51
|
Campbell K. Contribution of epithelial-mesenchymal transitions to organogenesis and cancer metastasis. Curr Opin Cell Biol 2018; 55:30-35. [PMID: 30006053 PMCID: PMC6284102 DOI: 10.1016/j.ceb.2018.06.008] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 05/10/2018] [Accepted: 06/14/2018] [Indexed: 02/06/2023]
Abstract
The epithelial-to-mesenchymal transition (EMT) plays crucial roles during development, and inappropriate activation of EMTs are associated with tumor progression and promoting metastasis. In recent years, increasing studies have identified developmental contexts where cells undergo an EMT and transition to a partial-state, downregulating just a subset of epithelial characteristics and increasing only some mesenchymal traits, such as invasive motility. In parallel, recent studies have shown that EMTs are rarely fully activated in tumor cells, generating a diverse array of transition states. As our appreciation of the full spectrum of intermediate phenotypes and the huge diversity in underlying mechanisms grows, cross-disciplinary collaborations investigating developmental-EMTs and cancer-EMTs will be fundamental in order to achieve a full mechanistic understanding of this complex cell process.
Collapse
Affiliation(s)
- Kyra Campbell
- Bateson Centre, Firth Court, University of Sheffield, Western Bank, Sheffield, UK; Department of Biomedical Science, Firth Court, University of Sheffield, Western Bank, Sheffield, UK.
| |
Collapse
|
52
|
Pal M, Bhattacharya S, Kalyan G, Hazra S. Cadherin profiling for therapeutic interventions in Epithelial Mesenchymal Transition (EMT) and tumorigenesis. Exp Cell Res 2018; 368:137-146. [DOI: 10.1016/j.yexcr.2018.04.014] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 03/15/2018] [Accepted: 04/13/2018] [Indexed: 12/14/2022]
|
53
|
Kobayashi W, Ozawa M. The epithelial-mesenchymal transition induced by transcription factor LEF-1 is independent of β-catenin. Biochem Biophys Rep 2018; 15:13-18. [PMID: 29998192 PMCID: PMC6038150 DOI: 10.1016/j.bbrep.2018.06.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 06/04/2018] [Accepted: 06/07/2018] [Indexed: 11/02/2022] Open
Abstract
Transcription factor lymphoid-enhancer-binding factor 1 (LEF-1) is a key molecule in the Wnt/β-catenin signaling pathway. Slug is one of the Wnt/β-catenin target genes and can induce epithelial-mesenchymal transition (EMT). Previously, we have shown that not only wild-type LEF-1 but also LEF-1 lacking the amino-terminal β-catenin-binding region can induce EMT, suggesting that LEF-1 acts independently of β-catenin. Because it has been reported that LEF-1 interacts with β-catenin outside the amino-terminal domain, namely, in the middle part of the molecule, the possible participation of β-catenin has not been formally ruled out. To determine the involvement of β-catenin in the LEF-1-induced EMT, we produced MDCK cells with a deletion of the β-catenin gene and then expressed LEF-1 in the cells. We found that LEF-1 induced EMT in those cells. In the absence of β-catenin, γ-catenin has been shown to take over the role of β-catenin. To examine this possibility, we first established MDCK cells with a double knockout of β-catenin and γ-catenin genes and then expressed LEF-1 in these cells. We found that LEF-1 can induce EMT in these cells; therefore, we conclude that neither β-catenin nor γ-catenin expression is necessary for the LEF-1-mediated induction of EMT.
Collapse
Affiliation(s)
- Wakako Kobayashi
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Masayuki Ozawa
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| |
Collapse
|
54
|
Divya T, Velavan B, Sudhandiran G. Regulation of Transforming Growth Factor-β/Smad-mediated Epithelial-Mesenchymal Transition by Celastrol Provides Protection against Bleomycin-induced Pulmonary Fibrosis. Basic Clin Pharmacol Toxicol 2018; 123:122-129. [PMID: 29394529 DOI: 10.1111/bcpt.12975] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/24/2018] [Indexed: 12/21/2022]
Abstract
The respiratory disease pulmonary fibrosis (PF), which is characterized by scar formation throughout the lung, imposes a serious health burden. No effective drug without side effects has been proven to prevent this fatal lung disease. In this context, this study was undertaken to elucidate the protective effect of celastrol, a quinine methide pentacyclic triterpenoid from a Chinese medicinal plant 'thunder god vine' against bleomycin (BLM)-induced PF. We also attempted to study how the cytokine transforming growth factor-β (TGF-β) stimulates fibrosis through the induction of epithelial-mesenchymal transition (EMT) and the role of celastrol in regulating EMT. TGF-β (5 ng/ml) was administered to human alveolar epithelial adenocarcinoma A549 cells to induce fibrotic response in cells. Induction of EMT was analysed in cells through morphological analysis and expression of epithelial and mesenchymal markers by Western blotting. Bleomycin at a concentration of 3 U/Kg b.w was used to induce fibrosis in adult male rat lungs. Celastrol (5 mg/kg b.w) was given to rats twice a week after BLM administration for a period of 28 days. Western blot and immunofluorescence analyses were performed with lung tissue sample to find out the potential of celastrol in regulating EMT during the progression of fibrosis. TGF-β induces EMT in A549 cells as demonstrated by changes in epithelial cell morphology and expression of epithelial and mesenchymal marker proteins. The expressions of epithelial marker proteins E-cadherin and claudin were found to be reduced in the BLM-induced group of rats. Expression of mesenchymal markers, such as N-cadherin, snail, slug, vimentin and β-catenin, was enhanced in BLM-induced rat lungs. Celastrol reverts these cellular changes in rat lungs, and it was found that celastrol regulates EMT through the inhibition of heat shock protein 90 (HSP 90). Together, the results indicate that EMT is a crucial phenomenon for the progression of fibrosis, and celastrol provides protection against PF through the regulation of EMT.
Collapse
Affiliation(s)
- Thomas Divya
- Cell Biology Laboratory, Department of Biochemistry, University of Madras, Chennai, India
| | | | - Ganapasam Sudhandiran
- Cell Biology Laboratory, Department of Biochemistry, University of Madras, Chennai, India
| |
Collapse
|
55
|
Simon-Tillaux N, Hertig A. Snail and kidney fibrosis. Nephrol Dial Transplant 2018; 32:224-233. [PMID: 28186539 DOI: 10.1093/ndt/gfw333] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 08/04/2016] [Indexed: 12/13/2022] Open
Abstract
Snail family zinc finger 1 (SNAI1) is a transcription factor expressed during renal embryogenesis, and re-expressed in various settings of acute kidney injury (AKI). Subjected to tight regulation, SNAI1 controls major biological processes responsible for renal fibrogenesis, including mesenchymal reprogramming of tubular epithelial cells, shutdown of fatty acid metabolism, cell cycle arrest and inflammation of the microenvironment surrounding tubular epithelial cells. The present review describes in detail the interactions of SNAI1 with AKI-associated signalling pathways. We also discuss how this central factor has been iteratively (and promisingly) targeted in a number of animal models in order to prevent or slow down renal fibrogenesis.
Collapse
Affiliation(s)
- Noémie Simon-Tillaux
- French National Institute of Health and Medical Research (INSERM), UMR_S1155, Remodeling and Repair of Renal Tissue, Hôpital Tenon, Paris, France
| | - Alexandre Hertig
- French National Institute of Health and Medical Research (INSERM), UMR_S1155, Remodeling and Repair of Renal Tissue, Hôpital Tenon, Paris, France.,Sorbonne Universités, UPMC Paris 06, UMR S_1155, Paris, France
| |
Collapse
|
56
|
Yang R, Liu W, Miao L, Yang X, Fu J, Dou B, Cai A, Zong X, Tan C, Chen H, Wang X. Induction of VEGFA and Snail-1 by meningitic Escherichia coli mediates disruption of the blood-brain barrier. Oncotarget 2018; 7:63839-63855. [PMID: 27588479 PMCID: PMC5325408 DOI: 10.18632/oncotarget.11696] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 08/24/2016] [Indexed: 12/20/2022] Open
Abstract
Escherichia coli is the most common Gram-negative bacterium that possesses the ability to cause neonatal meningitis, which develops as circulating bacteria penetrate the blood-brain barrier (BBB). However, whether meningitic E. coli could induce disruption of the BBB and the underlying mechanisms are poorly understood. Our current work highlight for the first time the participation of VEGFA and Snail-1, as well as the potential mechanisms, in meningitic E. coli induced disruption of the BBB. Here, we characterized a meningitis-causing E. coli PCN033, and demonstrated that PCN033 invasion could increase the BBB permeability through downregulating and remodeling the tight junction proteins (TJ proteins). This process required the PCN033 infection-induced upregulation of VEGFA and Snail-1, which involves the activation of TLR2-MAPK-ERK1/2 signaling cascade. Moreover, production of proinflammatory cytokines and chemokines in response to infection also promoted the upregulation of VEGFA and Snail-1, therefore further mediating the BBB disruption. Our observations reported here directly support the involvement of VEGFA and Snail-1 in meningitic E. coli induced BBB disruption, and VEGFA and Snail-1 would therefore represent the essential host targets for future prevention of clinical E. coli meningitis.
Collapse
Affiliation(s)
- Ruicheng Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Wentong Liu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Ling Miao
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Xiaopei Yang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Jiyang Fu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Beibei Dou
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Aoling Cai
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Xin Zong
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Chen Tan
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei 430070, China.,Key Laboratory of development of veterinary diagnostic products of Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei 430070, China.,Key Laboratory of development of veterinary diagnostic products of Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Xiangru Wang
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China.,The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan, Hubei 430070, China.,Key Laboratory of development of veterinary diagnostic products of Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| |
Collapse
|
57
|
Tom CTMB, Crellin JE, Motiwala HF, Stone MB, Davda D, Walker W, Kuo YH, Hernandez JL, Labby KJ, Gomez-Rodriguez L, Jenkins PM, Veatch SL, Martin BR. Chemoselective ratiometric imaging of protein S-sulfenylation. Chem Commun (Camb) 2018; 53:7385-7388. [PMID: 28613292 DOI: 10.1039/c7cc02285a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Here we report a ratiometric fluorescent probe for chemoselective conjugation to sulfenic acids in living cells. Our approach couples an α-fluoro-substituted dimedone to an aminonaphthalene fluorophore (F-DiNap), which upon sulfenic acid conjugation is locked as the 1,3-diketone, changing the fluorophore excitation. F-DiNap reacts with S-sulfenylated proteins at equivalent rates to current probes, but the α-fluorine substitution blocks side-reactions with biological aldehydes.
Collapse
Affiliation(s)
- Christopher T M B Tom
- Department of Chemistry and Program in Chemical Biology, University of Michigan, 930 N. University Ave., Ann Arbor, MI, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Kim KS, Kim J, Oh N, Kim MY, Park KS. ELK3-GATA3 axis modulates MDA-MB-231 metastasis by regulating cell-cell adhesion-related genes. Biochem Biophys Res Commun 2018; 498:509-515. [PMID: 29510139 DOI: 10.1016/j.bbrc.2018.03.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 03/02/2018] [Indexed: 10/25/2022]
Abstract
GATA3 is a master regulator that drives mammary epithelial cell differentiation, and the suppression of GATA3 expression is associated with the development of aggressive breast cancer. However, the mechanism through which GATA3 loss drives cancer development is poorly understood. Previously, we reported that ELK3 suppression in MDA-MB-231 (ELK3 KD) resulted in the reprogramming of these cells from a basal to luminal subtype, which was associated with the induction of GATA3 expression, and that the ELK3-GATA3 axis orchestrated the metastatic characteristics of MDA-MB-231. Here, we show that GATA3 suppression in ELK3 knockdown MDA-MB-231 cells (ELK3/GATA3 DKD) restores the metastatic ability comparably to that of control MDA-MB-231 cells, even though the epithelial cell morphology and TGF-β signaling of ELK3 KD are not recovered in ELK3/GATA3 DKD. The expression of E-cadherin and tight junctional proteins, including occludin, claudin and ZO-1, which is activated in ELK3 KD, is suppressed in ELK3/GATA3 DKD. These results reveal the possibility that the ELK3-GATA3 axis determines the metastatic characteristics of MDA-MB-231 by regulating the expression of cell-cell adhesion factors.
Collapse
Affiliation(s)
- Kwang-Soo Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
| | - Jiewan Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
| | - Nuri Oh
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
| | - Mi-Young Kim
- Digestive Disease Center, CHA Bundang Medical Center, CHA University, Seongnam, South Korea.
| | - Kyung-Soon Park
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea.
| |
Collapse
|
59
|
Looi K, Buckley AG, Rigby PJ, Garratt LW, Iosifidis T, Zosky GR, Larcombe AN, Lannigan FJ, Ling KM, Martinovich KM, Kicic-Starcevich E, Shaw NC, Sutanto EN, Knight DA, Kicic A, Stick SM. Effects of human rhinovirus on epithelial barrier integrity and function in children with asthma. Clin Exp Allergy 2018; 48:513-524. [PMID: 29350877 DOI: 10.1111/cea.13097] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/13/2017] [Accepted: 11/21/2017] [Indexed: 11/29/2022]
Abstract
BACKGROUND Bronchial epithelial tight junctions (TJ) have been extensively assessed in healthy airway epithelium. However, no studies have yet assessed the effect of human rhinovirus (HRV) infection on the expression and resultant barrier function in epithelial tight junctions (TJ) in childhood asthma. OBJECTIVES To investigate the impact of HRV infection on airway epithelial TJ expression and barrier function in airway epithelial cells (AECs) of children with and without asthma. Furthermore, to test the hypothesis that barrier integrity and function is compromised to a greater extent by HRV in AECs from asthmatic children. METHODS Primary AECs were obtained from children with and without asthma, differentiated into air-liquid interface (ALI) cultures and infected with rhinovirus. Expression of claudin-1, occludin and zonula occluden-1 (ZO-1) was assessed via qPCR, immunocytochemistry (ICC), in-cell western (ICW) and confocal microscopy. Barrier function was assessed by transepithelial electrical resistance (TER; RT ) and permeability to fluorescent dextran. RESULTS Basal TJ gene expression of claudin-1 and occludin was significantly upregulated in asthmatic children compared to non-asthmatics; however, no difference was seen with ZO-1. Interestingly, claudin-1, occludin and ZO-1 protein expression was significantly reduced in AEC of asthmatic children compared to non-asthmatic controls suggesting possible post-transcriptional inherent differences. HRV infection resulted in a transient dissociation of TJ and airway barrier integrity in non-asthmatic children. Although similar dissociation of TJ was observed in asthmatic children, a significant and sustained reduction in TJ expression concurrent with both a significant decrease in TER and an increase in permeability in asthmatic children was observed. CONCLUSION This study demonstrates novel intrinsic differences in TJ gene and protein expression between AEC of children with and without asthma. Furthermore, it correlates directly the relationship between HRV infection and the resultant dissociation of epithelial TJ that causes a continued altered barrier function in children with asthma.
Collapse
Affiliation(s)
- K Looi
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - A G Buckley
- Centre for Microscopy, Characterisation and Analysis (CMCA), University of Western Australia, Crawley, WA, Australia
| | - P J Rigby
- Centre for Microscopy, Characterisation and Analysis (CMCA), University of Western Australia, Crawley, WA, Australia
| | - L W Garratt
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - T Iosifidis
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, University of Western Australia, Nedlands, WA, Australia
| | - G R Zosky
- School of Medicine, Faculty of Health, University of Tasmania, Hohart, TAS, Australia
| | - A N Larcombe
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Occupation and Environment, School of Public Health, Curtin University, Perth, WA, Australia
| | - F J Lannigan
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,School of Medicine, Notre Dame University, Fremantle, WA, Australia
| | - K-M Ling
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - K M Martinovich
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - E Kicic-Starcevich
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia
| | - N C Shaw
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia
| | - E N Sutanto
- Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia
| | - D A Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, Australia.,Priority Research Centre for Asthma and Respiratory Disease, Hunter Medical Research Institute, Newcastle, NSW, Australia.,Department of Anaesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - A Kicic
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, University of Western Australia, Nedlands, WA, Australia.,Occupation and Environment, School of Public Health, Curtin University, Perth, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia
| | - S M Stick
- School of Paediatrics and Child Health, University of Western Australia, Nedlands, WA, Australia.,Telethon Kids Institute, University of Western Australia, Subiaco, WA, Australia.,Centre for Cell Therapy and Regenerative Medicine, University of Western Australia, Nedlands, WA, Australia.,Department of Respiratory Medicine, Princess Margaret Hospital for Children, Perth, WA, Australia
| |
Collapse
|
60
|
Lo UG, Lee CF, Lee MS, Hsieh JT. The Role and Mechanism of Epithelial-to-Mesenchymal Transition in Prostate Cancer Progression. Int J Mol Sci 2017; 18:ijms18102079. [PMID: 28973968 PMCID: PMC5666761 DOI: 10.3390/ijms18102079] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 09/21/2017] [Accepted: 09/27/2017] [Indexed: 12/12/2022] Open
Abstract
In prostate cancer (PCa), similar to many other cancers, distant organ metastasis symbolizes the beginning of the end disease, which eventually leads to cancer death. Many mechanisms have been identified in this process that can be rationalized into targeted therapy. Among them, epithelial-to-mesenchymal transition (EMT) is originally characterized as a critical step for cell trans-differentiation during embryo development and now recognized in promoting cancer cells invasiveness because of high mobility and migratory abilities of mesenchymal cells once converted from carcinoma cells. Nevertheless, the underlying pathways leading to EMT appear to be very diverse in different cancer types, which certainly represent a challenge for developing effective intervention. In this article, we have carefully reviewed the key factors involved in EMT of PCa with clinical correlation in hope to facilitate the development of new therapeutic strategy that is expected to reduce the disease mortality.
Collapse
Affiliation(s)
- U-Ging Lo
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Cheng-Fan Lee
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 10617, Taiwan.
| | - Ming-Shyue Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 10617, Taiwan.
| | - Jer-Tsong Hsieh
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
61
|
Ma SC, Li Q, Peng JY, Zhouwen JL, Diao JF, Niu JX, Wang X, Guan XD, Jia W, Jiang WG. Claudin-5 regulates blood-brain barrier permeability by modifying brain microvascular endothelial cell proliferation, migration, and adhesion to prevent lung cancer metastasis. CNS Neurosci Ther 2017; 23:947-960. [PMID: 28961379 DOI: 10.1111/cns.12764] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 09/13/2017] [Accepted: 09/13/2017] [Indexed: 02/06/2023] Open
Abstract
AIMS To investigate the roles of Claudin-5 (CLDN5) in regulating the permeability of the blood-brain barrier (BBB) during lung cancer brain metastasis. RESULTS By silencing and overexpressing the CLDN5 gene in human brain vascular endothelial (hCMEC/D3) cells, we demonstrated the attenuation of cell migration ability and CLDN5's significant positive role in cell proliferation in CLDN5-overexpressing hCMEC/D3 cells and observed the opposite result in the CLDN5 knockdown group. The reinforced CLDN5 expression reduced the paracellular permeability of hCMEC/D3 cells and decreased the invasion of lung adenocarcinoma A549 cells. Overall, 1685 genes were found to be differentially expressed between the CLDN5-overexpressing cells and the control cells using the Affymetrix Human Transcriptome Array 2.0 (HTA 2.0), and the function of these genes was determined by Gene Ontology and pathway analyses. The possible biological functions of the 1685 genes include cell proliferation, adhesion molecules, and the Jak-STAT, PI3K-Akt, Wnt, and Notch signaling pathways. The identified sets of mRNAs that were specific to CLDN5-overexpressing hCMEC/D3 cells were verified by a qRT-PCR experiment. CONCLUSION CLDN5 regulates the permeability of BBB by regulating the proliferation, migration, and permeability of hCMEC/D3 cells, especially through the cell adhesion molecule signaling pathway, to enhance the function of the tight junctions, which was involved in reducing the formation of lung cancer brain metastasis.
Collapse
Affiliation(s)
- Shun-Chang Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Department of Neurosurgery, Fuxing Hospital, Capital Medical University, Beijing, China
| | - Qi Li
- Core Laboratory for Clinical Medical Research, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China
| | - Jia-Yi Peng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jian-Long Zhouwen
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jin-Fu Diao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jian-Xing Niu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiu-Dong Guan
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wang Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wen-Guo Jiang
- Metastasis and Angiogenesis Research Group, University Department of Surgery, Cardiff University School of Medicine, Cardiff, UK
| |
Collapse
|
62
|
Sauer RS, Kirchner J, Yang S, Hu L, Leinders M, Sommer C, Brack A, Rittner HL. Blood-spinal cord barrier breakdown and pericyte deficiency in peripheral neuropathy. Ann N Y Acad Sci 2017; 1405:71-88. [DOI: 10.1111/nyas.13436] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 06/07/2017] [Accepted: 06/23/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Reine-Solange Sauer
- Department of Anesthesiology and Critical Care; University Hospital of Würzburg; Würzburg Germany
| | - Juliane Kirchner
- Department of Anesthesiology and Critical Care; University Hospital of Würzburg; Würzburg Germany
| | - Shaobing Yang
- Department of Anesthesiology and Critical Care; University Hospital of Würzburg; Würzburg Germany
- Department of Anesthesiology and Pain Medicine, Tongji Hospital of Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
| | - Liu Hu
- Department of Anesthesiology and Critical Care; University Hospital of Würzburg; Würzburg Germany
- Department of Anesthesiology and Pain Medicine, Tongji Hospital of Tongji Medical College; Huazhong University of Science and Technology; Wuhan China
| | - Mathias Leinders
- Department of Neurology; University Hospital of Würzburg; Würzburg Germany
| | - Claudia Sommer
- Department of Neurology; University Hospital of Würzburg; Würzburg Germany
| | - Alexander Brack
- Department of Anesthesiology and Critical Care; University Hospital of Würzburg; Würzburg Germany
| | - Heike L. Rittner
- Department of Anesthesiology and Critical Care; University Hospital of Würzburg; Würzburg Germany
| |
Collapse
|
63
|
Li H, Zhong A, Li S, Meng X, Wang X, Xu F, Lai M. The integrated pathway of TGFβ/Snail with TNFα/NFκB may facilitate the tumor-stroma interaction in the EMT process and colorectal cancer prognosis. Sci Rep 2017; 7:4915. [PMID: 28687755 PMCID: PMC5501852 DOI: 10.1038/s41598-017-05280-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 05/26/2017] [Indexed: 12/14/2022] Open
Abstract
Substantial evidence has shown that epithelial-mesenchymal transition (EMT) plays critical roles in colorectal cancer (CRC) development and prognosis. To uncover the pivotal regulators that function in the cooperative interactions between cancer cells and their microenvironment and consequently affect the EMT process, we carried out a systematic analysis and evaluated prognosis in CRC specimens. Tumor buds and their surrounding stroma were captured using laser microdissection. We used gene expression profiling, bioinformatics analysis and regulatory network construction for molecular selection. The clinical significance of potential biomarkers was investigated. We identified potential EMT biomarkers, including BGN, MMP1, LGALS1, SERPINB5, and TM4SF4, all of which participated in the integrated pathway of TGFβ/Snail with TNFα/NFκB. We also found that BGN, MMP1, LGALS1, SERPINB5 and TM4SF4 were related to CRC patient prognosis. Patients with higher expression of these individual potential biomarkers had poorer prognosis. Among the identified biomarkers, BGN and TM4SF4 are reported, for the first time, to probably be involved in the EMT process and to predict CRC prognosis. Our results strongly suggest that the integrated pathway of TGFβ/Snail with TNFα/NFκB may be the principal axis that links cancer cells to their microenvironment during the EMT process and results in poor prognosis in CRC patients.
Collapse
Affiliation(s)
- Hui Li
- Department of Pathology, School of Medicine, Zhejiang University, Hangzhou, 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, 310058, China
| | - Anjing Zhong
- Department of Pathology, School of Medicine, Zhejiang University, Hangzhou, 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, 310058, China
| | - Si Li
- Department of Pathology, School of Medicine, Zhejiang University, Hangzhou, 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, 310058, China
| | - Xianwen Meng
- Department of Bioinformatics, State Key Laboratory of Plant Physiology and Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xue Wang
- China Pharmaceutical University, Nanjing, 320100, China
| | - Fangying Xu
- Department of Pathology, School of Medicine, Zhejiang University, Hangzhou, 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, 310058, China
| | - Maode Lai
- Department of Pathology, School of Medicine, Zhejiang University, Hangzhou, 310058, China. .,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, 310058, China.
| |
Collapse
|
64
|
Wu J, Zhou XJ, Sun X, Xia TS, Li XX, Shi L, Zhu L, Zhou WB, Wei JF, Ding Q. RBM38 is involved in TGF-β-induced epithelial-to-mesenchymal transition by stabilising zonula occludens-1 mRNA in breast cancer. Br J Cancer 2017; 117:675-684. [PMID: 28683467 PMCID: PMC5572167 DOI: 10.1038/bjc.2017.204] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/31/2017] [Accepted: 06/07/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The transforming growth factor-β (TGF-β) pathway plays a vital role in driving cancer cell epithelial-mesenchymal transition (EMT). Zonula occludens-1 (ZO-1), which is downregulated in response to TGF-β, is able to control endothelial cell-cell tension, cell migration, and barrier formation. However, the molecular mechanism of how TGF-β regulates ZO-1 expression remains unclear. METHODS Breast cancer cells were treated with TGF-β to induce an EMT progress. Chromatin immunoprecipitation and dual-luciferase reporter assay were performed to investigate direct relationship between Snail and RNA binding motif protein 38 (RBM38). The RNA immunoprecipitation combined with RNA electrophoretic mobility shift assay and dual-luciferase reporter assay were conducted to testify direct relationship between RBM38 and ZO-1. The ZO-1 siRNA was transfected to breast cancer cells that overexpress RBM38 and the control, followed by transwell and Matrigel invasion assays to examine cell migratory and invasive ability. RESULTS Transforming growth factor-β induced a remarkable downregulation of RBM38 in breast cancer that was directly regulated by transcription repressor Snail targeting the E-box elements in promoter region of RBM38 gene. Additionally, RBM38 positively regulated ZO-1 transcript via directly binding to AU/U-rich elements in its mRNA 3'-UTR. Moreover, by magnifying RBM38 expression, cell migration and invasion mediated by knockdown of ZO-1 in breast cancer were reversed. CONCLUSIONS All the results clarified a linear regulation relationship among Snail, RBM38, and ZO-1, implicating RBM38 as a pivotal mediator in TGF-β-induced EMT in breast cancer.
Collapse
Affiliation(s)
- Jing Wu
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Xu-Jie Zhou
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Xi Sun
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Tian-Song Xia
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Xiao-Xia Li
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Liang Shi
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Lei Zhu
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Wen-Bin Zhou
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Ji-Fu Wei
- Research Division of Clinical Pharmacology, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| | - Qiang Ding
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, 300 Guangzhou Road, Nanjing 210029, China
| |
Collapse
|
65
|
Wang P, Voronkova M, Luanpitpong S, He X, Riedel H, Dinu CZ, Wang L, Rojanasakul Y. Induction of Slug by Chronic Exposure to Single-Walled Carbon Nanotubes Promotes Tumor Formation and Metastasis. Chem Res Toxicol 2017; 30:1396-1405. [PMID: 28598615 DOI: 10.1021/acs.chemrestox.7b00049] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Carbon nanotubes (CNTs) represent a major class of engineered nanomaterials that are being used in diverse fields. However, their use has increasingly become a concern because of their carcinogenic potential. Accumulating evidence has demonstrated that certain types of CNTs are carcinogenic or tumor-promoting in animal models. However, the underlying molecular and cellular mechanisms are unclear. Here, we report that chronic exposure to single-walled (SW) CNTs results in the induction of Slug, a key transcription factor that induces an epithelial-mesenchymal transition (EMT), in human lung epithelial cells. We show that SWCNT-induced Slug upregulation plays a critical role in the aggressive phenotype of SWCNT-exposed cells, which includes increased cell migration, invasion, and anchorage-independent cell growth. Our in vivo studies also show that SWCNT-induced Slug upregulation and EMT activation play a pivotal role in tumor formation and metastasis. Our findings illustrate a direct link between CNT-induced Slug upregulation, EMT activation, and tumor formation and metastasis, and they highlight the potential of CNT-induced Slug upregulation as a target for future risk assessment and prevention of CNT-associated diseases.
Collapse
Affiliation(s)
| | | | - Sudjit Luanpitpong
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine, Mahidol University , Bangkok 10700, Thailand
| | | | | | | | - Liying Wang
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health , Morgantown, West Virginia 26505, United States
| | | |
Collapse
|
66
|
Epithelial-to-mesenchymal transition in tumor progression. Med Oncol 2017; 34:122. [PMID: 28560682 DOI: 10.1007/s12032-017-0980-8] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 05/24/2017] [Indexed: 12/13/2022]
Abstract
The epithelial-to-mesenchymal transition (EMT) is a biological process in which a non-motile epithelial cell changes to a mesenchymal state with invasive capacities. However, the EMT program is involved in both physiological and pathological processes. Cancer-associated EMT is known to contribute to increase invasiveness and metastasis, resistance to therapies, and generation of cell populations with stem cell-like characteristics and therefore is deeply involved in tumor progression. This process is finely orchestrated by multiple signaling pathways and regulatory transcriptional networks. The hallmark of EMT is the loss of epithelial surface markers, mainly E-cadherin, and the acquisition of mesenchymal phenotype. These events can be mediated by EMT transcription factors which can cooperate with several enzymes to repress the E-cadherin expression and regulate EMT at the epigenetic and post-translational level. A growing body of evidence indicates that cancer cells can reside in various phenotypic states along the EMT spectrum, where cells can jointly retain epithelial traits with mesenchymal ones. This type of phenotypic plasticity endows cancer cells with tumor-initiating potential. The identification of the signaling pathways and modulators that lead to activation of EMT programs during these disease processes is providing new insights into the plasticity of cellular phenotypes and possible therapeutic interventions.
Collapse
|
67
|
Lopez-Anton M, Lambie M, Lopez-Cabrera M, Schmitt CP, Ruiz-Carpio V, Bartosova M, Schaefer B, Davies S, Stone T, Jenkins R, Taylor PR, Topley N, Bowen T, Fraser D. miR-21 Promotes Fibrogenesis in Peritoneal Dialysis. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:1537-1550. [PMID: 28495592 DOI: 10.1016/j.ajpath.2017.03.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 03/28/2017] [Indexed: 12/13/2022]
Abstract
Peritoneal dialysis (PD) is a life-saving form of renal replacement therapy for those with end-stage kidney disease. Mesothelial cells (MCs) line the peritoneal cavity and help define peritoneal response to treatment-associated injury, a major reason for treatment failure. miRNAs are important regulators, but their roles in peritoneal fibrosis are largely unknown. In this study, miR-21 was one of the most abundant miRNAs in primary MCs, and was up-regulated by the profibrotic cytokine transforming growth factor-β1 and in PD effluent-derived MCs exhibiting mesenchymal phenotypic change. Increased miR-21 was found in peritoneal membrane biopsy specimens from PD patients compared to healthy controls (PD biocompatible, 5.86×, P = 0.0001; PD conventional, 7.09×, P < 0.0001, n = 11 per group). In PD effluent from a cohort of 230 patients, miR-21 was higher in those receiving the therapy long-term compared to new starters (n = 230, miR-21 3.26×, P = 0.001) and associated with icodextrin use (R = 0.52; 95% CI, 0.20-0.84), peritonitis count (R = 0.16; 95% CI, 0.03-0.29), and dialysate cytokines. miR-21 down-regulated programmed cell death 4 and programmed cell death 4 protein was decreased in peritoneal membrane biopsy specimens from PD patients compared to healthy controls. New miR-21 targets were identified that may be important during PD fibrogenesis. These data identify miR-21 as an important effector of fibrosis in the peritoneal membrane, and a promising biomarker in the dialysis effluent for membrane change in patients receiving PD.
Collapse
Affiliation(s)
- Melisa Lopez-Anton
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | | | - Manuel Lopez-Cabrera
- Centro de Biología Molecular Severo Ochoa, the Spanish National Research Council, Madrid, Spain
| | | | - Vicente Ruiz-Carpio
- Centro de Biología Molecular Severo Ochoa, the Spanish National Research Council, Madrid, Spain
| | | | | | | | - Timothy Stone
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | - Robert Jenkins
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | - Philip R Taylor
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | - Nicholas Topley
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | - Timothy Bowen
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | - Donald Fraser
- Wales Kidney Research Unit, Division of Infection and Immunity, School of Medicine, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom.
| |
Collapse
|
68
|
Li J, Qu J, Shi Y, Perfetto M, Ping Z, Christian L, Niu H, Mei S, Zhang Q, Yang X, Wei S. Nicotinic acid inhibits glioma invasion by facilitating Snail1 degradation. Sci Rep 2017; 7:43173. [PMID: 28256591 PMCID: PMC5335718 DOI: 10.1038/srep43173] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 01/23/2017] [Indexed: 01/22/2023] Open
Abstract
Malignant glioma is a formidable disease that commonly leads to death, mainly due to the invasion of tumor cells into neighboring tissues. Therefore, inhibition of tumor cell invasion may provide an effective therapy for malignant glioma. Here we report that nicotinic acid (NA), an essential vitamin, inhibits glioma cell invasion in vitro and in vivo. Treatment of the U251 glioma cells with NA in vitro results in reduced invasion, which is accompanied by a loss of mesenchymal phenotype and an increase in cell-cell adhesion. At the molecular level, transcription of the adherens junction protein E-cadherin is upregulated, leading to accumulation of E-cadherin protein at the cell-cell boundary. This can be attributed to NA's ability to facilitate the ubiquitination and degradation of Snail1, a transcription factor that represses E-cadherin expression. Similarly, NA transiently inhibits neural crest migration in Xenopus embryos in a Snail1-dependent manner, indicating that the mechanism of action for NA in cell migration is evolutionarily conserved. We further show that NA injection blocks the infiltration of tumor cells into the adjacent brain tissues and improves animal survival in a rat model of glioma. These results suggest that NA treatment may be developed into a potential therapy for malignant glioma.
Collapse
Affiliation(s)
- Jiejing Li
- Department of Clinical Laboratory, The Affiliated Hospital of KMUST, Medical School, Kunming University of Science and Technology, Kunming 650032, China.,Department of Biology, West Virginia University, Morgantown, WV 26506, United States
| | - Jiagui Qu
- Department of Clinical Laboratory, The Affiliated Hospital of KMUST, Medical School, Kunming University of Science and Technology, Kunming 650032, China
| | - Yu Shi
- Department of Clinical Laboratory, Children's Hospital of Chongqing Medical University, Chongqing 400014, China.,Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics; Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing 400014, China
| | - Mark Perfetto
- Department of Biology, West Virginia University, Morgantown, WV 26506, United States.,Department of Biological Sciences, University of Delaware, Newark, DE 19716, United States
| | - Zhuxian Ping
- Department of Clinical Laboratory, The Affiliated Hospital of KMUST, Medical School, Kunming University of Science and Technology, Kunming 650032, China
| | - Laura Christian
- Department of Biology, West Virginia University, Morgantown, WV 26506, United States
| | - Hua Niu
- Department of Clinical Laboratory, The Affiliated Hospital of KMUST, Medical School, Kunming University of Science and Technology, Kunming 650032, China
| | - Shuting Mei
- Department of Gerontology, First People's Hospital of Yunnan Province, Kunming 650032, China
| | - Qin Zhang
- Department of Clinical Laboratory, The Affiliated Hospital of KMUST, Medical School, Kunming University of Science and Technology, Kunming 650032, China
| | - Xiangcai Yang
- Department of Clinical Laboratory, The Affiliated Hospital of KMUST, Medical School, Kunming University of Science and Technology, Kunming 650032, China
| | - Shuo Wei
- Department of Biology, West Virginia University, Morgantown, WV 26506, United States.,Department of Biological Sciences, University of Delaware, Newark, DE 19716, United States
| |
Collapse
|
69
|
Wang D, Zhu C, Zhang Y, Zheng Y, Ma F, Su L, Shao G. MicroRNA-30e-3p inhibits cell invasion and migration in clear cell renal cell carcinoma by targeting Snail1. Oncol Lett 2017; 13:2053-2058. [PMID: 28454361 PMCID: PMC5403512 DOI: 10.3892/ol.2017.5690] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 10/28/2016] [Indexed: 01/16/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common type of neoplasm affecting the adult kidney. Previous studies on ccRCC have focused on microRNAs, a class of small non-coding RNAs that are important in cancer development and progression. The present study aimed to investigate the potential role of microRNA (miR)-30e-3p in ccRCC. The results revealed that overexpression of miR-30e-3p in the A498 and 786O ccRCC cell lines was able to inhibit cell invasion and migration. The expression level of Snail1, a potential target gene of miR-30e-3p, was inversely correlated with miR-30e-3p expression in ccRCC tissues and cell lines. Furthermore, Snail1 was revealed to be directly regulated by miR-30e-3p and had an important role in mediating the biological effects of miR-30e-3p in ccRCC. Restoration of Snail1 expression was able to reverse the inhibitory properties of miR-30e-3p. Therefore, the results of the current study suggest that miR-30e-3p exerts its anticancer functions through direct targeting of Snail1 in ccRCC cells, and may be a novel therapeutic agent for this form of cancer.
Collapse
Affiliation(s)
- Daya Wang
- Department of Urology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Chao Zhu
- Department of Nephrology, People's Liberation Army No. 411 Hospital, Shanghai 200081, P.R. China
| | - Yifan Zhang
- Department of Nephrology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Yuenan Zheng
- Department of Nephrology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Feiju Ma
- Department of Urology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Li Su
- School of Pharmacy, Second Military Medical University, Shanghai 200433, P.R. China
| | - Guojian Shao
- Department of Nephrology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
70
|
EMT and stemness: flexible processes tuned by alternative splicing in development and cancer progression. Mol Cancer 2017; 16:8. [PMID: 28137272 PMCID: PMC5282733 DOI: 10.1186/s12943-016-0579-2] [Citation(s) in RCA: 219] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 12/25/2016] [Indexed: 12/17/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is associated with metastasis formation as well as with generation and maintenance of cancer stem cells. In this way, EMT contributes to tumor invasion, heterogeneity and chemoresistance. Morphological and functional changes involved in these processes require robust reprogramming of gene expression, which is only partially accomplished at the transcriptional level. Alternative splicing is another essential layer of gene expression regulation that expands the cell proteome. This step in post-transcriptional regulation of gene expression tightly controls cell identity between epithelial and mesenchymal states and during stem cell differentiation. Importantly, dysregulation of splicing factor function and cancer-specific splicing isoform expression frequently occurs in human tumors, suggesting the importance of alternative splicing regulation for cancer biology. In this review, we briefly discuss the role of EMT programs in development, stem cell differentiation and cancer progression. Next, we focus on selected examples of key factors involved in EMT and stem cell differentiation that are regulated post-transcriptionally through alternative splicing mechanisms. Lastly, we describe relevant oncogenic splice-variants that directly orchestrate cancer stem cell biology and tumor EMT, which may be envisioned as novel targets for therapeutic intervention.
Collapse
|
71
|
Sivagurunathan S, Palanisamy K, Arunachalam JP, Chidambaram S. Possible role of HIWI2 in modulating tight junction proteins in retinal pigment epithelial cells through Akt signaling pathway. Mol Cell Biochem 2016; 427:145-156. [DOI: 10.1007/s11010-016-2906-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 12/03/2016] [Indexed: 12/22/2022]
|
72
|
Yeung KT, Yang J. Epithelial-mesenchymal transition in tumor metastasis. Mol Oncol 2016; 11:28-39. [PMID: 28085222 PMCID: PMC5242415 DOI: 10.1002/1878-0261.12017] [Citation(s) in RCA: 503] [Impact Index Per Article: 55.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/13/2016] [Accepted: 10/21/2016] [Indexed: 12/15/2022] Open
Abstract
The epithelial-mesenchymal transition (EMT) is a developmental program that enables stationary epithelial cells to gain the ability to migrate and invade as single cells. Tumor cells reactivate EMT to acquire molecular alterations that enable the partial loss of epithelial features and partial gain of a mesenchymal phenotype. Our understanding of the contribution of EMT to tumor invasion, migration, and metastatic outgrowth has evolved over the past decade. In this review, we provide a summary of both historic and recent studies on the role of EMT in the metastatic cascade from various experimental systems, including cancer cell lines, genetic mouse tumor models, and clinical human breast cancer tissues.
Collapse
Affiliation(s)
- Kay T Yeung
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA.,Department of Medicine, University of California, San Diego, La Jolla, CA, USA.,Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Jing Yang
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA.,Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.,Department of Pediatrics, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
73
|
Campbell K, Casanova J. A common framework for EMT and collective cell migration. Development 2016; 143:4291-4300. [DOI: 10.1242/dev.139071] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
During development, cells often switch between static and migratory behaviours. Such transitions are fundamental events in development and are linked to harmful consequences in pathology. It has long been considered that epithelial cells either migrate collectively as epithelial cells, or undergo an epithelial-to-mesenchymal transition and migrate as individual mesenchymal cells. Here, we assess what is currently known about in vivo cell migratory phenomena and hypothesise that such migratory behaviours do not fit into alternative and mutually exclusive categories. Rather, we propose that these categories can be viewed as the most extreme cases of a general continuum of morphological variety, with cells harbouring different degrees or combinations of epithelial and mesenchymal features and displaying an array of migratory behaviours.
Collapse
Affiliation(s)
- Kyra Campbell
- Institut de Biologia Molecular de Barcelona (CSIC), C/Baldiri Reixac 10, Barcelona, Catalonia 08028, Spain
- Institut de Recerca Biomèdica de Barcelona, C/Baldiri Reixac 10, Barcelona, Catalonia 08028, Spain
| | - Jordi Casanova
- Institut de Biologia Molecular de Barcelona (CSIC), C/Baldiri Reixac 10, Barcelona, Catalonia 08028, Spain
- Institut de Recerca Biomèdica de Barcelona, C/Baldiri Reixac 10, Barcelona, Catalonia 08028, Spain
| |
Collapse
|
74
|
Bhat AA, Ahmad R, Uppada SB, Singh AB, Dhawan P. Claudin-1 promotes TNF-α-induced epithelial-mesenchymal transition and migration in colorectal adenocarcinoma cells. Exp Cell Res 2016; 349:119-127. [PMID: 27742576 PMCID: PMC6166648 DOI: 10.1016/j.yexcr.2016.10.005] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 09/02/2016] [Accepted: 10/05/2016] [Indexed: 02/07/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is an important mechanism in cancer progression and malignancy including colorectal cancer (CRC). Importantly, inflammatory mediators are critical constituents of the local tumor environment and an intimate link between CRC progression and inflammation is now validated. We and others have reported key role of the deregulated claudin-1 expression in colon carcinogenesis including colitis-associated colon cancer (CAC). However, the causal association between claudin-1 expression and inflammation-induced colon cancer progression remains unclear. Here we demonstrate, TNF-α, a pro-inflammatory cytokine, regulates claudin-1 to modulate epithelial to mesenchymal transition (EMT) and migration in colon adenocarcinoma cells. Importantly, colon cancer cells cultured in the presence of TNF-α (10ng/ml), demonstrated a sharp decrease in E-cadherin expression and an increase in vimentin expression (versus control cells). Interestingly, TNF-α treatment also upregulated (and delocalized) claudin-1 expression in a time-dependent manner accompanied by increase in proliferation and wound healing. Furthermore, similar to our previous observation that claudin-1 overexpression in CRC cells induces ERK1/2 and Src- activation, signaling associated with colon cancer cell survival and transformation, TNF-α-treatment induced upregulation of phospho-ERK1/2 and -Src expression. The shRNA-mediated inhibition of claudin-1 expression largely abrogated the TNF-α-induced changes in EMT, proliferation, migration, p-Erk and p-Src expression. Taken together, our data demonstrate TNF-α mediated regulation of claudin-1 and tumorigenic abilities of colon cancer cells and highlights a key role of deregulated claudin-1 expression in inflammation-induced colorectal cancer growth and progression, through the regulation of the ERK and Src-signaling.
Collapse
Affiliation(s)
- Ajaz A Bhat
- Surgery, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Rizwan Ahmad
- Departments of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68022, United States
| | - SrijayaPrakash B Uppada
- Departments of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68022, United States
| | - Amar B Singh
- From the Department of Veterans Affairs, University of Nebraska Medical Center, Omaha, NE 68022, United States; Departments of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68022, United States; Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68022, United States
| | - Punita Dhawan
- From the Department of Veterans Affairs, University of Nebraska Medical Center, Omaha, NE 68022, United States; Departments of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68022, United States; Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68022, United States.
| |
Collapse
|
75
|
Faris AN, Ganesan S, Chattoraj A, Chattoraj SS, Comstock AT, Unger BL, Hershenson MB, Sajjan US. Rhinovirus Delays Cell Repolarization in a Model of Injured/Regenerating Human Airway Epithelium. Am J Respir Cell Mol Biol 2016; 55:487-499. [PMID: 27119973 DOI: 10.1165/rcmb.2015-0243oc] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Rhinovirus (RV), which causes exacerbation in patients with chronic airway diseases, readily infects injured airway epithelium and has been reported to delay wound closure. In this study, we examined the effects of RV on cell repolarization and differentiation in a model of injured/regenerating airway epithelium (polarized, undifferentiated cells). RV causes only a transient barrier disruption in a model of normal (mucociliary-differentiated) airway epithelium. However, in the injury/regeneration model, RV prolongs barrier dysfunction and alters the differentiation of cells. The prolonged barrier dysfunction caused by RV was not a result of excessive cell death but was instead associated with epithelial-to-mesenchymal transition (EMT)-like features, such as reduced expression of the apicolateral junction and polarity complex proteins, E-cadherin, occludin, ZO-1, claudins 1 and 4, and Crumbs3 and increased expression of vimentin, a mesenchymal cell marker. The expression of Snail, a transcriptional repressor of tight and adherence junctions, was also up-regulated in RV-infected injured/regenerating airway epithelium, and inhibition of Snail reversed RV-induced EMT-like features. In addition, compared with sham-infected cells, the RV-infected injured/regenerating airway epithelium showed more goblet cells and fewer ciliated cells. Inhibition of epithelial growth factor receptor promoted repolarization of cells by inhibiting Snail and enhancing expression of E-cadherin, occludin, and Crumbs3 proteins, reduced the number of goblet cells, and increased the number of ciliated cells. Together, these results suggest that RV not only disrupts barrier function, but also interferes with normal renewal of injured/regenerating airway epithelium by inducing EMT-like features and subsequent goblet cell hyperplasia.
Collapse
Affiliation(s)
- Andrea N Faris
- 1 Departments of Pediatrics and Communicable Diseases and
| | | | | | | | | | | | - Marc B Hershenson
- 1 Departments of Pediatrics and Communicable Diseases and.,2 Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | | |
Collapse
|
76
|
Yun J, Song SH, Kim HP, Han SW, Yi EC, Kim TY. Dynamic cohesin-mediated chromatin architecture controls epithelial-mesenchymal plasticity in cancer. EMBO Rep 2016; 17:1343-59. [PMID: 27466323 PMCID: PMC5007572 DOI: 10.15252/embr.201541852] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 06/03/2016] [Accepted: 06/22/2016] [Indexed: 01/26/2023] Open
Abstract
Epithelial to mesenchymal transition (EMT) and mesenchymal to epithelial transition (MET) are important interconnected events in tumorigenesis controlled by complex genetic networks. However, the cues that activate EMT-initiating factors and the mechanisms that reversibly connect EMT/MET are not well understood. Here, we show that cohesin-mediated chromatin organization coordinates EMT/MET by regulating mesenchymal genes. We report that RAD21, a subunit of the cohesin complex, is expressed in epithelial breast cancer cells, whereas its expression is decreased in mesenchymal cancer. Depletion of RAD21 in epithelial cancer cells causes transcriptional activation of TGFB1 and ITGA5, inducing EMT. Reduced binding of RAD21 changes intrachromosomal chromatin interactions within the TGFB1 and ITGA5 loci, creating an active transcriptional environment. Similarly, stem cell-like cancer cells also show an open chromatin structure at both genes, which correlates with high expression levels and mesenchymal fate characteristics. Conversely, overexpression of RAD21 in mesenchymal cancer cells induces MET-specific expression patterns. These findings indicate that dynamic cohesin-mediated chromatin structures are responsible for the initiation and regulation of essential EMT-related cell fate changes in cancer.
Collapse
Affiliation(s)
- Jiyeon Yun
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology Seoul National University College of Medicine, Seoul, Korea
| | - Sang-Hyun Song
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hwang-Phill Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Sae-Won Han
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Eugene C Yi
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology Seoul National University College of Medicine, Seoul, Korea
| | - Tae-You Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology Seoul National University College of Medicine, Seoul, Korea Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
77
|
Tanaka S, Kobayashi W, Haraguchi M, Ishihata K, Nakamura N, Ozawa M. Snail1 expression in human colon cancer DLD-1 cells confers invasive properties without N-cadherin expression. Biochem Biophys Rep 2016; 8:120-126. [PMID: 28955947 PMCID: PMC5613769 DOI: 10.1016/j.bbrep.2016.08.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 07/19/2016] [Accepted: 08/17/2016] [Indexed: 11/30/2022] Open
Abstract
The epithelial-mesenchymal transition (EMT) is a fundamental characteristic of carcinoma cells. EMT is generally associated with a change in cellular morphology from cobblestone to spindle shape, reduced expression of epithelial markers such as E-cadherin, and enhanced expression of mesenchymal markers such as N-cadherin. This EMT-associated reciprocal expression of E-cadherin and N-cadherin has been called the "cadherin switch". Downregulation of E-cadherin enables cells to dissociate from colonies while upregulation of N-cadherin is associated with increased invasiveness. The transcription factor Snail1 induces these changes in various epithelial cell lines, including canine MDCK cells and human A431 cells. In the present study, we introduced a Snail1 expression vector into human DLD-1 cells and isolated stable transfectants. These cells showed changes in morphology, reduced expression of epithelial marker E-cadherin and occludin, and elevated invasion and migration. However, neither expression of N-cadherin protein nor its corresponding mRNA was detected. Therefore, elevated N-cadherin expression is not required for invasiveness of the cells.
Collapse
Affiliation(s)
- Shoko Tanaka
- Department of Biochemistry and Molecular Biology, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan.,Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Wakako Kobayashi
- Department of Biochemistry and Molecular Biology, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Misako Haraguchi
- Department of Biochemistry and Molecular Biology, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Kiyohide Ishihata
- Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Norifumi Nakamura
- Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Masayuki Ozawa
- Department of Biochemistry and Molecular Biology, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| |
Collapse
|
78
|
Salvador E, Burek M, Förster CY. Tight Junctions and the Tumor Microenvironment. CURRENT PATHOBIOLOGY REPORTS 2016; 4:135-145. [PMID: 27547510 PMCID: PMC4978755 DOI: 10.1007/s40139-016-0106-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE OF REVIEW Tight junctions (TJs) are specialized differentiations of epithelial and endothelial cell membranes. TJs play an important role in the adhesion of cells and their interaction with each other. Most cancers originate from epithelial cells. Thus, it is of significance to examine the role of TJs in the tumor microenvironment (TME) and how they affect cancer metastasis. RECENT FINDINGS In epithelium-derived cancers, intactness of the primary tumor mass is influenced by intercellular structures as well as cell-to-cell adhesion. Irregularities of these factors may lead to tumor dissociation and subsequent metastasis. Low expression of TJs is observed among highly metastatic cancer cells. SUMMARY In this review, we summarized findings from current literature in consideration of the role of TJs in relation to the TME and cancer. Deeper understanding of the mechanisms leading to TJ dysregulation is needed to facilitate the design and conceptualization of new and better therapeutic strategies for cancer.
Collapse
Affiliation(s)
- Ellaine Salvador
- Department of Anesthesia and Critical Care, University of Wurzburg, Oberdürrbacher Straße 6, 97080 Würzburg, Germany
| | - Malgorzata Burek
- Department of Anesthesia and Critical Care, University of Wurzburg, Oberdürrbacher Straße 6, 97080 Würzburg, Germany
| | - Carola Y. Förster
- Department of Anesthesia and Critical Care, University of Wurzburg, Oberdürrbacher Straße 6, 97080 Würzburg, Germany
| |
Collapse
|
79
|
Multifaceted role of E-cadherin in hepatitis C virus infection and pathogenesis. Proc Natl Acad Sci U S A 2016; 113:7298-300. [PMID: 27342863 DOI: 10.1073/pnas.1607732113] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
80
|
Moly PK, Cooley JR, Zeltzer SL, Yatskievych TA, Antin PB. Gastrulation EMT Is Independent of P-Cadherin Downregulation. PLoS One 2016; 11:e0153591. [PMID: 27097030 PMCID: PMC4838233 DOI: 10.1371/journal.pone.0153591] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 03/31/2016] [Indexed: 11/24/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is an evolutionarily conserved process during which cells lose epithelial characteristics and gain a migratory phenotype. Although downregulation of epithelial cadherins by Snail and other transcriptional repressors is generally considered a prerequisite for EMT, recent studies have challenged this view. Here we investigate the relationship between E-cadherin and P-cadherin expression and localization, Snail function and EMT during gastrulation in chicken embryos. Expression analyses show that while E-cadherin transcripts are detected in the epiblast but not in the primitive streak or mesoderm, P-cadherin mRNA and protein are present in the epiblast, primitive and mesoderm. Antibodies that specifically recognize E-cadherin are not presently available. During EMT, P-cadherin relocalizes from the lateral surfaces of epithelial epiblast cells to a circumferential distribution in emerging mesodermal cells. Cells electroporated with an E-cadherin expression construct undergo EMT and migrate into the mesoderm. An examination of Snail function showed that reduction of Slug (SNAI2) protein levels using a morpholino fails to inhibit EMT, and expression of human or chicken Snail in epiblast cells fails to induce EMT. In contrast, cells expressing the Rho inhibitor peptide C3 rapidly exit the epiblast without activating Slug or the mesoderm marker N-cadherin. Together, these experiments show that epiblast cells undergo EMT while retaining P-cadherin, and raise questions about the mechanisms of EMT regulation during avian gastrulation.
Collapse
Affiliation(s)
- Pricila K. Moly
- Department of Cellular and Molecular Medicine, University of Arizona, 1656 E. Mabel Street, P.O. Box 245217, Tucson, AZ, 85724, United States of America
| | - James R. Cooley
- Department of Cellular and Molecular Medicine, University of Arizona, 1656 E. Mabel Street, P.O. Box 245217, Tucson, AZ, 85724, United States of America
| | - Sebastian L. Zeltzer
- Department of Cellular and Molecular Medicine, University of Arizona, 1656 E. Mabel Street, P.O. Box 245217, Tucson, AZ, 85724, United States of America
| | - Tatiana A. Yatskievych
- Department of Cellular and Molecular Medicine, University of Arizona, 1656 E. Mabel Street, P.O. Box 245217, Tucson, AZ, 85724, United States of America
| | - Parker B. Antin
- Department of Cellular and Molecular Medicine, University of Arizona, 1656 E. Mabel Street, P.O. Box 245217, Tucson, AZ, 85724, United States of America
- * E-mail:
| |
Collapse
|
81
|
Inada M, Izawa G, Kobayashi W, Ozawa M. 293 cells express both epithelial as well as mesenchymal cell adhesion molecules. Int J Mol Med 2016; 37:1521-7. [PMID: 27121032 PMCID: PMC4866952 DOI: 10.3892/ijmm.2016.2568] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 02/24/2016] [Indexed: 11/06/2022] Open
Abstract
The 293 cell line, used extensively in various types of studies due to the ease with which these cells can be transfected, was thought to be derived by the transformation of primary cultures of human embryonic kidney cells with sheared adenovirus type 5 DNA. Although the 293 cells were assumed to originate from epithelial cells, the exact origin of these cells remains unknown. Previous attempts to characterize these cells combined immunostaining, immunoblot analysis and microarray analysis to demonstrate that 293 cells express neurofilament subunits, α-internexin, and several other proteins typically found in neurons. These findings raised the possibility that the 293 cell line may have originated from human neuronal lineage cells. Contrary to this suggestion, in this study, we found that the 293 cells expressed N-cadherin and vimentin, which are marker proteins expressed in mesenchymal cells. Furthermore, the 293 cells also expressed E-cadherin, cytokeratins 5/8 and desmoglein 2, which are epithelial cell markers. When the cells, primarily cultured from the kidneys of Clawn miniature swine and passaged 10–15 generations [termed porcine kidney epithelial (PKE) cells] were examined, they were found to be positive for the expression of both mesenchymal and epithelial markers. Thus, transformation by adenovirus was not necessary for the cells to express N-cadherin. Occludin and zonula occludens (ZO)-1, two components of tight junctions in epithelial and endothelial cells, were detected in the 293 and the PKE cells. Thus, the findings of the present study demonstrate that 293 cells retain several characteristics of epithelial cells.
Collapse
Affiliation(s)
- Masakazu Inada
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Genya Izawa
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Wakako Kobayashi
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Masayuki Ozawa
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| |
Collapse
|
82
|
α-Actinin-4 induces the epithelial-to-mesenchymal transition and tumorigenesis via regulation of Snail expression and β-catenin stabilization in cervical cancer. Oncogene 2016; 35:5893-5904. [PMID: 27065319 DOI: 10.1038/onc.2016.117] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 12/30/2016] [Accepted: 01/08/2016] [Indexed: 02/08/2023]
Abstract
α-Actinin-4 (ACTN4) is frequently amplified and overexpressed in various cancers. Although ACTN4 functions in cancer cell migration and invasion, the roles of ACTN4 during the epithelial-to-mesenchymal transition (EMT) and cervical cancer tumorigenesis are unknown. In this study, we investigated the function of ACTN4 in the progression of cervical cancer and the mechanisms of EMT and tumorigenesis induced by ACTN4. We found that ACTN4 induced EMT by upregulating Snail, which was dependent on the Akt signaling pathway in cervical cancer. ACTN4 induced cell migration and invasion through Snail-mediated matrix metalloproteinase-9 expression. ACTN4 expression level was correlated with stabilization of β-catenin. Accumulatioin of β-catenin owing to ACTN4 induced tumorigenesis via upregulation of genes involved in cell proliferation, including cyclin D1 and c-myc. ACTN4 knockdown reduced cervical cancer cell proliferation and tumor formation in vivo. The expression level of ACTN4 is highly elevated in human cervical tumors, compared with that in normal cervical tissues. ACTN4-overexpressing MDCK cells induced tumor formation and metastatic nodules in nude mice. Our findings indicate that ACTN4 promotes EMT and tumorigenesis by regulating Snail expression and the Akt pathway in cervical cancer. We propose a novel mechanism for EMT and tumorigenesis in cervical cancer.
Collapse
|
83
|
Dissecting Stages of Human Kidney Development and Tumorigenesis with Surface Markers Affords Simple Prospective Purification of Nephron Stem Cells. Sci Rep 2016; 6:23562. [PMID: 27020553 PMCID: PMC4810363 DOI: 10.1038/srep23562] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 03/08/2016] [Indexed: 01/16/2023] Open
Abstract
When assembling a nephron during development a multipotent stem cell pool becomes
restricted as differentiation ensues. A faulty differentiation arrest in this
process leads to transformation and initiation of a Wilms’ tumor.
Mapping these transitions with respective surface markers affords accessibility to
specific cell subpopulations. NCAM1 and CD133 have been previously suggested to mark
human renal progenitor populations. Herein, using cell sorting, RNA sequencing,
in vitro studies with serum-free media and in vivo
xenotransplantation we demonstrate a sequential map that links human kidney
development and tumorigenesis; In nephrogenesis,
NCAM1+CD133− marks
SIX2+ multipotent renal stem cells transiting to
NCAM1+CD133+ differentiating segment-specific
SIX2− epithelial progenitors and
NCAM1−CD133+ differentiated nephron
cells. In tumorigenesis, NCAM1+CD133−
marks SIX2+ blastema that includes the ALDH1+ WT
cancer stem/initiating cells, while NCAM1+CD133+ and
NCAM1−CD133+ specifying early and late
epithelial differentiation, are severely restricted in tumor initiation capacity and
tumor self-renewal. Thus, negative selection for CD133 is required for defining
NCAM1+ nephron stem cells in normal and malignant
nephrogenesis.
Collapse
|
84
|
Xu XY, Chai JJ, Chen YP, Rui HL, Wang YY, Dong HR, Man YL, Cheng H. Hirsutella sinensis Attenuates Aristolochic Acid-Induced Renal Tubular Epithelial-Mesenchymal Transition by Inhibiting TGF-β1 and Snail Expression. PLoS One 2016; 11:e0149242. [PMID: 26890569 PMCID: PMC4759455 DOI: 10.1371/journal.pone.0149242] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 01/28/2016] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE To investigate the inhibitory effect of Hirsutella sinensis (HS) on epithelial-mesenchymal transition (EMT) of renal tubular epithelial cells induced by aristolochic acid (AA) and its possible mechanism. METHODS 18 male Sprague-Dawley rats were randomly and equally divided into the following 3 groups: AA group, AA+HS group and control group. Urinary protein excretion and creatinine clearance (CCr) were measured. All rats were sacrificed at the end of 12th week. The pathological examination of renal tissue was performed and the mRNA and protein expression of transforming growth factor-β1 (TGF-β1), α-smooth muscle actin (α-SMA), cytokeratin-18 and Snail in renal cortex were determined by real time quantitative PCR and immunohistochemical staining respectively. In addition, human renal proximal tubule epithelial cells line (HKC) was divided into the following 4 groups: AA group, AA+HS group, HS control group and control group. The above mRNA and protein expression in HKC was determined by real time quantitative PCR and Western blot respectively. RESULTS (1) CCr was significantly decreased, and the urinary protein excretion and relative area of renal interstitial fibrosis were significantly increased in the rats of AA and AA+HS group compared to those in control group (P<0.05 or P<0.01); all the above abnormalities significantly lightened in the rats of AA+HS group compared to those in AA group (P<0.05). (2) The mRNA and protein expression of TGF-β1, α-SMA and Snail was significantly up-regulated and the expression of cytokeratin-18 was significantly down-regulated in the rat renal cortex as well as in the cultured HKC cells in AA and AA+HS groups compared to those in control group (P<0.05 or P<0.01); all the above abnormalities significantly alleviated in AA+HS group compared to those in AA group (P<0.05 or P<0.01). (3) Knockdown endogenous Snail expression by siRNA could ameliorate AA-induced EMT of HKC cells, while overexpression of Snail by plasmid transfection diminished the antagonistic effect of HS on AA-induced EMT. These results suggest Snail might be a potential target of HS effect. CONCLUSION HS is able to antagonize, to some extent, tubular EMT and renal interstitial fibrosis caused by AA, which might be related to its inhibitory effects on the TGF-β1 and Snail expression.
Collapse
Affiliation(s)
- Xiao-yi Xu
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Jing-jing Chai
- Emergency Department, Peking Union Medical College Hospital, Beijing 100730, China
| | - Yi-pu Chen
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
- * E-mail:
| | - Hong-liang Rui
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Yan-yan Wang
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Hong-rui Dong
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Yu-lin Man
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Hong Cheng
- Division of Nephrology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| |
Collapse
|
85
|
Zhu Y, Tan J, Xie H, Wang J, Meng X, Wang R. HIF-1α regulates EMT via the Snail and β-catenin pathways in paraquat poisoning-induced early pulmonary fibrosis. J Cell Mol Med 2016; 20:688-97. [PMID: 26781174 PMCID: PMC5126389 DOI: 10.1111/jcmm.12769] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 11/28/2015] [Indexed: 12/21/2022] Open
Abstract
Paraquat (PQ) poisoning‐induced pulmonary fibrosis is one of the primary causes of death in patients with PQ poisoning. Hypoxia‐inducible factor‐1α (HIF‐1α) and epithelial‐mesenchymal transition (EMT) are involved in the progression of pulmonary fibrosis. Snail and β‐catenin are two other factors involved in promoting EMT. However, the relationship among HIF‐1α, Snail and β‐catenin in PQ poisoning‐induced pulmonary fibrosis is not clear. Our research aimed to determine whether the regulation of HIF‐1α in EMT occurs via the Snail and β‐catenin pathways in PQ poisoning‐induced pulmonary fibrosis. Sixty‐six Sprague–Dawley rats were randomly and evenly divided into a control group and a PQ group. The PQ group was treated with an intragastric infusion of a 20% PQ solution (50 mg/kg) for 2, 6, 12, 24, 48 and 72 hrs. A549 and RLE‐6TN cell lines were transfected with HIF‐1α siRNA for 48 hrs before being exposed to PQ. Western blotting, real‐time quantitative PCR, immunofluorescence, immunohistochemistry and other assays were used in our research. In vivo, the protein levels of HIF‐1α and α‐SMA were increased at 2 hrs and the level of ZO‐1 (Zonula Occluden‐1) was reduced at 12 hrs. In vitro, the transient transfection of HIF‐1α siRNA resulted in a decrease in the degree of EMT. The expression levels of Snail and β‐catenin were significantly reduced when HIF‐α was silenced. These data demonstrate that EMT may be involved in PQ poisoning‐induced pulmonary fibrosis and regulated by HIF‐1α via the Snail and β‐catenin pathways. Hypoxia‐inducible factor‐1α may be a therapeutic target for the treatment of PQ poisoning‐induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Yong Zhu
- Department of Critical Care Medicine, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jiuting Tan
- Department of Critical Care Medicine, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Hui Xie
- Department of Critical Care Medicine, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jinfeng Wang
- Department of Critical Care Medicine, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xiaoxiao Meng
- Department of Critical Care Medicine, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ruilan Wang
- Department of Critical Care Medicine, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
86
|
Heijink IH, Jonker MR, de Vries M, van Oosterhout AJM, Telenga E, Ten Hacken NHT, Postma DS, van den Berge M. Budesonide and fluticasone propionate differentially affect the airway epithelial barrier. Respir Res 2016; 17:2. [PMID: 26739349 PMCID: PMC4704248 DOI: 10.1186/s12931-015-0318-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 12/23/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND COPD patients have a higher risk of pneumonia when treated with fluticasone propionate (FP) than with placebo, and a lower risk with budesonide (BUD). We hypothesized that BUD and FP differentially affect the mucosal barrier in response to viral infection and/or cigarette smoke. METHODS We assessed protective effects of equivalent concentrations of BUD and FP on cytokine production and barrier function (electrical resistance) in human bronchial epithelial 16HBE cells and primary bronchial epithelial cells (PBECs) upon exposure to viral mimetic poly-(I:C) and/or cigarette smoke extract (CSE) or epidermal growth factor (EGF). RESULTS BUD and FP were equally effective in suppressing poly-(I:C)- and/or CSE-induced IL-8 secretion in 16HBE and PBECs. Poly-(I:C) substantially decreased electrical resistance in 16HBE cells and both BUD and FP fully counteracted this effect. However, FP hardly affected 16HBE barrier dysfunction induced by CSE with/without poly-(I:C), whereas BUD (16 nM) provided full protection, an effect likely mediated by affecting EGFR-downstream target GSK-3β. Similarly, BUD, but not FP, significantly improved CSE-induced barrier dysfunction in PBECs. Finally, BUD, but not FP, exerted a modest but significant protective effect against Streptococcus Pneumoniae-induced barrier dysfunction, and BUD, but not FP, prevented cellular adhesion and/or internalization of these bacteria induced by poly-(I:C) in 16HBE. CONCLUSIONS Collectively, both BUD and FP efficiently control epithelial pro-inflammatory responses and barrier function upon mimicry of viral infection. Of potential clinical relevance, BUD more effectively counteracted CSE-induced barrier dysfunction, reinforcing the epithelial barrier and potentially limiting access of pathogens upon smoking in vivo.
Collapse
Affiliation(s)
- I H Heijink
- Department of Pathology & Medical Biology, Experimental Pulmonology and Inflammation Research, University of Groningen, University Medical Center Groningen,, Hanzeplein 1, NL-9713 GZ, Groningen, The Netherlands. .,Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands. .,University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands.
| | - M R Jonker
- Department of Pathology & Medical Biology, Experimental Pulmonology and Inflammation Research, University of Groningen, University Medical Center Groningen,, Hanzeplein 1, NL-9713 GZ, Groningen, The Netherlands
| | - M de Vries
- Department of Pathology & Medical Biology, Experimental Pulmonology and Inflammation Research, University of Groningen, University Medical Center Groningen,, Hanzeplein 1, NL-9713 GZ, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - A J M van Oosterhout
- Department of Pathology & Medical Biology, Experimental Pulmonology and Inflammation Research, University of Groningen, University Medical Center Groningen,, Hanzeplein 1, NL-9713 GZ, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - E Telenga
- Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - N H T Ten Hacken
- Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - D S Postma
- Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| | - M van den Berge
- Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, GRIAC Research Institute, Groningen, The Netherlands
| |
Collapse
|
87
|
Pattabiraman DR, Weinberg RA. Targeting the Epithelial-to-Mesenchymal Transition: The Case for Differentiation-Based Therapy. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2016; 81:11-19. [PMID: 28057845 PMCID: PMC5722631 DOI: 10.1101/sqb.2016.81.030957] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Although important strides have been made in targeted therapy for certain leukemias and subtypes of breast cancer, the standard of care for most carcinomas still involves chemotherapy, radiotherapy, surgery, or a combination of these. Two processes serve as obstacles to the successful treatment of carcinomas. First, a majority of deaths from these types of cancers occurs as a result of distant metastases and not the primary tumors themselves. Second, subsets of cells that are able to survive conventional therapy drive the aggressive relapse of the tumors, often in forms that are resistant to treatment. A frequently observed feature of malignant carcinomas is the loss of epithelial traits and the gain of certain mesenchymal ones that are programmed by the cell-biological program termed the epithelial-to-mesenchymal transition (EMT). The EMT program can confer (i) an ability to disseminate, (ii) an ability to become stem-like tumor-initiating cells, (iii) an ability to found new tumor colonies at distant anatomical sites, and (iv) an elevated resistance to therapy. These multiple powers of the EMT program explain why it has become an attractive target for therapeutic intervention. Recent work has revealed the variable nature of the EMT, with multiple versions of the program being observed depending on the tissue context and the stage of tumor progression. In this review, we attempt to crystallize emerging concepts in the research on EMT and stemness and discuss the benefits of using a differentiation-based therapeutic strategy for the eradication of stem-like populations that have adopted various versions of the EMT program.
Collapse
Affiliation(s)
| | - Robert A Weinberg
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142
- Ludwig/MIT Center for Molecular Oncology, Cambridge, Massachusetts 02139
| |
Collapse
|
88
|
Hong JY, Oh IH, McCrea PD. Phosphorylation and isoform use in p120-catenin during development and tumorigenesis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:102-14. [PMID: 26477567 DOI: 10.1016/j.bbamcr.2015.10.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Revised: 10/12/2015] [Accepted: 10/13/2015] [Indexed: 12/12/2022]
Abstract
P120-catenin is essential to vertebrate development, modulating cadherin and small-GTPase functions, and growing evidence points also to roles in the nucleus. A complexity in addressing p120-catenin's functions is its many isoforms, including optional splicing events, alternative points of translational initiation, and secondary modifications. In this review, we focus upon how choices in the initiation of protein translation, or the earlier splicing of the RNA transcript, relates to primary sequences that harbor established or putative regulatory phosphorylation sites. While certain p120 phosphorylation events arise via known kinases/phosphatases and have defined outcomes, in most cases the functional consequences are still to be established. In this review, we provide examples of p120-isoforms as they relate to phosphorylation events, and thereby to isoform dependent protein-protein associations and downstream functions. We also provide a view of upstream pathways that determine p120's phosphorylation state, and that have an impact upon development and disease. Because other members of the p120 subfamily undergo similar processing and phosphorylation, as well as related catenins of the plakophilin subfamily, what is learned regarding p120 will by extension have wide relevance in vertebrates.
Collapse
Affiliation(s)
- Ji Yeon Hong
- Division of Cardiology, Department of Medicine, Severance Biomedical Science Institute, Yonsei University College of Medicine, 50 Yonsei-ro, Seodaemun-gu, Seoul 120-752, Republic of Korea.
| | - Il-Hoan Oh
- The Catholic University of Korea, Catholic High Performance Cell Therapy Center, 505 Banpo-dong, Seocho-Ku, Seoul 137-701, Republic of Korea
| | - Pierre D McCrea
- Department of Genetics, University of Texas MD Anderson Cancer Center, University of Texas Graduate School of Biomedical Science, Houston, TX 77030, USA.
| |
Collapse
|
89
|
Kumar SR, Patel H, Tomlinson A. Wingless mediated apoptosis: How cone cells direct the death of peripheral ommatidia in the developing Drosophila eye. Dev Biol 2015; 407:183-94. [PMID: 26428511 DOI: 10.1016/j.ydbio.2015.09.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Revised: 09/25/2015] [Accepted: 09/26/2015] [Indexed: 11/24/2022]
Abstract
Morphogen gradients play pervasive roles in development, and understanding how they are established and decoded is a major goal of contemporary developmental biology. Here we examine how a Wingless (Wg) morphogen gradient patterns the peripheral specialization of the fly eye. The outermost specialization is the pigment rim; a thick band of pigment cells that circumscribes the eye and optically insulates the sides of the retina. It results from the coalescence of pigment cells that survive the death of the outermost row of developing ommatidia. We investigate here how the Wg target genes expressed in the moribund ommatidia direct the intercellular signaling, the morphogenetic movements, and ultimately the ommatidial death. A salient feature of this process is the secondary expression of the Wg morphogen elicited in the ommatidia by the primary Wg signal. We find that neither the primary nor secondary sources of Wg alone are able to promote ommatidial death, but together they suffice to drive the apoptosis. This represents an unusual gradient read-out process in which a morphogen induces its own expression in its target cells to generate a concentration spike required to push the local cellular responses to the next threshold response.
Collapse
Affiliation(s)
- Sudha R Kumar
- Department of Genetics and Development College of Physicians and Surgeons Columbia University, United States
| | - Hina Patel
- Department of Genetics and Development College of Physicians and Surgeons Columbia University, United States
| | - Andrew Tomlinson
- Department of Genetics and Development College of Physicians and Surgeons Columbia University, United States.
| |
Collapse
|
90
|
Shimatsu Y, Horii W, Nunoya T, Iwata A, Fan J, Ozawa M. Production of human apolipoprotein(a) transgenic NIBS miniature pigs by somatic cell nuclear transfer. Exp Anim 2015; 65:37-43. [PMID: 26411321 PMCID: PMC4783649 DOI: 10.1538/expanim.15-0057] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Most cases of ischemic heart disease and stroke occur as a result of atherosclerosis. The
purpose of this study was to produce a new Nippon Institute for Biological Science (NIBS)
miniature pig model by somatic cell nuclear transfer (SCNT) for studying atherosclerosis.
The human apolipoprotein(a) (apo(a)) genes were transfected into kidney epithelial cells
derived from a male and a female piglet. Male cells were used as donors initially, and 275
embryos were transferred to surrogates. Three offspring were delivered, and the production
efficiency was 1.1% (3/275). Serial female cells were injected into 937 enucleated
oocytes. Eight offspring were delivered (production efficiency: 0.9%) from surrogates. One
male and 2 female transgenic miniature pigs matured well. Lipoprotein(a) was found in the
male and one of the female transgenic animals. These results demonstrate successful
production of human apo(a) transgenic NIBS miniature pigs by SCNT. Our goal is to
establish a human apo(a) transgenic NIBS miniature pig colony for studying
atherosclerosis.
Collapse
Affiliation(s)
- Yoshiki Shimatsu
- NIBS Laboratory Animal Research Station, Nippon Institute for Biological Science, 3331-114 Kamisasao, Kobuchisawa, Hokuto, Yamanashi 408-0041, Japan
| | | | | | | | | | | |
Collapse
|
91
|
Buhrmann C, Shayan P, Kraehe P, Popper B, Goel A, Shakibaei M. Resveratrol induces chemosensitization to 5-fluorouracil through up-regulation of intercellular junctions, Epithelial-to-mesenchymal transition and apoptosis in colorectal cancer. Biochem Pharmacol 2015; 98:51-68. [PMID: 26310874 DOI: 10.1016/j.bcp.2015.08.105] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 08/20/2015] [Indexed: 12/18/2022]
Abstract
5-Fluorouracil (5-FU), a common chemotherapeutic agent used for the treatment of colorectal cancer (CRC), by itself has inadequate response rates; highlighting the need for novel and improved treatment regimens for these patients. Resveratrol, a naturally-occurring polyphenol, has been linked with chemosensitizing potential and anticancer properties; however, the underlying mechanisms for these effects remain poorly understood. The effect of resveratrol in parental CRC cell lines (HCT116, SW480) and their corresponding isogenic 5-FU-chemoresistant derived clones (HCT116R, SW480R) was examined by MTT assays, intercellular junction formation and apoptosis by electron- and immunoelectron microscopy, nuclear factor-kappaB (NF-κB) and NF-κB regulated gene products by western blot analysis in a 3D-alginate microenvironment. Resveratrol blocked the proliferation of all four CRC cell lines and synergized the invasion inhibitory effects of 5-FU. Interestingly, resveratrol induced a transition from 5-FU-induced formation of microvilli to a planar cell surface, which was concomitant with up-regulation of desmosomes, gap- and tight junctions (claudin-2) and adhesion molecules (E-cadherin) expression in HCT116 and HCT116R cells. Further, resveratrol significantly attenuated drug resistance through inhibition of epithelial-mesenchymal transition (EMT) factors (decreased vimentin and slug, increased E-cadherin) and down-regulation of NF-κB activation and its translocation to the nucleus and abolished NF-κB-regulated gene end-products (MMP-9, caspase-3). Moreover, this suppression was mediated through inhibition of IκBα kinase and IκBα phosphorylation and degradation. Our results demonstrate that resveratrol can potentiate the anti-tumor effects of 5-FU on CRC cells by chemosensitizing them, inhibiting an EMT phenotype via up-regulation of intercellular junctions and by down-regulation of NF-κB pathway.
Collapse
Affiliation(s)
- Constanze Buhrmann
- Institute of Anatomy, Ludwig-Maximilian-University Munich, Pettenkoferstrasse 11, D-80336 Munich, Germany
| | - Parviz Shayan
- Investigating Institute of Molecular Biological System Transfer, Tehran 1417863171, Iran; Department of Parasitology, Faculty of Veterinary Medicine, University of Tehran, Tehran, 141556453, Iran
| | - Patricia Kraehe
- Institute of Anatomy, Ludwig-Maximilian-University Munich, Pettenkoferstrasse 11, D-80336 Munich, Germany
| | - Bastian Popper
- Department of Anatomy and Cell Biology, Biomedical Center, Ludwig-Maximilian-University Munich, D-80336 Munich, Germany
| | - Ajay Goel
- Center for Gastrointestinal Research; Center for Epigenetics, Cancer Prevention and Cancer Genomics, Baylor Research Institute and Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas, USA
| | - Mehdi Shakibaei
- Institute of Anatomy, Ludwig-Maximilian-University Munich, Pettenkoferstrasse 11, D-80336 Munich, Germany.
| |
Collapse
|
92
|
Shirato K, Ujike M, Kawase M, Matsuyama S. Identification of CCL2, RARRES2 and EFNB2 as host cell factors that influence the multistep replication of respiratory syncytial virus. Virus Res 2015; 210:213-26. [PMID: 26277777 DOI: 10.1016/j.virusres.2015.08.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 08/10/2015] [Accepted: 08/10/2015] [Indexed: 01/20/2023]
Abstract
Human respiratory syncytial virus (RSV) is a major causative agent of respiratory tract infections in children worldwide. Preterm children or those with underlying cardiopulmonary disorders are at particularly high risk of developing severe and lethal RSV respiratory tract infections; however, there are currently no effective vaccines or anti-viral drugs. To identify targets for the development of drugs to treat RSV infections, we investigated host cell factors involved in the replication of RSV. To this end, MDCK cells with low susceptibility to RSV were transfected with cDNA libraries derived from RSV-susceptible human lung or HeLa cells. A microarray analysis was subsequently performed on parental MDCK cells and MDCK cells that were converted to an RSV-susceptible form. Among the genes identified, chemokine (C-C motif) ligand 2 (CCL2), retinoic acid receptor responder protein 2 (RARRES2) and ephrin-B2 (EFNB2) had a positive effect on RSV replication. Expression of these genes in MDCK cells resulted in a 10- to 100-fold increase in RSV replication. CCL2 expression also disrupted the distribution of claudin-1, a tight junction protein, suggesting that CCL2 plays a role in claudin-based tight junction formation during RSV replication. The knockdown of EFNB2 and RARRES2 by siRNA in RSV-susceptible cell lines (HEp-2 and A549) resulted in reduced RSV replication, suggesting that EFNB2 and RARRES2 participate in RSV replication. Together, our findings suggest that CCL2, RARRES2 and EFNB2 are host cell factors involved in RSV replication.
Collapse
Affiliation(s)
- Kazuya Shirato
- Laboratory of Acute Respiratory Viral Diseases and Cytokines, Department of Virology III, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama, Tokyo 208-0011, Japan.
| | - Makoto Ujike
- Laboratory of Virology and Viral Infections, Faculty of Veterinary Medicine, Nippon Veterinary and Life Science University, Kyonan-cho 1-7-1, Musashino, Tokyo 180-8602, Japan
| | - Miyuki Kawase
- Laboratory of Acute Respiratory Viral Diseases and Cytokines, Department of Virology III, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama, Tokyo 208-0011, Japan
| | - Shutoku Matsuyama
- Laboratory of Acute Respiratory Viral Diseases and Cytokines, Department of Virology III, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama, Tokyo 208-0011, Japan
| |
Collapse
|
93
|
Zarns K, Desell T, Nechaev S, Dhasarathy A. Searching the Human Genome for Snail and Slug With DNA@Home. PROCEEDINGS ... IEEE INTERNATIONAL CONFERENCE ON ESCIENCE. IEEE INTERNATIONAL CONFERENCE ON ESCIENCE 2015; 2015:429-438. [PMID: 26998498 PMCID: PMC4794263 DOI: 10.1109/escience.2015.27] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
DNA@Home is a volunteer computing project that aims to use Gibbs Sampling for the identification and location of DNA control signals on full genome-scale datasets. A fault tolerant and asynchronous implementation of Gibbs sampling using the Berkeley Open Infrastructure for Network Computing (BOINC) was used to identify the location of binding sites of the SNAI1 (Snail) and SNAI2 (Slug) transcription factors across the human genome. Genes regulated by Slug but not Snail, and genes regulated by Snail but not Slug provided two datasets with known motifs. These datasets contained up to 994 DNA sequences which to our knowledge is largest scale use of Gibbs sampling for discovery of binding sites. 1000 parallel sampling walks were used to search for the presence of 1, 2 or 3 possible motifs using small, medium, and full size sets of these sequences. These runs were performed over a period of two months using over 1500 volunteered computing hosts and generated over 2.2 Terabytes of sampling data. High performance computing resources were used for post processing. This paper presents intra and inter walk analyses used to determine walk convergence. The results were validated against current biological knowledge of the Snail and Slug promoter regions and present avenues for further biological study.
Collapse
Affiliation(s)
- Kristopher Zarns
- Department of Computer Science, University of North Dakota, Grand Forks, North Dakota 58202-9015
| | - Travis Desell
- Department of Computer Science, University of North Dakota, Grand Forks, North Dakota 58202-9015
| | - Sergei Nechaev
- Department of Basic Sciences, University of North Dakota, Grand Forks, North Dakota 58202-9061
| | - Archana Dhasarathy
- Department of Basic Sciences, University of North Dakota, Grand Forks, North Dakota 58202-9061
| |
Collapse
|
94
|
Haraguchi M, Sato M, Ozawa M. CRISPR/Cas9n-Mediated Deletion of the Snail 1Gene (SNAI1) Reveals Its Role in Regulating Cell Morphology, Cell-Cell Interactions, and Gene Expression in Ovarian Cancer (RMG-1) Cells. PLoS One 2015; 10:e0132260. [PMID: 26161782 PMCID: PMC4498756 DOI: 10.1371/journal.pone.0132260] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 06/11/2015] [Indexed: 01/09/2023] Open
Abstract
Snail1 is a transcription factor that induces the epithelial to mesenchymal transition (EMT). During EMT, epithelial cells lose their junctions, reorganize their cytoskeletons, and reprogram gene expression. Although Snail1 is a prominent repressor of E-cadherin transcription, its precise roles in each of the phenomena of EMT are not completely understood, particularly in cytoskeletal changes. Previous studies have employed gene knockdown systems to determine the functions of Snail1. However, incomplete protein knockdown is often associated with these systems, which may cause incorrect interpretation of the data. To more precisely evaluate the functions of Snail1, we generated a stable cell line with a targeted ablation of Snail1 (Snail1 KO) by using the CRISPR/Cas9n system. Snail1 KO cells show increased cell–cell adhesion, decreased cell–substrate adhesion and cell migration, changes to their cytoskeletal organization that include few stress fibers and abundant cortical actin, and upregulation of epithelial marker genes such as E-cadherin, occludin, and claudin-1. However, morphological changes were induced by treatment of Snail1 KO cells with TGF-beta. Other transcription factors that induce EMT were also induced by treatment with TGF-beta. The precise deletion of Snail1 by the CRISPR/Cas9n system provides clear evidence that loss of Snail1 causes changes in the actin cytoskeleton, decreases cell–substrate adhesion, and increases cell–cell adhesion. Treatment of RMG1 cells with TGF-beta suggests redundancy among the transcription factors that induce EMT.
Collapse
Affiliation(s)
- Misako Haraguchi
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
- * E-mail:
| | - Masahiro Sato
- Section of Gene Expression Regulation, Frontier Science Research Center, Kagoshima University, Kagoshima, Japan
| | - Masayuki Ozawa
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
95
|
Production of Cloned Miniature Pigs Expressing High Levels of Human Apolipoprotein(a) in Plasma. PLoS One 2015; 10:e0132155. [PMID: 26147378 PMCID: PMC4492603 DOI: 10.1371/journal.pone.0132155] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 06/10/2015] [Indexed: 11/19/2022] Open
Abstract
High lipoprotein(a) [Lp(a)] levels are a major risk factor for the development of atherosclerosis. However, because apolipoprotein(a) [apo(a)], the unique component of Lp(a), is found only in primates and humans, the study of human Lp(a) has been hampered due to the lack of appropriate animal models. Using somatic cell nuclear transfer (SCNT) techniques, we produced transgenic miniature pigs expressing human apo(a) in the plasma. First, we placed the hemagglutinin (HA)-tagged cDNA of human apo(a) under the control of the β-actin promoter and cytomegalovirus enhancer, and then introduced this construct into kidney epithelial cells. Immunostaining of cells with anti-HA antibody allowed identification of cells stably expressing apo(a); one of the positive clones was used to provide donor cells for SCNT, yielding blastocysts that expressed apo(a). Immunohistochemical analysis of tissue sections and RT-PCR analysis of total RNA from organs of cloned piglet revealed that apo(a) is expressed in various tissues/organs including heart, liver, kidney, and intestine. More importantly, a transgenic line exhibited a high level (>400 mg/dL) of Lp(a) in plasma, and the transgenic apo(a) gene was transmitted to the offspring. Thus, we generated a human apo(a)–transgenic miniature pig that can be used as a model system to study advanced atherosclerosis related to human disease. The anatomical and physiological similarities between the swine and human cardiovascular systems will make this pig model a valuable source of information on the role of apo(a) in the formation of atherosclerosis, as well as the mechanisms underlying vascular health and disease.
Collapse
|
96
|
Mei M, Xiang RL, Cong X, Zhang Y, Li J, Yi X, Park K, Han JY, Wu LL, Yu GY. Claudin-3 is required for modulation of paracellular permeability by TNF-α through ERK1/2/slug signaling axis in submandibular gland. Cell Signal 2015; 27:1915-27. [PMID: 26148935 DOI: 10.1016/j.cellsig.2015.07.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 07/01/2015] [Indexed: 02/07/2023]
Abstract
TNF-α plays an important role in the pathogenesis of salivary inflammatory diseases. Salivary dysfunction, which leads to impaired saliva secretion, can be caused by TNF-α-induced disrupted epithelial barrier. However, the signaling mechanism involved in TNF-α-modulated tight junction barrier in salivary gland remains unclear. Here, we found that TNF-α reduced transepithelial resistance (TER) and increased FITC-dextran flux in a rat submandibular cell line SMG-C6. Claudin (Cln)-3 was selectively downregulated and disrupted by TNF-α, whereas Cln-1, Cln-4, and β-catenin were not affected. Overexpression of Cln-3 retained and Cln-3 knockdown abolished the TNF-α-induced alterations. Moreover, TNF-α increased extracellular signal-regulated kinase (ERK1/2) phosphorylation and the expression of transcriptional factor slug. ERK1/2 kinase inhibitor PD98059 abrogated TNF-α-induced increase in paracellular permeability, alterations of Cln-3, and elevation of slug. Overexpression of slug decreased and slug knockdown increased Cln-3 expression. In addition, slug bind to the E-box elements of Cln-3 promoter in TNF-α-treated cells, and this response was blocked by PD98059. Furthermore, TNF-α decreased Cln-3 expression and increased slug content in cultured human submandibular gland. Taken together, our data suggest that Cln-3 plays a vital role in TNF-α-modulated paracellular permeability in submandibular epithelium and ERK1/2/slug signaling axis is involved in alteration of Cln-3 redistribution and downregulation.
Collapse
Affiliation(s)
- Mei Mei
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Ruo-Lan Xiang
- Department of Physiology and Pathophysiology, Peking University Health Science Center and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Xin Cong
- Department of Physiology and Pathophysiology, Peking University Health Science Center and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Yan Zhang
- Department of Physiology and Pathophysiology, Peking University Health Science Center and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Jing Li
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Xia Yi
- Department of Biochemistry and Molecular Biology, Peking University Health Science Center and Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Beijing 100191, China
| | - Kyungpyo Park
- Department of Physiology, School of Dentistry, Seoul National University, Seoul 110-749, South Korea
| | - Jing-Yan Han
- Department of Integration of Chinese and Western Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Li-Ling Wu
- Department of Physiology and Pathophysiology, Peking University Health Science Center and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China.
| | - Guang-Yan Yu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing 100081, China.
| |
Collapse
|
97
|
Epithelial-Mesenchymal Transition Markers β-catenin, Snail, and E-Cadherin do not Predict Disease Free Survival in Prostate Adenocarcinoma: a Prospective Study. Pathol Oncol Res 2015; 21:1209-16. [DOI: 10.1007/s12253-015-9958-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 05/26/2015] [Indexed: 11/25/2022]
|
98
|
Izawa G, Kobayashi W, Haraguchi M, Sudo A, Ozawa M. The ectopic expression of Snail in MDBK cells does not induce epithelial-mesenchymal transition. Int J Mol Med 2015; 36:166-72. [PMID: 25998899 PMCID: PMC4494602 DOI: 10.3892/ijmm.2015.2215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 04/29/2015] [Indexed: 01/06/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT), a key process in the tumor metastatic cascade, is characterized by the loss of cell-cell junctions and cell polarity, as well as by the acquisition of migratory and invasive properties. However, the precise molecular events that initiate this complex EMT process are poorly understood. Snail expression induces EMT in Madin-Darby canine kidney (MDCK) cells and the human epidermoid carcinoma cell line, A431. Snail is a zinc finger transcription factor and triggers EMT by suppressing E-cadherin expression. In the present study, to broaden our knowledge of Snail-induced EMT, we generated stable Snail transfectants using Madin-Darby bovine kidney (MDBK) cells. Contrary to the MDCK or A431 cells examined in our previous studies, the MDBK cells transfected with the Snail construct maintained an epithelial morphology and showed no sign of reduced cell-cell adhesiveness compared to the control cells. Consistent with these observations, the down-regulation of epithelial marker proteins, e.g. E-cadherin and desmoglein, and the upregulation of mesenchymal marker proteins, e.g., N-cadherin and fibronectin, were not detected. Furthermore, the E-cadherin promoter was not methylated. Therefore, in the MDBK cells, the ectopic expression of Snail failed to induce EMT. As previously demonstrated, in MDCK cells, Snail expression is accompanied by the increased expression of other EMT-inducing transcription factors, e.g., Slug and zinc finger E-box-binding homeobox 1 (ZEB1). However, the MDBK cells transfected with the Snail construct did not exhibit an increased expression of these factors. Thus, it is possible that the failure to upregulate other EMT-related transcription factors may explain the lack of Snail-mediated induction of EMT in MDBK cells.
Collapse
Affiliation(s)
- Genya Izawa
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Wakako Kobayashi
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Misako Haraguchi
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Akiharu Sudo
- Department of Sport and Physical Education, Faculty of Physical Education, Kokushikan University, Tama-shi, Tokyo 206-8515, Japan
| | - Masayuki Ozawa
- Department of Biochemistry and Molecular Biology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| |
Collapse
|
99
|
Kim BJ, Hancock BM, Bermudez A, Del Cid N, Reyes E, van Sorge NM, Lauth X, Smurthwaite CA, Hilton BJ, Stotland A, Banerjee A, Buchanan J, Wolkowicz R, Traver D, Doran KS. Bacterial induction of Snail1 contributes to blood-brain barrier disruption. J Clin Invest 2015; 125:2473-83. [PMID: 25961453 DOI: 10.1172/jci74159] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 04/06/2015] [Indexed: 12/23/2022] Open
Abstract
Bacterial meningitis is a serious infection of the CNS that results when blood-borne bacteria are able to cross the blood-brain barrier (BBB). Group B Streptococcus (GBS) is the leading cause of neonatal meningitis; however, the molecular mechanisms that regulate bacterial BBB disruption and penetration are not well understood. Here, we found that infection of human brain microvascular endothelial cells (hBMECs) with GBS and other meningeal pathogens results in the induction of host transcriptional repressor Snail1, which impedes expression of tight junction genes. Moreover, GBS infection also induced Snail1 expression in murine and zebrafish models. Tight junction components ZO-1, claudin 5, and occludin were decreased at both the transcript and protein levels in hBMECs following GBS infection, and this repression was dependent on Snail1 induction. Bacteria-independent Snail1 expression was sufficient to facilitate tight junction disruption, promoting BBB permeability to allow bacterial passage. GBS induction of Snail1 expression was dependent on the ERK1/2/MAPK signaling cascade and bacterial cell wall components. Finally, overexpression of a dominant-negative Snail1 homolog in zebrafish elevated transcription of tight junction protein-encoding genes and increased zebrafish survival in response to GBS challenge. Taken together, our data support a Snail1-dependent mechanism of BBB disruption and penetration by meningeal pathogens.
Collapse
|
100
|
IGFBP-rP1 suppresses epithelial-mesenchymal transition and metastasis in colorectal cancer. Cell Death Dis 2015; 6:e1695. [PMID: 25789970 PMCID: PMC4385937 DOI: 10.1038/cddis.2015.59] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 02/02/2015] [Accepted: 02/05/2015] [Indexed: 11/08/2022]
Abstract
Epithelial-mesenchymal transition (EMT) was initially recognized during organogenesis and has recently been reported to be involved in promoting cancer invasion and metastasis. Cooperation of transforming growth factor-β (TGF-β) and other signaling pathways, such as Ras and Wnt, is essential to inducing EMT, but the molecular mechanisms remain to be fully determined. Here, we reported that insulin-like growth factor binding protein-related protein 1 (IGFBP-rP1), a potential tumor suppressor, controls EMT in colorectal cancer progression. We revealed the inhibitory role of IGFBP-rP1 through analyses of clinical colorectal cancer samples and various EMT and metastasis models in vitro and in vivo. Moreover, we demonstrated that IGFBP-rP1 suppresses EMT and tumor metastasis by repressing TGF-β-mediated EMT through the Smad signaling cascade. These data establish that IGFBP-rP1 functions as a suppressor of EMT and metastasis in colorectal cancer.
Collapse
|