|
8
|
Li Y, Gong Y, Zhang X, Wang J, Cheng Y, Liu F, Shi X, Xu W, Dong L. Exploring the synergistic mechanism of Gegen Qinlian Decoction on the Wnt signaling pathway using an integrated strategy of network pharmacology and RNA-seq. JOURNAL OF ETHNOPHARMACOLOGY 2021; 278:114283. [PMID: 34098017 DOI: 10.1016/j.jep.2021.114283] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/27/2021] [Accepted: 05/30/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Gegen Qinlian Decoction (GQD) (including: Puerariae lobatae (Willd.) Ohwi, radix; (short for Gengen) Glycyrrhiza uralensis Fisch., root and rhizome (short for Gancao), honeyed; Coptis chinensis Franch., rhizome (short for Huanglian); Scutellaria baicalensis Georgi, radix, boiled (short for S. baicalensis) has been widely used to treat inflammatory bowel disease (IBD) and colorectal cancer (CRC). To explore compatibility mechanism of GQD could be of advantage to investigate the complex principle of TCM, which might be conducive to the exploration of the modernization of TCM. AIM OF REVIEW In this study, a strategy based on system pharmacology was constructed to uncover the multi-target regulation and compatibility mechanism of GQD on the Wnt signaling pathways. MATERIAL AND METHODS The pharmacological network of GQD was constructed by TCMSP, DAVID, Uniprote database. The cell growth inhibitory effects of puerarin (PUE), wogonin (WOG), berberine (BER), and glycyrrhetinic acid (GLY) on SW480 cells were assessed using CCK-8 assay. The multi-target regulation and compatibility mechanism of combination PUE with GLY were examined by RNA-seq, HPLC-QQQ/MS, qRT- PCR and Western blot analysis. RESULTS Network pharmacology analysis indicated that PUE, WOG, BER and GLY were the active components in GQD and had a synergistic effect on the targets of the Wnt signaling pathway. Additionally, pharmacological experiments revealed that WOG, BER, and GLY inhibited activity of colorectal cancer (CRC) cell lines SW480 cells, and that PUE only exhibited effective antitumour activity when combined with GLY. CTNNB1, CCND1 and SMAD4 were identified as synergistic targets inhibited by PUE-GLY. Moreover, PUE-GLY could influence the Wnt signaling pathway by upregulating GSK3B and downregulating CTNNB1 synergistically. It also showed that GLY could effectively increase the intracellular content of PUE based on HPLC-QQQ/MS analysis, and this process was achieved by influencing the targets of the membrane's pathway, such as cell adhesion molecules, focal adhesion, and tight junctions. CONCLUSION GLY was revealed a multi-target mechanism, which could downregulate CTNNB1 as the active component and intervene in membrane proteins (CDH1, CADM1, ITGB2, ICAM1, ITGA1) as 'guide' in the formulae. Moreover, the mechanism of synergistic antitumour action of PUE (the active component of Monarch drug) and GLY (the active component of Guide drug) on the Wnt signaling pathway was explored systematically. It was a promising breakthrough for elucidating the scientific connotation of the compatibility of TCM formulae and provide a valuable and practicable methodology for clarifying the mechanisms of TCM.
Collapse
Affiliation(s)
- Yanping Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China
| | - Yiting Gong
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China
| | - Xin Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China
| | - Jiaxin Wang
- School of Life Sciences, Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China
| | - Yaru Cheng
- School of Life Sciences, Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China
| | - Fen Liu
- School of Life Sciences, Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China
| | - Xiujia Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China
| | - Wenjuan Xu
- School of Life Sciences, Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China.
| | - Ling Dong
- School of Life Sciences, Beijing University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
9
|
Carbajo-García MC, Corachán A, Segura-Benitez M, Monleón J, Escrig J, Faus A, Pellicer A, Cervelló I, Ferrero H. 5-aza-2'-deoxycitidine inhibits cell proliferation, extracellular matrix formation and Wnt/β-catenin pathway in human uterine leiomyomas. Reprod Biol Endocrinol 2021; 19:106. [PMID: 34233687 PMCID: PMC8265104 DOI: 10.1186/s12958-021-00790-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/18/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Uterine leiomyoma is a benign tumor with unclear pathogenesis and inaccurate treatment. This tumor exhibits altered DNA methylation related to disease progression. DNMT inhibitors as 5-aza-2'-deoxycytidine (5-aza-CdR), have been suggested to treat tumors in which DNA methylation is altered. We aimed to evaluate whether DNA methylation reversion with 5-aza-CdR reduces cell proliferation and extracellular matrix (ECM) formation in uterine leiomyoma cells to provide a potential treatment option. METHODS Prospective study using uterine leiomyoma and adjacent myometrium tissues and human uterine leiomyoma primary (HULP) cells (n = 16). In tissues, gene expression was analyzed by qRT-PCR and DNMT activity by ELISA. Effects of 5-aza-CdR treatment on HULP cells were assessed by CellTiter, western blot, and qRT-PCR. RESULTS DNMT1 gene expression was higher in uterine leiomyoma vs myometrium. Similarly, DNMT activity was greater in uterine leiomyoma and HULP cells (6.5 vs 3.8 OD/h/mg; 211.3 vs 63.7 OD/h/mg, respectively). After 5-aza-CdR treatment on HULP cells, cell viability was reduced, significantly so at 10 μM (85.3%). Treatment with 10 μM 5-aza-CdR on HULP cells significantly decreased expression of proliferation marker PCNA (FC = 0.695) and of ECM proteins (COLLAGEN I FC = 0.654; PAI-1, FC = 0.654; FIBRONECTIN FC = 0.733). 5-aza-CdR treatment also decreased expression of Wnt/β-catenin pathway final targets, including WISP1 protein expression (10 μM, FC = 0.699), c-MYC gene expression (2 μM, FC = 0.745 and 10 μM, FC = 0.728), and MMP7 gene expression (5 μM, FC = 0.520 and 10 μM, FC = 0.577). CONCLUSIONS 5-aza-CdR treatment inhibits cell proliferation, ECM formation, and Wnt/β-catenin signaling pathway targets in HULP cells, suggesting that DNA methylation inhibition is a viable therapeutic target in uterine leiomyoma.
Collapse
Affiliation(s)
- María Cristina Carbajo-García
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell 106, Torre A, Planta 1ª, 46026, Valencia, Spain
- Departamento de Pediatría, Obstetricia y Ginecología, Universidad de Valencia, Valencia, Spain
| | - Ana Corachán
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell 106, Torre A, Planta 1ª, 46026, Valencia, Spain
- Departamento de Pediatría, Obstetricia y Ginecología, Universidad de Valencia, Valencia, Spain
| | - Marina Segura-Benitez
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell 106, Torre A, Planta 1ª, 46026, Valencia, Spain
- Departamento de Pediatría, Obstetricia y Ginecología, Universidad de Valencia, Valencia, Spain
| | - Javier Monleón
- Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Julia Escrig
- Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Amparo Faus
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell 106, Torre A, Planta 1ª, 46026, Valencia, Spain
| | - Antonio Pellicer
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell 106, Torre A, Planta 1ª, 46026, Valencia, Spain
- IVIRMA Rome, Rome, Italy
| | - Irene Cervelló
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell 106, Torre A, Planta 1ª, 46026, Valencia, Spain
| | - Hortensia Ferrero
- Fundación IVI, Instituto de Investigación Sanitaria La Fe, Avenida Fernando Abril Martorell 106, Torre A, Planta 1ª, 46026, Valencia, Spain.
| |
Collapse
|
10
|
Liu X, Su K, Sun X, Jiang Y, Wang L, Hu C, Zhang C, Lu M, Du X, Xing B. Sec62 promotes stemness and chemoresistance of human colorectal cancer through activating Wnt/β-catenin pathway. J Exp Clin Cancer Res 2021; 40:132. [PMID: 33858476 PMCID: PMC8051072 DOI: 10.1186/s13046-021-01934-6] [Citation(s) in RCA: 113] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Cancer stem cell (CSC)-related chemoresistance leads to poor outcome of the patients with colorectal cancer (CRC). In this study, we identified the chemoresistance-relevant molecules and decipher the involved mechanisms to provide potential therapeutic target for CRC. We focused on Sec62, a novel target with significantly increased expression in chemoresistant CRC tissues, and further investigated its role in the progression of CRC. METHODS Through analyzing the differentially-expressed genes between chemoresistant and chemosensitive CRCs, we selected Sec62 as a novel chemoresistance-related target in CRC. The expression and clinical significance of Sec62 were determined by immunoblotting and immunohistochemistry in tissues and cell lines of CRC. The roles of Sec62 in drug resistance, stemness and tumorigenesis were evaluated in vitro and in vivo using functional experiments. GST pull-down, western blot, coimmunoprecipitation and Me-RIP assays were performed to further explore the downstream molecular mechanisms. RESULTS Sec62 upregulation was associated with the chemoresistance of CRC and poor outcome of CRC patients. Depletion of Sec62 sensitized CRC cells to chemotherapeutic drugs. Sec62 promoted the stemness of CRC cells through activating Wnt/β-catenin signaling. Mechanistically, Sec62 bound to β-catenin and inhibited the degradation of β-catenin. Sec62 competitively disrupted the interaction between β-catenin and APC to inhibit the β-catenin destruction complex assembly. Moreover, Sec62 expression was upregulated by the m6A-mediated stabilization of Sec62 mRNA. CONCLUSIONS Sec62 upregulated by the METTL3-mediated m6A modification promotes the stemness and chemoresistance of CRC by binding to β-catenin and enhancing Wnt signalling. Thus, m6A modification-Sec62-β-catenin molecular axis might act as therapeutic targets in improving treatment of CRC.
Collapse
Affiliation(s)
- Xiaofeng Liu
- Hepatopancreatobiliary Surgery Department I, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Kunqi Su
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Xiaoyan Sun
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yang Jiang
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Lijun Wang
- Hepatopancreatobiliary Surgery Department I, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Chenyu Hu
- Hepatopancreatobiliary Surgery Department I, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Chunfeng Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Min Lu
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Xiaojuan Du
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
| | - Baocai Xing
- Hepatopancreatobiliary Surgery Department I, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
11
|
Morphofunctional Characterization of Different Tissue Factors in Congenital Diaphragmatic Hernia Affected Tissue. Diagnostics (Basel) 2021; 11:diagnostics11020289. [PMID: 33673194 PMCID: PMC7918239 DOI: 10.3390/diagnostics11020289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 12/20/2022] Open
Abstract
Congenital diaphragm hernia (CDH) is a congenital disease that occurs during prenatal development. Although the morbidity and mortality rate is rather significant, the pathogenesis of CDH has been studied insignificantly due to the decreased accessibility of human pathological material. Therefore the aim of our work was to evaluate growth factors (transforming growth factor-beta (TGF-β), basic fibroblast growth factor (bFGF), insulin-like growth factor 1 (IGF-1), hepatocyte growth factor (HGF)) and their receptors (fibroblast growth factor receptor 1 (FGFR1), insulin-like growth factor 1 (IGF-1R)), muscle (dystrophin, myosin, alpha actin) and nerve quality (nerve growth factor (NGF), nerve growth factor receptor (NGFR), neurofilaments (NF)) factors, local defense factors (ß-defensin 2, ß-defensin 4), programmed cell death (TUNEL), and separate gene (Wnt-1) expression in human pathological material to find immunohistochemical marker differences between the control and the CDH patient groups. A semi-quantitative counting method was used for the evaluation of the tissues and structures in the Biotin-Streptavidin-stained slides. Various statistically significant differences were found in immunoreactive expression between the patient and the control group tissue and the morphological structures as well as very strong, strong, and moderate correlations between immunoreactives in different diaphragm cells and structures. These significant changes and various correlations indicate that multiple morphopathogenetic pathways are affected in CDH pathogenesis. This work contains the evaluation of the causes for these changes and their potential involvement in CDH pathogenesis.
Collapse
|
12
|
Zhang J, Zhang S, Li X, Zhang F, Zhao L. HOXB5 promotes the progression of breast cancer through wnt/beta-catenin pathway. Pathol Res Pract 2020; 224:153117. [PMID: 34118725 DOI: 10.1016/j.prp.2020.153117] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 06/28/2020] [Accepted: 07/07/2020] [Indexed: 01/20/2023]
Abstract
OBJECTIVE The present study was designed to explore the function of HOXB5 in breast cancer and related signaling pathway. METHODS Breast cancer tissues and non-cancerous tissues were collected from 82 cases who were pathologically diagnosed with breast cancer. The mRNA level of HOXB5 was detected via quantitative real-time polymerase chain reaction (qRT-PCR). Chi-square test was adopted to analyze the association of HOXB5 with clinical features. The viability, migration and invasion of breast cancer cells were detected through MTT and Transwell assays, respectively. Protein analysis was performed adopting western blot analysis. RESULTS HOXB5 expression was increased in breast cancer tissues and cells, and showed positive correlation with tumor size (P = 0.028), TNM stage (P = 0.048), and lymph node metastasis (P = 0.002). Losing HOXB5 expression suppressed clone formation, proliferation, migration and invasion of breast cancer cells. The knockdown of HOXB5 significantly inactivated wnt/β-catenin pathway. Furthermore, wnt/β-catenin pathway had the potential to neutralize the oncogenic function of HOXB5 in breast cancer. CONCLUSION HOXB5 may be involved in the invasive progression of breast cancer. The function of HOXB5 in breast cancer was mediated by wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Oncology, PLA General Hospital, PLA School of Medicine, Beijing, China
| | - Sujie Zhang
- Department of Oncology, PLA General Hospital, PLA School of Medicine, Beijing, China
| | - Xiaoyan Li
- Department of Oncology, PLA General Hospital, PLA School of Medicine, Beijing, China
| | - Fan Zhang
- Department of Oncology, PLA General Hospital, PLA School of Medicine, Beijing, China
| | - Lei Zhao
- National Clinical Research Center for Normal Aging and Geriatric & The Key Lab of Normal Aging and Geriatric, Institute of Geriatric, PLA General Hospital, Beijing, China.
| |
Collapse
|
13
|
Zu ML, Piao XL, Gao JM, Xing SF, Liu LH. Monomer gypenoside LI from Gynostemma pentaphyllum inhibits cell proliferation and upregulates expression of miR-128-3p in melanoma cells. J Biochem Mol Toxicol 2020; 34:e22460. [PMID: 32022984 DOI: 10.1002/jbt.22460] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 01/14/2020] [Accepted: 01/21/2020] [Indexed: 12/16/2022]
Abstract
Gypenosides have anticancer activity against many cancers. Gypenoside LI is a gypenoside monomer from Gynostemma pentaphyllum, its pharmacological functions in melanoma have not been reported. In this study, we found that gypenoside LI had a potent cytotoxic effect on melanoma cells. Gypenoside LI can induce intrinsic apoptosis along with S phase arrest. Furthermore, gypenoside LI inhibited the colony formation ability of melanoma through inhibition of the Wnt/β-catenin signaling pathway. Interestingly, we also found that gypenoside LI can induce the upregulation of the tumor suppressor miR-128-3p during melanoma apoptosis. In contrast, gypenoside LI induced apoptosis, cell cycle arrest, and inhibition of the Wnt/β-catenin signaling pathway, which were abolished by overexpression of the miR-128-3p inhibitor in A375 cells. Taken together, these results showed that gypenoside LI could inhibit human melanoma cells through inducing apoptosis, arresting cell cycle at the S phase and suppressing the Wnt/β-catenin signaling pathway in a miR-128-3p dependent manner.
Collapse
Affiliation(s)
- Ma-Li Zu
- Department of Biochemistry, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning, China.,School of Pharmacy, Minzu University of China, Beijing, China
| | - Xiang-Lan Piao
- School of Pharmacy, Minzu University of China, Beijing, China
| | - Jia-Mei Gao
- Department of Biochemistry, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Shao-Fang Xing
- School of Pharmacy, Minzu University of China, Beijing, China
| | - Lin-Hua Liu
- Department of Biochemistry, College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
14
|
Liu H, Zhang L, Li G, Gao Z. Xanthohumol protects against Azoxymethane-induced colorectal cancer in Sprague-Dawley rats. ENVIRONMENTAL TOXICOLOGY 2020; 35:136-144. [PMID: 31714664 DOI: 10.1002/tox.22849] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 08/27/2019] [Accepted: 08/30/2019] [Indexed: 06/10/2023]
Abstract
Colorectal cancer (CRC) is a major health problem and third most common deaths in western world. Dietary interventions together with modified dietary style can prevent the CRC in humans. Xanthohumol (XHA), a polyphenol isolated from Humulus lupulus L. contains many beneficial effects. The aim of the study is to analyze the effect of XHA on Azoxymethane (AOM)-induced experimental CRC in rats. Levels of MDA were increased and enzymic antioxidants levels were decreased in AOM-induced rats. However, these levels were reversed upon XHA treatment. Further, the mRNA expressions of iNOS and COX-2 were also downregulated in XHA treated rats compared to AOM-induced rats. Further, we found that administration of XHA suppressed the wnt/β-catenin signaling together with modulation of apoptotic proteins Bax, Bcl-2, and caspase 3. We conclude that XHA can able to quench the free radicals, inhibits cell proliferation and induces apoptosis, thus it can be a chemopreventive/therapeutic agent against CRC.
Collapse
Affiliation(s)
- Hualin Liu
- Endoscopy Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan City, Shandong Province, China
| | - Lei Zhang
- Health Management Center, Qingdao Municipal Hospital, Qingdao City, Shandong Province, China
| | - Guanghua Li
- Department of Gastrointestinal Surgery, The Second Hospital of Shandong University, Jinan City, Shandong Province, China
| | - Zhuanglei Gao
- Department of Gastrointestinal Surgery, The Second Hospital of Shandong University, Jinan City, Shandong Province, China
| |
Collapse
|
15
|
Brocca G, Ferraresso S, Zamboni C, Martinez-Merlo EM, Ferro S, Goldschmidt MH, Castagnaro M. Array Comparative Genomic Hybridization Analysis Reveals Significantly Enriched Pathways in Canine Oral Melanoma. Front Oncol 2019; 9:1397. [PMID: 31921654 PMCID: PMC6920211 DOI: 10.3389/fonc.2019.01397] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 11/26/2019] [Indexed: 12/28/2022] Open
Abstract
Human Mucosal Melanoma (hMM) is an aggressive neoplasm of neuroectodermal origin with distinctive features from the more common cutaneous form of malignant melanoma (cMM). At the molecular level, hMMs are characterized by large chromosomal aberrations rather than single-nucleotide mutations, typically observed in cMM. Given the scarcity of available cases, there have been many attempts to establish a reliable animal model. In pet dogs, Canine Oral Melanoma (COM) is the most common malignant tumor of the oral cavity, sharing clinical and histological aspects with hMM. To improve the knowledge about COM's genomic DNA alterations, in the present work, formalin-fixed, paraffin-embedded (FFPE) samples of COM from different European archives were collected to set up an array Comparative Genomic Hybridization (aCGH) analysis to estimate recurrent Copy Number Aberrations (CNAs). DNA was extracted in parallel from tumor and healthy fractions and 19 specimens were successfully submitted to labeling and competitive hybridization. Data were statistically analyzed through GISTIC2.0 and a pathway-enrichment analysis was performed with ClueGO. Recurrent gained regions were detected, affecting chromosomes CFA 10, 13 and 30, while lost regions involved chromosomes CFA 10, 11, 22, and 30. In particular, CFA 13 showed a whole-chromosome gain in 37% of the samples, while CFA 22 showed a whole-chromosome loss in 25%. A distinctive sigmoidal trend was observed in CFA 10 and 30 in 25 and 30% of the samples, respectively. Comparative analysis revealed that COM and hMM share common chromosomal changes in 32 regions. MAPK- and PI3K-related genes were the most frequently involved, while pathway analysis revealed statistically significant perturbation of cancer-related biological processes such as immune response, drug metabolism, melanocytes homeostasis, and neo-angiogenesis. The latter is a new evidence of a significant involvement of neovascularization-related pathways in COMs and can provide the rationale for future application in anti-cancer targeted therapies.
Collapse
Affiliation(s)
- Ginevra Brocca
- Department of Comparative Biomedicine and Food Science, University of Padua, Legnaro, Italy
| | - Serena Ferraresso
- Department of Comparative Biomedicine and Food Science, University of Padua, Legnaro, Italy
| | - Clarissa Zamboni
- Department of Comparative Biomedicine and Food Science, University of Padua, Legnaro, Italy
| | | | - Silvia Ferro
- Department of Comparative Biomedicine and Food Science, University of Padua, Legnaro, Italy
| | - Michael H Goldschmidt
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Massimo Castagnaro
- Department of Comparative Biomedicine and Food Science, University of Padua, Legnaro, Italy
| |
Collapse
|
16
|
Bu H, Liu D, Cui J, Cai K, Shen F. Wnt/β-catenin signaling pathway is involved in induction of apoptosis by oridonin in colon cancer COLO205 cells. Transl Cancer Res 2019; 8:1782-1794. [PMID: 35116929 PMCID: PMC8797304 DOI: 10.21037/tcr.2019.08.25] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 08/07/2019] [Indexed: 11/20/2022]
Abstract
Background Oridonin has been demonstrated to have anticancer effect on all kinds of cancer cells and it has shown anti-tumor activity in some tumors partially via the inactivation of Wnt/β-catenin signaling pathway. The study investigated the anticancer effect of oridonin on colon carcinoma cell line COLO205 and explored underlying mechanism. Methods Cell Counting Kit-8 (CCK-8) assay was performed to assess cell viability. Flow cytometry was performed to analyze the apoptosis. The key target genes and proteins involved in Wnt/β-catenin pathway were detected by quantitative polymerase chain reaction (qPCR) and Western blotting. The xenograft tumor model of colon cancer COLO205 cell was introduced to detect anti-tumor effects in vivo. Transferase-mediated dUTP nick end labeling (TUNEL) assay was adopted to test the apoptotic cells in the tumor tissues. Results Oridonin inhibited the proliferation of colon cancer COLO205 cells in a dose-dependent and time-dependent manner. Oridonin induced apoptosis by increasing the cleavage of caspases in vitro. Furthermore, the expression levels of β-catenin and its downstream targets, including c-myc, cyclinD1 and survivin were significantly reduced. Nevertheless the knockdown of β-catenin by specific small interfering RNA (siRNA) could augment the anti-proliferative and pro-apoptotic effects by oridonin in COLO205 cells. Meanwhile, oridonin also increased protein expression level of glycogen synthase kinase 3β (GSK3β) and decreased the phosphorylation level of GSK3β. In vivo, oridonin treatment significantly suppressed tumor growth of COLO205 cell xenografts, and which was accompanied by the restrain of Wnt/β-catenin pathway. Conclusions Our present study demonstrated that the growth inhibition and apoptosis induction in colon cancer COLO205 cells by oridonin could be partially mediated through discontinuing Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Heqi Bu
- Department of Coloproctological Surgery, Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| | - Dianlei Liu
- Department of Surgery, Women's Hospital School of Medical Zhejiang University, Hangzhou 310006, China
| | - Junhui Cui
- Department of Coloproctological Surgery, Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| | - Ke Cai
- Department of Coloproctological Surgery, Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| | - Feng Shen
- Department of Coloproctological Surgery, Tongde Hospital of Zhejiang Province, Hangzhou 310012, China
| |
Collapse
|
17
|
Chai WX, Sun LG, Dai FH, Shao HS, Zheng NG, Cai HY. Inhibition of PRRX2 suppressed colon cancer liver metastasis via inactivation of Wnt/β-catenin signaling pathway. Pathol Res Pract 2019; 215:152593. [PMID: 31471104 DOI: 10.1016/j.prp.2019.152593] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/23/2019] [Accepted: 08/09/2019] [Indexed: 02/07/2023]
Abstract
The aim of this study was to investigate whether PRRX2 may regulate the liver metastasis of colon cancer via the Wnt/β-catenin signaling pathway. PRRX2 and β-catenin in patients with the liver metastases of colon cancer was detected by immunochemistry. Colon cancer cells (CT-26 and CMT93) were divided into Normal, si-Ctrl, si-PRRX2 and si-PRRX2 +LiCl groups. Cell invasive and migrating abilities and the related proteins were detected. Liver-metastatic mice model was constructed consisting of Normal, NC shRNA and PRRX2 shRNA groups to examine the function of PRRX2 shRNA on liver metastasis. We found that PRRX2 and β-catenin positive rate was elevated in colon cancer tissues, especially in those tissues with liver metastasis, and there was a close relation between PRRX2 and the clinical staging, lymph node metastasis and numbers of liver metastases of colon cancer patients with liver metastasis. In vitro, the invasive and migrating abilities of CT-26 and CMT93 cells decreased apparently in the si-PRRX2 group, with down-regulation of PRRX2, p-GSK3βSer9/GSK3β, nucleus and cytoplasm β-catenin, TCF4 and Vimentin but up-regulation of E-cadherin. However, LiCl, the Wnt/β-catenin pathway activator, can reverse the inhibitory effect of si-PRRX2 on invasive and migrating ability of colon cancer cells. In vivo, the volume and weight of transplanted tumor and the number of liver metastases in the PRRX2 shRNA group were significantly reduced, with the similar protein expression patterns as in vitro. In a word, PRRX2 inhibition may reduce invasive and migrating abilities to hinder epithelial-mesenchymal transition (EMT), and suppress colon cancer liver metastasis through inactivation of Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Wen-Xiao Chai
- Department of Interventional Oncology, Gansu Provincial People's Hospital, Lanzhou 730000, Gansu, China
| | - Li-Guo Sun
- Department of Surgical Oncology, Dingxi City People's Hospital, Dingxi 743000, Gansu, China
| | - Fu-Hong Dai
- Department of Interventional Oncology, Gansu Provincial People's Hospital, Lanzhou 730000, Gansu, China
| | - Hong-Sheng Shao
- Department of Radiology, Rehabilitation Center Hospital of Gansu Province, Lanzhou 730000, Gansu, China
| | - Ning-Gang Zheng
- Department of Interventional Oncology, Gansu Provincial People's Hospital, Lanzhou 730000, Gansu, China
| | - Hong-Yi Cai
- Department of Radiation oncology, Gansu Provincial People's Hospital, Lanzhou 730000, Gansu, China.
| |
Collapse
|
18
|
Allgayer H, Leupold JH, Patil N. Defining the "Metastasome": Perspectives from the genome and molecular landscape in colorectal cancer for metastasis evolution and clinical consequences. Semin Cancer Biol 2019; 60:1-13. [PMID: 31362074 DOI: 10.1016/j.semcancer.2019.07.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 02/07/2023]
Abstract
Metastasis still poses the highest challenge for personalized therapy in cancer, partly due to a still incomplete understanding of its molecular evolution. We recently presented the most comprehensive whole-genome study of colorectal metastasis vs. matched primary tumors and suggested novel components of disease progression and metastasis evolution, some of them potentially relevant for targeted therapy. In this review, we try to put these findings into perspective with latest discoveries of colleagues and recent literature, and propose a systematic international team effort to collectively define the "metastasome", a term we introduce to summarize all genomic, epigenomic, transcriptomic, further -omic, molecular and functional characteristics rendering metastases different from primary tumors. Based on recent discoveries, we propose a revised metastasis model for colorectal cancer which is based on a common ancestor clone, early dissemination but flexible early or late stage clonal separation paralleling stromal interactions. Furthermore, we discuss hypotheses on site-specific metastasis, colorectal cancer progression, metastasis-targeted diagnosis and therapy, and metastasis prevention based on latest metastasome data.
Collapse
Affiliation(s)
- Heike Allgayer
- Department of Experimental Surgery - Cancer Metastasis, Medical Faculty Mannheim, Theodor Kutzer Ufer 1-3, 68135, Mannheim, Ruprecht Karls University of Heidelberg, Germany; Centre for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Ludolf-Krehl-Str. 6, 68135, Mannheim, Ruprecht Karls University of Heidelberg, Germany.
| | - Jörg H Leupold
- Department of Experimental Surgery - Cancer Metastasis, Medical Faculty Mannheim, Theodor Kutzer Ufer 1-3, 68135, Mannheim, Ruprecht Karls University of Heidelberg, Germany; Centre for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Ludolf-Krehl-Str. 6, 68135, Mannheim, Ruprecht Karls University of Heidelberg, Germany
| | - Nitin Patil
- Department of Experimental Surgery - Cancer Metastasis, Medical Faculty Mannheim, Theodor Kutzer Ufer 1-3, 68135, Mannheim, Ruprecht Karls University of Heidelberg, Germany; Centre for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Ludolf-Krehl-Str. 6, 68135, Mannheim, Ruprecht Karls University of Heidelberg, Germany
| |
Collapse
|
19
|
Yücel ÇY, Erden G, Yılmaz FM, Sezer S, Çalcı E. IGF-I and IGFBP-3 levels and their correlations with carcinoembryonic antigen in colorectal cancer patients. ALEXANDRIA JOURNAL OF MEDICINE 2019. [DOI: 10.1016/j.ajme.2017.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Affiliation(s)
| | - Gönül Erden
- Ankara Numune Training and Research Hospital, Biochemistry Clinic, Turkey
| | - Fatma Meriç Yılmaz
- Yıldırım Beyazıt University, Medical Faculty, Department of Biochemistry, Turkey
| | - Sevilay Sezer
- Ankara Numune Training and Research Hospital, Biochemistry Clinic, Turkey
| | - Esin Çalcı
- Ankara Numune Training and Research Hospital, Biochemistry Clinic, Turkey
| |
Collapse
|
20
|
Yang WD, Wang L. MCM10 facilitates the invaded/migrated potentials of breast cancer cells via Wnt/β-catenin signaling and is positively interlinked with poor prognosis in breast carcinoma. J Biochem Mol Toxicol 2019; 33:e22330. [PMID: 30990947 DOI: 10.1002/jbt.22330] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/27/2019] [Accepted: 03/15/2019] [Indexed: 12/16/2022]
Abstract
The minichromosome maintenance protein 10 (MCM10) is one of the MCM proteins that initiate DNA replication by interacting with CDC45-MCM2-7. It has been reported that MCM10 has a role in breast cancer progression. However, MCM10 in breast cancer is still not comprehensively studied and further research is needed. This study was aimed at investigating the potential effects of MCM10 on metastasis, the prognosis of breast carcinoma, and its underlying mechanisms. Using the ONCOMINE database and the Kaplan-Meier Plotter, MCM10 was significantly overexpressed in cancers, and high expression of MCM10 was involved in the poor prognosis of breast carcinoma. MCM10 can promote the proliferation, migration, and invasion of MDA-MB-231 cells. MCM10 knockdown brought about a radical reversal in cell behaviors. Meanwhile, decreased expression of β-catenin and cyclin Dl was detected in MCM10 short hairpin RNA cells, implying that MCM10 might induce breast cancer metastasis via the Wnt/β-catenin pathway.MCM10 can be defined as a potential diagnostic tool and a promising target for breast carcinoma.
Collapse
Affiliation(s)
- Wei-Dong Yang
- Department of Thyroid and Breast Surgery, People's Hospital of Three Gorges University, Yichang, Hubei, China
| | - Lu Wang
- Department of Thyroid and Breast Surgery, People's Hospital of Three Gorges University, Yichang, Hubei, China
| |
Collapse
|
21
|
Gowrikumar S, Ahmad R, Uppada SB, Washington MK, Shi C, Singh AB, Dhawan P. Upregulated claudin-1 expression promotes colitis-associated cancer by promoting β-catenin phosphorylation and activation in Notch/p-AKT-dependent manner. Oncogene 2019; 38:5321-5337. [PMID: 30971761 PMCID: PMC6597297 DOI: 10.1038/s41388-019-0795-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 12/21/2018] [Accepted: 01/31/2019] [Indexed: 12/18/2022]
Abstract
In IBD patients, integration between a hyper-activated immune system and epithelial cell plasticity underlies colon cancer development. However, molecular regulation of such a circuity remains undefined. Claudin-1(Cld-1), a tight-junction integral protein deregulation alters colonic epithelial cell (CEC) differentiation, and promotes colitis severity while impairing colitis-associated injury/repair. Tumorigenesis is a product of an unregulated wound healing process and therefore we postulated that upregulated Cld-1 levels render IBD patients susceptible to the colitis-associated cancer (CAC). Villin Cld-1 mice is used to carryout overexpressed studies in mice. The role of deregulated Cld-1 expression in CAC and underlying mechanism using a well-constructed study scheme and mouse models of DSS colitis/recovery and CAC. Using an inclusive investigative scheme, we here report that upregulated Cld-1 expression promotes susceptibility to the CAC and its malignancy. Increased mucosal inflammation, defective epithelial homeostasis accompanied the increased CAC in Villin-Cld1-Tg mice. We further found significantly increased levels of pro-tumorigenic M2 macrophages and β-CateninSer552 (β-CatSer552) expression in the CAC in Cld-1Tg versus WT mice. Mechanistic studies identified the role of PI3K/Akt signaling in Cld-1 dependent activation of the β-CatSer552, which, in turn, was dependent on pro-inflammatory signals. Our studies identify a critical role of Cld-1 in promoting susceptibility to CAC. Importantly, these effects of deregulated Cld-1 were not associated with altered tight junction integrity, but on its non-canonical role in regulating Notch/PI3K/Wnt/ β-CatSer552 signaling. Overall, outcome from our current studies identifies Cld-1 as potential prognostic biomarker for IBD severity and CAC, and a novel therapeutic target.
Collapse
Affiliation(s)
- Saiprasad Gowrikumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rizwan Ahmad
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Mary K Washington
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Chanjuan Shi
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Amar B Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA.,Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA. .,VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA. .,Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
22
|
Gil-Martín E, Egea J, Reiter RJ, Romero A. The emergence of melatonin in oncology: Focus on colorectal cancer. Med Res Rev 2019; 39:2239-2285. [PMID: 30950095 DOI: 10.1002/med.21582] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/04/2019] [Accepted: 03/16/2019] [Indexed: 12/17/2022]
Abstract
Within the last few decades, melatonin has increasingly emerged in clinical oncology as a naturally occurring bioactive molecule with substantial anticancer properties and a pharmacological profile optimal for joining the currently available pharmacopeia. In addition, extensive experimental data shows that this chronobiotic agent exerts oncostatic effects throughout all stages of tumor growth, from initial cell transformation to mitigation of malignant progression and metastasis; additionally, melatonin alleviates the side effects and improves the welfare of radio/chemotherapy-treated patients. Thus, the support of clinicians and oncologists for the use of melatonin in both the treatment and proactive prevention of cancer is gaining strength. Because of its epidemiological importance and symptomatic debut in advanced stages of difficult clinical management, colorectal cancer (CRC) is a preferential target for testing new therapies. In this regard, the development of effective forms of clinical intervention for the improvement of CRC outcome, specifically metastatic CRC, is urgent. At the same time, the need to reduce the costs of conventional anti-CRC therapy results is also imperative. In light of this status quo, the therapeutic potential of melatonin, and the direct and indirect critical processes of CRC malignancy it modulates, have aroused much interest. To illuminate the imminent future on CRC research, we focused our attention on the molecular mechanisms underlying the multiple oncostatic actions displayed by melatonin in the onset and evolution of CRC and summarized epidemiological evidence, as well as in vitro, in vivo and clinical findings that support the broadly protective potential demonstrated by melatonin.
Collapse
Affiliation(s)
- Emilio Gil-Martín
- Department of Biochemistry, Genetics and Immunology, Biomedical Research Center (CINBIO, 'Centro Singular de Investigación de Galicia'), University of Vigo, Vigo, Spain
| | - Javier Egea
- Molecular Neuroinflammation and Neuronal Plasticity Laboratory, Research Unit, Hospital Universitario Santa Cristina, Madrid, Spain.,Servicio de Farmacología Clínica, Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, Madrid, Spain.,Departamento de Farmacología y Terapéutica, Instituto-Fundación Teófilo Hernando, Universidad Autónoma de Madrid, Madrid, Spain
| | - Russel J Reiter
- Department of Cellular and Structural Biology, UT Health Science Center, San Antonio, Texas, USA
| | - Alejandro Romero
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
23
|
Yang X, Pan Q, Lu Y, Jiang X, Zhang S, Wu J. MNX1 promotes cell proliferation and activates Wnt/β-catenin signaling in colorectal cancer. Cell Biol Int 2019; 43:402-408. [PMID: 30614606 DOI: 10.1002/cbin.11096] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/03/2019] [Indexed: 02/07/2023]
Abstract
Aberrant Wnt/β-catenin signaling is a characteristic feature of colorectal cancer (CRC), therefore, understanding the underlying mechanisms of aberrant Wnt/β-catenin signaling will improve the treatment outcome of CRC. Expression of MNX1 in paired fresh CRC tissues and corresponding adjacent normal tissues were examined by qPCR and Western blotting. The levels of MNX1 in paraffin-embedded CRC specimens were detected by immunohistochemistry (IHC). The role of MNX1 in growth and proliferation of CRC cells was evaluated by MTT and colony formation assay. Luciferase reporter analysis and western blotting were carried out to explore the influence of MNX1 on Wnt/β-catenin signaling. The results showed that expression of MNX1 is markedly upregulated in CRC tissues and positively correlated with level of Ki67, and overexpression of MNX1 significantly promotes the proliferation of CRC cells. Further study showed that ectopic expression of MNX1 activates the Wnt/β-catenin signaling and upregulates the expression of c-Myc and CCND1, the downstream genes of Wnt/β-catenin signaling. Therefore, MNX1 plays an indispensable role in promoting of human CRC progression and may represent a novel therapeutic target for CRC.
Collapse
Affiliation(s)
- Xiangrong Yang
- Respiratory and digestive internal medicine, the Second People's of Longgang District, Shenzhen 518112, Guangdong, China
| | - Qimei Pan
- Guangzhou YouSheng Biotech Co. Ltd, Guangzhou 510006, Guangdong, China
| | - Yijie Lu
- Department of general surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou 215002, Jiangsu, China
| | - Xinwei Jiang
- Department of general surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou 215002, Jiangsu, China
| | - Sirong Zhang
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Jianwu Wu
- Department of general surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou 215002, Jiangsu, China
| |
Collapse
|
24
|
Datta S, Choudhury D, Das A, Mukherjee DD, Dasgupta M, Bandopadhyay S, Chakrabarti G. Autophagy inhibition with chloroquine reverts paclitaxel resistance and attenuates metastatic potential in human nonsmall lung adenocarcinoma A549 cells via ROS mediated modulation of β-catenin pathway. Apoptosis 2019; 24:414-433. [DOI: 10.1007/s10495-019-01526-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
25
|
Manilkara zapota (L.) P. Royen leaf water extract triggered apoptosis and activated caspase-dependent pathway in HT-29 human colorectal cancer cell line. Biomed Pharmacother 2018; 110:748-757. [PMID: 30554113 DOI: 10.1016/j.biopha.2018.12.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 12/05/2018] [Accepted: 12/05/2018] [Indexed: 01/08/2023] Open
Abstract
Manilkara zapota (L.) P. Royen (Family: Sapotaceae), commonly called as sapodilla, has been applied as traditional folk medicine for diarrhea and pulmonary infections. Conventional therapy in colorectal cancer is not likely effective due to undesirable outcomes. The anti-colon cancer properties of Manilkara zapota leaf water extract have yet to be investigated thus far. Therefore, our present study aimed to evaluate the ability to induce apoptosis and the underlying mechanisms of Manilkara zapota leaf water extract against human colorectal cancer (HT-29) cells. The cytotoxicity of Manilkara zapota leaf water extract was screened in different cancer cell lines using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and lactate dehydrogenase (LDH) analyses. The morphological changes in HT-29 cell lines after exposure to Manilkara zapota leaf water extract were viewed under fluorescence and inverted light microscope. The apoptotic cell was measured by Annexin V-propidium iodide staining. The caspase-3 and -8 activities were assessed by colorimetric assay. Overall analyses revealed that treatment with Manilkara zapota leaf water extract for 72 h can inhibit the viability of HT-29 cells. Incubation with Manilkara zapota leaf water extract for 24, 48, and 72 h significantly increased (p < 0.05) the total apoptotic cells compared to the control. Treatment with 21, 42, and 84 μg/mL of Manilkara zapota leaf water extract for 72 h triggered both caspase-3 and -8 activities in a concentration-dependent pattern. We also found that the catalase level in the two treatment groups (21 and 42 μg/mL) was significantly elevated after 24 h incubation. Incubation with Manilkara zapota leaf water extract for 72 h triggered the transcriptional elevation of the adenomatous polyposis coli (APC), glycogen synthase kinase 3β (GSK3β), AXIN1, and casein kinase 1 (CK1). The β-catenin mRNA levels were reduced accordingly when the concentration of the Manilkara zapota leaf water extract was increased. Our results suggested that Manilkara zapota leaf water extract offer great potential against colorectal cancer through modulation of Wnt/β-catenin signaling pathway, caspase-dependent pathway, and antioxidant enzyme.
Collapse
|
26
|
Wang Y, Hong D, Qian Y, Tu X, Wang K, Yang X, Shao S, Kong X, Lou Z, Jin L. Lupeol inhibits growth and migration in two human colorectal cancer cell lines by suppression of Wnt-β-catenin pathway. Onco Targets Ther 2018; 11:7987-7999. [PMID: 30519040 PMCID: PMC6235339 DOI: 10.2147/ott.s183925] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Lupeol, a triterpene isolated from various herbal plants, possesses an anti-inflammatory function and has been proposed as a candidate for anticancer agents. The purpose of this research was to investigate the effect of lupeol on the viability, apoptosis, cell-cycle distribution, and migration of colorectal cancer cell lines and its molecular mechanism. Methods Lupeol was assessed for its anticancer effect using two human colorectal cancer cell lines: SW480 and HCT116. These cells were treated with lupeol, and their viability, apoptosis, migration, and cycle distribution were detected by CCK8, flow cytometry, and the transwell method. Quantitative PCR, Western blot, and immunofluorescence were applied to detect the expressions of CTNNB1, TCF4, cMYC, CCND1, CLDN1, and CCNA2. Results Lupeol suppressed cell viability and migration and induced cellular apoptosis of both cell lines, with increased p53 and decreased Bcl2 protein levels (P<0.05). Cell cycles of both lupeol-treated cell lines were arrested in the S phase (P<0.05). Quantitative PCR and Western blot analyses showed significantly reduced expressions of CTNNB1, TCF4, and downstream genes of the Wnt–β-catenin pathway, including the cell-cycle-regulated genes of cMYC and CCND1 of both cell lines upon lupeol treatment (P<0.05). mRNA and protein levels of CLDN1 decreased in HCT116 cells, plus the expression of CCNA2 mRNA and protein decreased in SW480 cells (P<0.05). Immunofluorescence analysis confirmed decreased expression of Wnt–β-catenin signaling. Conclusion Our findings indicate that lupeol effectively inhibits proliferation and migration and induces apoptosis and cell-cycle arrest of two colorectal cell lines by inactivation of the Wnt–β-catenin signaling pathway and downregulation of cMYC, CCND1, CCNA2, and CLDN1, thereby making it a promising anticancer candidate.
Collapse
Affiliation(s)
- Yihao Wang
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, People's Republic of China, ; .,School of Ophthalmology and Optometry, Wenzhou Medical University, Zhejiang, People's Republic of China
| | - Dan Hong
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, People's Republic of China, ;
| | - Yuqin Qian
- School of the first Clinical Medical Sciences, Wenzhou Medical University, Zhejiang, People's Republic of China
| | - Xuezi Tu
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, People's Republic of China, ;
| | - Keke Wang
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, People's Republic of China, ;
| | - Xianhong Yang
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, People's Republic of China, ;
| | - Sijia Shao
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, People's Republic of China, ;
| | - Xinlong Kong
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, People's Republic of China, ;
| | - Zhefeng Lou
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, People's Republic of China, ;
| | - Longjin Jin
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Zhejiang, People's Republic of China, ;
| |
Collapse
|
27
|
Peters KM, Carlson BA, Gladyshev VN, Tsuji PA. Selenoproteins in colon cancer. Free Radic Biol Med 2018; 127:14-25. [PMID: 29793041 PMCID: PMC6168369 DOI: 10.1016/j.freeradbiomed.2018.05.075] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 05/18/2018] [Accepted: 05/20/2018] [Indexed: 02/07/2023]
Abstract
Selenocysteine-containing proteins (selenoproteins) have been implicated in the regulation of various cell signaling pathways, many of which are linked to colorectal malignancies. In this in-depth excurse into the selenoprotein literature, we review possible roles for human selenoproteins in colorectal cancer, focusing on the typical hallmarks of cancer cells and their tumor-enabling characteristics. Human genome studies of single nucleotide polymorphisms in various genes coding for selenoproteins have revealed potential involvement of glutathione peroxidases, thioredoxin reductases, and other proteins. Cell culture studies with targeted down-regulation of selenoproteins and studies utilizing knockout/transgenic animal models have helped elucidate the potential roles of individual selenoproteins in this malignancy. Those selenoproteins, for which strong links to development or progression of colorectal cancer have been described, may be potential future targets for clinical interventions.
Collapse
Affiliation(s)
- Kristin M Peters
- Dept. of Biological Sciences, Towson University, 8000 York Rd, Towson, MD 21252, United States.
| | - Bradley A Carlson
- National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, United States.
| | - Vadim N Gladyshev
- Dept. of Medicine, Brigham & Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, United States.
| | - Petra A Tsuji
- Dept. of Biological Sciences, Towson University, 8000 York Rd, Towson, MD 21252, United States.
| |
Collapse
|
28
|
HPV-18 E6 Oncoprotein and Its Spliced Isoform E6*I Regulate the Wnt/β-Catenin Cell Signaling Pathway through the TCF-4 Transcriptional Factor. Int J Mol Sci 2018; 19:ijms19103153. [PMID: 30322153 PMCID: PMC6214013 DOI: 10.3390/ijms19103153] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 09/29/2018] [Accepted: 10/09/2018] [Indexed: 01/03/2023] Open
Abstract
The Wnt/β-catenin signaling pathway regulates cell proliferation and differentiation and its aberrant activation in cervical cancer has been described. Persistent infection with high risk human papillomavirus (HR-HPV) is the most important factor for the development of this neoplasia, since E6 and E7 viral oncoproteins alter cellular processes, promoting cervical cancer development. A role of HPV-16 E6 in Wnt/β-catenin signaling has been proposed, although the participation of HPV-18 E6 has not been previously studied. The aim of this work was to investigate the participation of HPV-18 E6 and E6*I, in the regulation of the Wnt/β-catenin signaling pathway. Here, we show that E6 proteins up-regulate TCF-4 transcriptional activity and promote overexpression of Wnt target genes. In addition, it was demonstrated that E6 and E6*I bind to the TCF-4 (T cell factor 4) and β-catenin, impacting TCF-4 stabilization. We found that both E6 and E6*I proteins interact with the promoter of Sp5, in vitro and in vivo. Moreover, although differences in TCF-4 transcriptional activation were found among E6 intratype variants, no changes were observed in the levels of regulated genes. Furthermore, our data support that E6 proteins cooperate with β-catenin to promote cell proliferation.
Collapse
|
29
|
Chang YY, Yu LCH, Yu IS, Jhuang YL, Huang WJ, Yang CY, Jeng YM. Deletion of cadherin-17 enhances intestinal permeability and susceptibility to intestinal tumour formation. J Pathol 2018; 246:289-299. [PMID: 30047135 DOI: 10.1002/path.5138] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/28/2018] [Accepted: 06/05/2018] [Indexed: 01/08/2023]
Abstract
Cadherin-17 is an adhesion molecule expressed specifically in intestinal epithelial cells. It is frequently underexpressed in human colorectal cancer. The physiological function of cadherin-17 and its role in tumourigenesis have not yet been determined. We used the transcription activator-like effector nuclease technique to generate a Cdh17 knockout (KO) mouse model. Intestinal tissues were analysed with histological, immunohistochemical and ultrastructural methods. Colitis was induced by oral administration of dextran sulphate sodium (DSS), and, to study effects on intestinal tumourigenesis, mice were given azoxymethane (AOM) and DSS to induce colitis-associated cancer. Cdh17 KO mice were viable and fertile. The histology of their small and large intestines was similar to that of wild-type mice. The junctional architecture of the intestinal epithelium was preserved. The loss of cadherin-17 resulted in increased permeability and susceptibility to DSS-induced colitis. The AOM/DSS model demonstrated that Cdh17 KO enhanced tumour formation and progression in the intestine. Increased nuclear translocation of Yap1, but not of β-catenin, was identified in the tumours of Cdh17 KO mice. In conclusion, cadherin-17 plays a crucial role in intestinal homeostasis by limiting the permeability of the intestinal epithelium. Cadherin-17 is also a tumour suppressor for intestinal epithelia. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Ya-Yun Chang
- Graduate Institute of Pathology, National Taiwan University, Taipei, Taiwan
| | - Linda Chia-Hui Yu
- Graduate Institute of Physiology, National Taiwan University, Taipei, Taiwan
| | - I-Shing Yu
- Laboratory Animal Centre, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Lin Jhuang
- Graduate Institute of Pathology, National Taiwan University, Taipei, Taiwan.,Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Wei-Ju Huang
- Graduate Institute of Pathology, National Taiwan University, Taipei, Taiwan.,Department of Oral Hygiene, Hsin-Sheng College of Medical Care and Management, Taoyuan, Taiwan
| | - Ching-Yao Yang
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Yung-Ming Jeng
- Graduate Institute of Pathology, National Taiwan University, Taipei, Taiwan.,Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
30
|
Wang S, Yan S, Zhu S, Zhao Y, Yan J, Xiao Z, Bi J, Qiu J, Zhang D, Hong Z, Zhang L, Huang C, Li T, Liang L, Liao W, Jiao H, Ding Y, Ye Y. FOXF1 Induces Epithelial-Mesenchymal Transition in Colorectal Cancer Metastasis by Transcriptionally Activating SNAI1. Neoplasia 2018; 20:996-1007. [PMID: 30189360 PMCID: PMC6134153 DOI: 10.1016/j.neo.2018.08.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/07/2018] [Accepted: 08/07/2018] [Indexed: 02/07/2023] Open
Abstract
Forkhead Box F1 (FOXF1) has been recently implicated in cancer progression and metastasis of lung cancer and breast cancer. However, the biological functions and underlying mechanisms of FOXF1 in the regulation of the progression of colorectal cancer (CRC) are largely unknown. We showed that FOXF1 was up-regulated in 93 paraffin-embedded archived human CRC tissue, and both high expression and nuclear location of FOXF1 were significantly associated with the aggressive characteristics and poorer survival of CRC patients. The GSEA analysis showed that the higher level of FOXF1 was positively associated with an enrichment of EMT gene signatures, and exogenous overexpression of FOXF1 induced EMT by transcriptionally activating SNAI1. Exogenous overexpression FOXF1 functionally promoted invasion and metastasis features of CRC cells, and inhibition of SNAI1 attenuates the invasive phenotype and metastatic potential of FOXF1-overexpressing CRC cells. Furthermore, the results of the tissue chip showed that the expression of FOXF1 was positively correlated with SNAI1 in CRC tissues chip. These results suggested that FOXF1 plays a critical role in CRC metastasis by inducing EMT via transcriptional activation of SNAI1, highlighting a potential new therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Shuyang Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Shanshan Yan
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Shaowei Zhu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Yali Zhao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Junyu Yan
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Zhiyuan Xiao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Jiaxin Bi
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Junfeng Qiu
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Dan Zhang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Zexuan Hong
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Lingjie Zhang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Chengmei Huang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Tingting Li
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Li Liang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Wenting Liao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China
| | - Hongli Jiao
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China.
| | - Yanqing Ding
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China.
| | - Yaping Ye
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China; Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou, China.
| |
Collapse
|
31
|
Yang D, Zhang X, Zhang W, Rengarajan T. Vicenin-2 inhibits Wnt/β-catenin signaling and induces apoptosis in HT-29 human colon cancer cell line. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:1303-1310. [PMID: 29849451 PMCID: PMC5965372 DOI: 10.2147/dddt.s149307] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Colorectal cancer (CRC) is among highest prevailing cancers in the whole world, especially in western countries. For a diverse of reasons, patients prefer naturally occurring dietary substances over synthetic agents to prevent cancer. Vicenin-2 is largely available in a medicinal plant Ocimum sanctum and is an apigenin form, 6,8-di-C-glucoside, which has been reported to have a range of pharmacological values which includes antioxidant, hepatoprotective, anti-inflammatory and anti-cancer. This study was aimed to analyze the anti-proliferative effect of Vicenin-2 on human colon cancer cells via the Wnt/β-catenin signaling inhibition. Methods MTT assay was used to assess the cell viability at different concentrations and time point. Vicenin-2 at a concentration of 50 µM (IC50) decreased the phosphorylated (inactive) glycogen synthase kinase-3β, cyclin D1, and non-p-β-catenin expressions in HT-29 cells, which were evidenced through western blot analysis. Results Further, Vincenin-2 reduced the T-cell factor (TCF) / Leukocyte erythroid factor (LEF) reporter activity in HT-29 cells. Vicenin-2 also promoted substantial cell cycle arrest at the G2M phase of HT-29 cells, as well induced apoptosis in HT-29 cells, as revealed through flow cytometric analysis. Furthermore, immunoblot analysis showed that Vicenin-2 treatment enhanced the expression of Cytochrome C, Bax and caspase-3 whereas suppressed the Bcl-2 expression. Conclusion Together, these results revealed that Vicenin-2 can act as a potent inhibitor of HT-29 cell proliferation and can be used as an agent against CRC.
Collapse
Affiliation(s)
- Dong Yang
- Department of Anorectal Surgery, The First People's Hospital of Lianyungang, Lianyungang, China
| | - Xiling Zhang
- Department of Anorectal Surgery, Shaanxi Provincial People's Hospital, Shaanxi, Xi'an, China
| | - Wencun Zhang
- Department of Anorectal Surgery, The Fourth Hospital of Yu Lin City, Shaanxi Province, China
| | | |
Collapse
|
32
|
Novel Regulatory Roles of Wnt1 in Infection-Associated Colorectal Cancer. Neoplasia 2018; 20:499-509. [PMID: 29626750 PMCID: PMC5915993 DOI: 10.1016/j.neo.2018.03.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/28/2018] [Accepted: 03/01/2018] [Indexed: 12/11/2022] Open
Abstract
Salmonella infection is a major public health concern, and colonization in humans can be chronic and increases the risk of cancers. Wnt signaling is a key pathway for intestinal renewal and development, inflammation, and tumorigenesis. In the current study, we report a novel role of Wnt1 in infection and colon cancer using cell culture models, a Salmonella-colitis colon cancer model, and human samples. In contrast to the bacteria-induced increases in Wnt2 and Wnt11, Salmonella colonization significantly reduced the level of Wnt1 in intestinal epithelial cells in vivo and in vitro. The bacterial AvrA protein is known to activate the canonical Wnt pathway. Wnt1 expression level was downregulated by AvrA-expressing Salmonella but stabilized by AvrA-deficient Salmonella in the intestine of Salmonella-colitis mice. In a chronic Salmonella-infected cancer model, the Wnt1 protein level was decreased in the AvrA+ infected group. Thus, we further assessed the functional role of Wnt1 downregulation in the inflammatory response and colorectal cancer (CRC) progression. Moreover, downregulation of Wnt1 by the Crispr-Cas9 method promoted cancer cell invasion and migration. Interestingly, we found that Wnt1 was downregulated in human CRC tissue, and Wnt1 downregulation may be correlated with cancer progression. Our study provides insights into mechanisms by which enteric bacteria regulate Wnt1 expression and potentially contribute to infection-associated colon cancer.
Collapse
|
33
|
Fan X, Zhao J, Ren F, Wang Y, Feng Y, Ding L, Zhao L, Shang Y, Li J, Ni J, Jia B, Liu Y, Chang Z. Dimerization of p15RS mediated by a leucine zipper-like motif is critical for its inhibitory role on Wnt signaling. J Biol Chem 2018; 293:7618-7628. [PMID: 29618509 DOI: 10.1074/jbc.ra118.001969] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 03/27/2018] [Indexed: 01/31/2023] Open
Abstract
We previously demonstrated that p15RS, a newly discovered tumor suppressor, inhibits Wnt/β-catenin signaling by interrupting the formation of β-catenin·TCF4 complex. However, it remains unclear how p15RS helps exert such an inhibitory effect on Wnt signaling based on its molecular structure. In this study, we reported that dimerization of p15RS is required for its inhibition on the transcription regulation of Wnt-targeted genes. We found that p15RS forms a dimer through a highly conserved leucine zipper-like motif in the coiled-coil terminus domain. In particular, residues Leu-248 and Leu-255 were identified as being responsible for p15RS dimerization, as mutation of these two leucines into prolines disrupted the homodimer formation of p15RS and weakened its suppression of Wnt signaling. Functional studies further confirmed that mutations of p15RS at these residues results in diminishment of its inhibition on cell proliferation and tumor formation. We therefore concluded that dimerization of p15RS governed by the leucine zipper-like motif is critical for its inhibition of Wnt/β-catenin signaling and tumorigenesis.
Collapse
Affiliation(s)
- Xuanzi Fan
- From the State Key Laboratory of Membrane Biology, School of Medicine and.,the School of Life Sciences, Tsinghua University, Beijing 100084
| | - Juan Zhao
- From the State Key Laboratory of Membrane Biology, School of Medicine and
| | - Fangli Ren
- From the State Key Laboratory of Membrane Biology, School of Medicine and
| | - Yinyin Wang
- From the State Key Laboratory of Membrane Biology, School of Medicine and
| | - Yarui Feng
- From the State Key Laboratory of Membrane Biology, School of Medicine and
| | - Lidan Ding
- From the State Key Laboratory of Membrane Biology, School of Medicine and
| | - Linpeng Zhao
- the Department of Cell Biology, College of Life Sciences, Key Laboratory for Cell Proliferation and Regulation Biology of Ministry of Education, Beijing Normal University, Beijing 100875
| | - Yu Shang
- the Department of Cell Biology, College of Life Sciences, Key Laboratory for Cell Proliferation and Regulation Biology of Ministry of Education, Beijing Normal University, Beijing 100875
| | - Jun Li
- the Institute of Immunology, PLA, The Third Military Medical University, Chongqing 400038, and
| | - Jianquan Ni
- From the State Key Laboratory of Membrane Biology, School of Medicine and
| | - Baoqing Jia
- the Department of General Surgery/Pathology, Chinese PLA General Hospital, Beijing 100853, China
| | - Yule Liu
- the School of Life Sciences, Tsinghua University, Beijing 100084
| | - Zhijie Chang
- From the State Key Laboratory of Membrane Biology, School of Medicine and
| |
Collapse
|
34
|
Sharma M, Castro-Piedras I, Simmons GE, Pruitt K. Dishevelled: A masterful conductor of complex Wnt signals. Cell Signal 2018; 47:52-64. [PMID: 29559363 DOI: 10.1016/j.cellsig.2018.03.004] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 03/14/2018] [Accepted: 03/14/2018] [Indexed: 12/21/2022]
Abstract
The Dishevelled gene was first identified in Drosophila mutants with disoriented hair and bristle polarity [1-3]. The Dsh gene (Dsh/Dvl, in Drosophila and vertebrates respectively) gained popularity when it was discovered that it plays a key role in segment polarity during early embryonic development in Drosophila [4]. Subsequently, the vertebrate homolog of Dishevelled genes were identified in Xenopus (Xdsh), mice (Dvl1, Dvl2, Dvl3), and in humans (DVL1, DVL2, DVL3) [5-10]. Dishevelled functions as a principal component of Wnt signaling pathway and governs several cellular processes including cell proliferation, survival, migration, differentiation, polarity and stem cell renewal. This review will revisit seminal discoveries and also summarize recent advances in characterizing the role of Dishevelled in both normal and pathophysiological settings.
Collapse
Affiliation(s)
- Monica Sharma
- Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Isabel Castro-Piedras
- Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Glenn E Simmons
- Department of Biomedical Sciences, University of Minnesota, School of Medicine, Duluth, MN, USA
| | - Kevin Pruitt
- Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
| |
Collapse
|
35
|
Li Q, Yang T, Li D, Ding F, Bai G, Wang W, Sun H. Knockdown of aquaporin-5 sensitizes colorectal cancer cells to 5-fluorouracil via inhibition of the Wnt-β-catenin signaling pathway. Biochem Cell Biol 2018; 96:572-579. [PMID: 29390193 DOI: 10.1139/bcb-2017-0162] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Aquaporin-5 (AQP5), a water channel protein, has been reported to possess oncogenic potential in multiple types of malignancies, including colorectal cancer (CRC). However, its effect on the chemosensitivity of CRC cells remains elusive. Hence, this study investigated the effect of AQP5 silencing in CRC cells on 5-fluorouracil (5-FU) sensitivity and attempted to elucidate the underlying mechanisms. A short hairpin RNA construct targeting AQP5 was transfected into HCT116 or HT29 cells to generate stable AQP5-silenced cell lines. The effects of AQP5 knockdown on cell viability, apoptosis, tumor growth, and 5-FU chemoresistance were evaluated. Relative protein levels of Wnt-β-catenin pathway effectors were also measured. The results showed that silencing of AQP5 increased the chemosensitivity of CRC cells to 5-FU, facilitated 5-FU-mediated apoptosis, suppressed tumor growth, and reduced 5-FU chemoresistance in vivo. Furthermore, the effect of AQP5 on 5-FU chemosensitivity was mediated by the Wnt-β-catenin pathway. Silencing of AQP5 inhibited Wnt-β-catenin signaling, whereas overexpression of the degradation-resistant mutant of β-catenin (S33Y) reversed apoptosis induced by AQP5 silencing. Taken together, these results suggest that AQP5 silencing enhances the sensitivity of CRC cells to 5-FU, and the underlying mechanism is related to inhibition of the Wnt-β-catenin pathway. AQP5 could be a useful therapeutic target for CRC treatment.
Collapse
Affiliation(s)
- Qing Li
- a College of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China; Department of Internal Medicine, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, People's Republic of China
| | - Tao Yang
- b Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, People's Republic of China
| | - Dongsheng Li
- b Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, People's Republic of China
| | - Feng Ding
- b Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, People's Republic of China
| | - Guang Bai
- b Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, People's Republic of China
| | - Wei Wang
- b Department of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, People's Republic of China
| | - Hongzhi Sun
- c Department of Pathophysiology, College of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, People's Republic of China
| |
Collapse
|
36
|
Jiao S, Guan J, Chen M, Wang W, Li C, Wang Y, Cheng Y, Zhou Z. Targeting IRF3 as a YAP agonist therapy against gastric cancer. J Exp Med 2018; 215:699-718. [PMID: 29339449 PMCID: PMC5789414 DOI: 10.1084/jem.20171116] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 11/07/2017] [Accepted: 12/12/2017] [Indexed: 11/25/2022] Open
Abstract
Jiao et al. show that the key player of antiviral immunity IRF3 binds to and promotes the transactivation of the YAP–TEAD4 complex to coregulate transcription of Hippo pathway target genes and that therapeutic targeting of IRF3 suppresses YAP-driven gastric cancer. The Hippo pathway plays a vital role in tissue homeostasis and tumorigenesis. The transcription factor IRF3 is essential for innate antiviral immunity. In this study, we discovered IRF3 as an agonist of Yes-associated protein (YAP). The expression of IRF3 is positively correlated with that of YAP and its target genes in gastric cancer; the expression of both IRF3 and YAP is up-regulated and prognosticates patient survival. IRF3 interacts with both YAP and TEAD4 in the nucleus to enhance their interaction, promoting nuclear translocation and activation of YAP. IRF3 and YAP–TEAD4 are associated genome-wide to cobind and coregulate many target genes of the Hippo pathway. Overexpression of active IRF3 increased, but depletion of IRF3 reduced, the occupancy of YAP on the target genes. Knockdown or pharmacological targeting of IRF3 by Amlexanox, a drug used clinically for antiinflammatory treatment, inhibits gastric tumor growth in a YAP-dependent manner. Collectively, our study identifies IRF3 as a positive regulator for YAP, highlighting a new therapeutic target against YAP-driven cancers.
Collapse
Affiliation(s)
- Shi Jiao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jingmin Guan
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Min Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wenjia Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chuanchuan Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yugong Wang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yunfeng Cheng
- Department of Hematology and Institute of Clinical Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhaocai Zhou
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.,State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China .,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
37
|
Fiore D, Ramesh P, Proto MC, Piscopo C, Franceschelli S, Anzelmo S, Medema JP, Bifulco M, Gazzerro P. Rimonabant Kills Colon Cancer Stem Cells without Inducing Toxicity in Normal Colon Organoids. Front Pharmacol 2018; 8:949. [PMID: 29354056 PMCID: PMC5758598 DOI: 10.3389/fphar.2017.00949] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 12/14/2017] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer (CRC), like other tumor types, is a highly heterogeneous disease. Within the tumor bulk, intra-tumoral heterogeneity is also ascribable to Cancer Stem Cells (CSCs) subpopulation, characterized by high chemoresistance and the unique ability to retain tumorigenic potential, thus associated to tumor recurrence. High dynamic plasticity of CSCs, makes the development of winning therapeutic strategies even more complex to completely eradicate tumor fuel. Rimonabant, originally synthesized as antagonist/inverse agonist of Cannabinoid Receptor 1, is able to inactivate Wnt signaling, both in vitro and in vivo, in CRC models, through inhibition of p300-histone acetyltransferase activity. Since Wnt/β-Catenin pathway is the main player underlying CSCs dynamic, this finding candidates Rimonabant as potential modulator of cancer stemness, in CRC. In this work, using established 3D cultures of primary colon CSCs, taking into account the tumor heterogeneity through monitoring of Wnt activity, we demonstrated that Rimonabant was able to reduces both tumor differentiated cells and colon CSCs proliferation and to control their survival in long term cultures. Interestingly, in ex vivo model of wild type human organoids, retaining both architecture and heterogeneity of original tissue, Rimonabant showed no toxicity against cells from healthy colon epithelium, suggesting its potential selectivity toward cancer cells. Overall, results from this work provided new insights on anti-tumor efficacy of Rimonabant, strongly suggesting that it could be a novel lead compound for CRC treatment.
Collapse
Affiliation(s)
- Donatella Fiore
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | - Prashanthi Ramesh
- Laboratory of Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Academisch Medisch Centrum, University of Amsterdam, Amsterdam, Netherlands
| | - Maria C Proto
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | - Chiara Piscopo
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | | | - Serena Anzelmo
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | - Jan P Medema
- Laboratory of Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Academisch Medisch Centrum, University of Amsterdam, Amsterdam, Netherlands
| | - Maurizio Bifulco
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy
| | | |
Collapse
|