351
|
Gil D, Ciołczyk-Wierzbicka D, Dulińska-Litewka J, Laidler P. Integrin-linked kinase regulates cadherin switch in bladder cancer. Tumour Biol 2016; 37:15185-15191. [PMID: 27683053 PMCID: PMC5126188 DOI: 10.1007/s13277-016-5354-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 09/07/2016] [Indexed: 12/25/2022] Open
Abstract
Cadherin switch is specific of epithelial-mesenchymal transition (EMT) and is closely related to tumor cell invasion. However, the molecular mechanism that promotes the phenotypic changes remains unclear and elusive. We found that integrin-linked kinase (ILK) is a key factor involved in cadherin switch. The expression and activity of ILK are elevated in a variety of cancers but its mechanisms are not exactly understood. In this report, we studied the role and mechanism of ILK in EMT of human bladder cancer. We showed that silencing of ILK expression by small interfering RNA (siRNA) significantly abolished the nuclear translocation or the presence of markers associated with EMT like Snail, Twist, Zeb, and beta-catenin. ILK knockdown by siRNA suppressed N-cadherin expression and increased re-expression of E-cadherin in bladder cancer cells. We suggest that ILK is a major signaling factor involved in EMT. It is essential to understand the molecular mechanism of EMT in aim to possibly use it in search for new therapeutic targets.
Collapse
Affiliation(s)
- Dorota Gil
- Chair of Medical Biochemistry, Jagiellonian University Medical College, ul.Kopernika 7, 31-034, Kraków, Poland.
| | - Dorota Ciołczyk-Wierzbicka
- Chair of Medical Biochemistry, Jagiellonian University Medical College, ul.Kopernika 7, 31-034, Kraków, Poland
| | - Joanna Dulińska-Litewka
- Chair of Medical Biochemistry, Jagiellonian University Medical College, ul.Kopernika 7, 31-034, Kraków, Poland
| | - Piotr Laidler
- Chair of Medical Biochemistry, Jagiellonian University Medical College, ul.Kopernika 7, 31-034, Kraków, Poland
| |
Collapse
|
352
|
Huang H, Svoboda RA, Lazenby AJ, Saowapa J, Chaika N, Ding K, Wheelock MJ, Johnson KR. Up-regulation of N-cadherin by Collagen I-activated Discoidin Domain Receptor 1 in Pancreatic Cancer Requires the Adaptor Molecule Shc1. J Biol Chem 2016; 291:23208-23223. [PMID: 27605668 DOI: 10.1074/jbc.m116.740605] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinomas are highly malignant cancers characterized by extensive invasion into surrounding tissues, metastasis to distant organs, and a limited response to therapy. A main feature of pancreatic ductal adenocarcinomas is desmoplasia, which leads to extensive deposition of collagen I. We have demonstrated that collagen I can induce epithelial-mesenchymal transition (EMT) in pancreatic cancer cells. A hallmark of EMT is an increase in the expression of the mesenchymal cadherin N-cadherin. Previously we showed up-regulation of N-cadherin promotes tumor cell invasion and that collagen I-induced EMT is mediated by two collagen receptors, α2β1-integrin and discoidin domain receptor 1 (DDR1). DDR1 is a receptor-tyrosine kinase widely expressed during embryonic development and in many adult tissues and is also highly expressed in many different cancers. In the signaling pathway initiated by collagen, we have shown proline-rich tyrosine kinase 2 (Pyk2) is downstream of DDR1. In this study we found isoform b of DDR1 is responsible for collagen I-induced up-regulation of N-cadherin and tyrosine 513 of DDR1b is necessary. Knocking down Shc1, which binds to tyrosine 513 of DDR1b via its PTB (phosphotyrosine binding) domain, eliminates the up-regulation of N-cadherin. The signaling does not require a functional SH2 domain or the tyrosine residues commonly phosphorylated in Shc1 but is mediated by the interaction between a short segment of the central domain of Shc1 and the proline-rich region of Pyk2. Taken together, these data illustrate DDR1b, but not DDR1a, mediates collagen I-induced N-cadherin up-regulation, and Shc1 is involved in this process by coupling to both DDR1 and Pyk2.
Collapse
Affiliation(s)
- Huocong Huang
- From the Department of Biochemistry and Molecular Biology, College of Medicine
| | | | - Audrey J Lazenby
- Department of Pathology and Microbiology, College of Medicine, and
| | | | - Nina Chaika
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha Nebraska 68198
| | - Ke Ding
- State Key Laboratory of Respiratory Diseases, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, #190 Kaiyuan Avenue, Guangzhou 510530, China, and
| | - Margaret J Wheelock
- From the Department of Biochemistry and Molecular Biology, College of Medicine.,Department of Oral Biology, College of Dentistry.,Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha Nebraska 68198
| | - Keith R Johnson
- From the Department of Biochemistry and Molecular Biology, College of Medicine, .,Department of Oral Biology, College of Dentistry.,Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha Nebraska 68198.,Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
353
|
Lee E, Ewald ML, Sedarous M, Kim T, Weyers BW, Truong RH, Yamada S. Deletion of the cytoplasmic domain of N-cadherin reduces, but does not eliminate, traction force-transmission. Biochem Biophys Res Commun 2016; 478:1640-6. [PMID: 27596967 DOI: 10.1016/j.bbrc.2016.08.173] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 08/30/2016] [Indexed: 10/21/2022]
Abstract
Collective migration of epithelial cells is an integral part of embryonic development, wound healing, tissue renewal and carcinoma invasion. While previous studies have focused on cell-extracellular matrix adhesion as a site of migration-driving, traction force-transmission, cadherin mediated cell-cell adhesion is also capable of force-transmission. Using a soft elastomer coated with purified N-cadherin as a substrate and a Hepatocyte Growth Factor-treated, transformed MDCK epithelial cell line as a model system, we quantified traction transmitted by N-cadherin-mediated contacts. On a substrate coated with purified extracellular domain of N-cadherin, cell surface N-cadherin proteins arranged into puncta. N-cadherin mutants (either the cytoplasmic deletion or actin-binding domain chimera), however, failed to assemble into puncta, suggesting the assembly of focal adhesion like puncta requires the cytoplasmic domain of N-cadherin. Furthermore, the cytoplasmic domain deleted N-cadherin expressing cells exerted lower traction stress than the full-length or the actin binding domain chimeric N-cadherin. Our data demonstrate that N-cadherin junctions exert significant traction stress that requires the cytoplasmic domain of N-cadherin, but the loss of the cytoplasmic domain does not completely eliminate traction force transmission.
Collapse
Affiliation(s)
- Eliot Lee
- Biomedical Engineering Department, University of California, Davis, United States
| | - Makena L Ewald
- Biomedical Engineering Department, University of California, Davis, United States
| | - Mary Sedarous
- Biomedical Engineering Department, University of California, Davis, United States
| | - Timothy Kim
- Biomedical Engineering Department, University of California, Davis, United States
| | - Brent W Weyers
- Biomedical Engineering Department, University of California, Davis, United States
| | - Rose Hong Truong
- Biomedical Engineering Department, University of California, Davis, United States
| | - Soichiro Yamada
- Biomedical Engineering Department, University of California, Davis, United States.
| |
Collapse
|
354
|
Regev M, Sabanay H, Kartvelishvily E, Kam Z, Bershadsky AD. Involvement of Rho GAP GRAF1 in maintenance of epithelial phenotype. Cell Adh Migr 2016; 11:367-383. [PMID: 27588930 PMCID: PMC5569970 DOI: 10.1080/19336918.2016.1227910] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Adhesion of epithelial cell to each other and to extracellular matrix, as well as cell migration ability and cytoskeleton organization undergo significant alterations in the course of neoplastic transformation, but regulatory mechanisms involved in these processes are not fully understood. Here, we studied the role of a Rho GAP protein GRAF1 (GTPase Regulator Associated with Focal adhesion kinase-1) in the regulation of the epithelial phenotype in cells of breast derived, non-malignant, MCF10A cell line. GRAF1 was shown to be localized to cell-cell junctions, and its depletion resulted in accelerated cell migration velocity, elongation of the cells and cell colonies, impaired monolayer integrity and significant disruption of desmosomes with a loss of associated keratin filaments. These processes were accompanied by formation of larger focal adhesions, an increased number of contractile actin stress fibers, reduction in epithelial markers and increase in mesenchymal markers such as epithelial-mesenchymal transition (EMT)-specific transcription factors Snail-1 and Snail-2, as well as N-cadherin, and vimentin. Moreover, unlike control cells, GRAF1 knocked-down cells demonstrated anchorage-independent growth in soft agar. GRAF1 expression in several highly invasive breast cancer cell lines was low, as compared to the non-malignant MCF10A cells, while overexpressing of GRAF1 in the malignant BT-549 cell line led to a decrease of mesenchymal markers, especially the Snail-1 and 2. Altogether, our analysis suggests that GRAF1 plays a role in the maintenance of normal epithelial phenotype and its depletion leads to an EMT-like process that might be involved in neoplastic transformation.
Collapse
Affiliation(s)
- Miriam Regev
- a Department of Molecular Cell Biology , Weizmann Institute of Science , Rehovot , Israel.,b The Chaim Sheba Medical Center, Tel-Hashomer Hospital , Israel
| | - Helena Sabanay
- a Department of Molecular Cell Biology , Weizmann Institute of Science , Rehovot , Israel
| | - Elena Kartvelishvily
- a Department of Molecular Cell Biology , Weizmann Institute of Science , Rehovot , Israel
| | - Zvi Kam
- a Department of Molecular Cell Biology , Weizmann Institute of Science , Rehovot , Israel
| | - Alexander D Bershadsky
- a Department of Molecular Cell Biology , Weizmann Institute of Science , Rehovot , Israel.,c Mechanobiology Institute, National University of Singapore , Singapore
| |
Collapse
|
355
|
Gigantol Inhibits Epithelial to Mesenchymal Process in Human Lung Cancer Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:4561674. [PMID: 27651818 PMCID: PMC5019934 DOI: 10.1155/2016/4561674] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 07/11/2016] [Accepted: 08/01/2016] [Indexed: 12/16/2022]
Abstract
Lung cancer remains a leading public health problem as evidenced by its increasing death rate. The main cause of death in lung cancer patients is cancer metastasis. The metastatic behavior of lung cancer cells becomes enhanced when cancer cells undergo epithelial to mesenchymal transition (EMT). Gigantol, a bibenzyl compound extracted from the Thai orchid, Dendrobium draconis, has been shown to have promising therapeutic potential against cancer cells, which leads to the hypothesis that gigantol may be able to inhibit the fundamental EMT process in cancer cells. This study has demonstrated for the first time that gigantol possesses the ability to suppress EMT in non-small cell lung cancer H460 cells. Western blot analysis has revealed that gigantol attenuates the activity of ATP-dependent tyrosine kinase (AKT), thereby inhibiting the expression of the major EMT transcription factor, Slug, by both decreasing its transcription and increasing its degradation. The inhibitory effects of gigantol on EMT result in a decrease in the level of migration in H460 lung cancer cells. The results of this study emphasize the potential of gigantol for further development against lung cancer metastasis.
Collapse
|
356
|
Hahn JM, McFarland KL, Combs KA, Supp DM. Partial epithelial-mesenchymal transition in keloid scars: regulation of keloid keratinocyte gene expression by transforming growth factor-β1. BURNS & TRAUMA 2016; 4:30. [PMID: 27574697 PMCID: PMC4994224 DOI: 10.1186/s41038-016-0055-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 07/04/2016] [Indexed: 12/17/2022]
Abstract
Background Keloids are an extreme form of abnormal scarring that result from a pathological fibroproliferative wound healing process. The molecular mechanisms driving keloid pathology remain incompletely understood, hindering development of targeted, effective therapies. Recent studies in our laboratory demonstrated that keloid keratinocytes exhibit adhesion abnormalities and display a transcriptional signature reminiscent of cells undergoing epithelial-mesenchymal transition (EMT), suggesting a role for EMT in keloid pathology. In the current study, we further define the EMT-like phenotype of keloid scars and investigate regulation of EMT-related genes in keloid. Methods Primary keratinocytes from keloid scar and normal skin were cultured in the presence or absence of transforming growth factor beta 1 (TGF-β1) +/− inhibitors of TGF-β1 and downstream signaling pathways. Gene expression was measured using quantitative polymerase chain reaction. Migration was analyzed using an in vitro wound healing assay. Proteins in keloid scar and normal skin sections were localized by immunohistochemistry. Statistical analyses utilized SigmaPlot (SyStat Software, San Jose, CA) or SAS® (SAS Institute, Cary, NC). Results In keloid and normal keratinocytes, TGF-β1 regulated expression of EMT-related genes, including hyaluronan synthase 2, vimentin, cadherin-11, wingless-type MMTV integration site family, member 5A, frizzled 7, ADAM metallopeptidase domain 19, and interleukin-6. Inhibition of canonical TGF-β1 signaling in keloid keratinocytes significantly inhibited expression of these genes, and TGF-β1 stimulation of normal keratinocytes increased their expression. The inhibition of the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway or the p38 mitogen-activated protein kinase pathway attenuated TGF-β1-induced expression of subsets of these genes. Migration of keloid keratinocytes, previously shown to be increased compared with normal keratinocytes, was significantly reduced by inhibition of TGF-β1 or ERK1/2 signaling. Biomarkers of EMT, including reduced E-cadherin and increased active β-catenin, were observed in keloid epidermis in vivo. However, evidence of basement membrane breakdown in keloid scar was not observed. Conclusions The results suggest that keloid keratinocytes exist in an EMT-like metastable state, similar to activated keratinocytes in healing wounds. The EMT-like gene expression pattern of keloid keratinocytes is regulated by canonical and non-canonical TGF-β1 signaling pathways. Therefore, interventions targeting TGF-β1-regulated EMT-like gene expression in keloid keratinocytes may serve to suppress keloid scarring. Electronic supplementary material The online version of this article (doi:10.1186/s41038-016-0055-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jennifer M Hahn
- Research Department, Shriners Hospitals for Children - Cincinnati, Cincinnati, OH USA
| | - Kevin L McFarland
- Research Department, Shriners Hospitals for Children - Cincinnati, Cincinnati, OH USA
| | - Kelly A Combs
- Research Department, Shriners Hospitals for Children - Cincinnati, Cincinnati, OH USA
| | - Dorothy M Supp
- Research Department, Shriners Hospitals for Children - Cincinnati, Cincinnati, OH USA ; Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH USA
| |
Collapse
|
357
|
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
|
358
|
Stone RC, Pastar I, Ojeh N, Chen V, Liu S, Garzon KI, Tomic-Canic M. Epithelial-mesenchymal transition in tissue repair and fibrosis. Cell Tissue Res 2016; 365:495-506. [PMID: 27461257 DOI: 10.1007/s00441-016-2464-0] [Citation(s) in RCA: 422] [Impact Index Per Article: 46.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 06/24/2016] [Indexed: 12/28/2022]
Abstract
The epithelial-mesenchymal transition (EMT) describes the global process by which stationary epithelial cells undergo phenotypic changes, including the loss of cell-cell adhesion and apical-basal polarity, and acquire mesenchymal characteristics that confer migratory capacity. EMT and its converse, MET (mesenchymal-epithelial transition), are integral stages of many physiologic processes and, as such, are tightly coordinated by a host of molecular regulators. Converging lines of evidence have identified EMT as a component of cutaneous wound healing, during which otherwise stationary keratinocytes (the resident skin epithelial cells) migrate across the wound bed to restore the epidermal barrier. Moreover, EMT plays a role in the development of scarring and fibrosis, as the matrix-producing myofibroblasts arise from cells of the epithelial lineage in response to injury but are pathologically sustained instead of undergoing MET or apoptosis. In this review, we summarize the role of EMT in physiologic repair and pathologic fibrosis of tissues and organs. We conclude that further investigation into the contribution of EMT to the faulty repair of fibrotic wounds might identify components of EMT signaling as common therapeutic targets for impaired healing in many tissues. Graphical Abstract Model for injury-triggered EMT activation in physiologic wound repair (left) and fibrotic wound healing (right).
Collapse
Affiliation(s)
- Rivka C Stone
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, RMSB, Room 2023A, Miami, FL 33136, USA
- The Research Residency Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Fla., USA
| | - Irena Pastar
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, RMSB, Room 2023A, Miami, FL 33136, USA
| | - Nkemcho Ojeh
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, RMSB, Room 2023A, Miami, FL 33136, USA
- Faculty of Medical Sciences, The University of the West Indies, Bridgetown, Barbados
| | - Vivien Chen
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, RMSB, Room 2023A, Miami, FL 33136, USA
| | - Sophia Liu
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, RMSB, Room 2023A, Miami, FL 33136, USA
| | - Karen I Garzon
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, RMSB, Room 2023A, Miami, FL 33136, USA
| | - Marjana Tomic-Canic
- Wound Healing and Regenerative Medicine Research Program, Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, 1600 NW 10th Avenue, RMSB, Room 2023A, Miami, FL 33136, USA.
| |
Collapse
|
359
|
Targeting Epithelial-Mesenchymal Transition (EMT) to Overcome Drug Resistance in Cancer. Molecules 2016; 21:molecules21070965. [PMID: 27455225 PMCID: PMC6273543 DOI: 10.3390/molecules21070965] [Citation(s) in RCA: 565] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 07/16/2016] [Accepted: 07/19/2016] [Indexed: 12/24/2022] Open
Abstract
Epithelial–mesenchymal transition (EMT) is known to play an important role in cancer progression, metastasis and drug resistance. Although there are controversies surrounding the causal relationship between EMT and cancer metastasis, the role of EMT in cancer drug resistance has been increasingly recognized. Numerous EMT-related signaling pathways are involved in drug resistance in cancer cells. Cells undergoing EMT show a feature similar to cancer stem cells (CSCs), such as an increase in drug efflux pumps and anti-apoptotic effects. Therefore, targeting EMT has been considered a novel opportunity to overcome cancer drug resistance. This review describes the mechanism by which EMT contributes to drug resistance in cancer cells and summarizes new advances in research in EMT-associated drug resistance.
Collapse
|
360
|
Kolijn K, Verhoef EI, van Leenders GJLH. Morphological and immunohistochemical identification of epithelial-to-mesenchymal transition in clinical prostate cancer. Oncotarget 2016; 6:24488-98. [PMID: 26041890 PMCID: PMC4695200 DOI: 10.18632/oncotarget.4177] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/02/2015] [Indexed: 12/22/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a process known to be associated with aggressive tumor behavior, metastasis and treatment resistance. It is characterized by coincidental upregulation of mesenchymal markers such as vimentin, fibronectin and N-cadherin concurrent with E-cadherin downregulation. Studies on EMT are generally performed in cell lines and mouse models, while the histopathological and phenotypical properties in clinical prostate cancer (PCa) are still unclear. The objective of this study was to identify EMT in PCa patients. We demonstrated that N-cadherin, vimentin and fibronectin were generally not co-expressed in corresponding tumor regions. Immunofluorescent double stainings confirmed that co-expression of mesenchymal markers was uncommon, as we found no prostate cancer cells that co-expressed N-cadherin with fibronectin and only rare (<1%) cells that co-expressed N-cadherin with vimentin. Downregulation of E-cadherin was demonstrated in all N-cadherin positive tumor cells, but not in vimentin or fibronectin positive tumor cells. We further analyzed N-cadherin expression in morphologically distinct PCa growth patterns in a radical prostatectomy cohort (n = 77) and found that N-cadherin is preferentially expressed in ill-defined Gleason grade 4 PCa. In conclusion, we demonstrate that N-cadherin is the most reliable marker for EMT in clinical PCa and is preferentially expressed in ill-defined Gleason grade 4 growth pattern.
Collapse
Affiliation(s)
- Kimberley Kolijn
- Department of Pathology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Esther I Verhoef
- Department of Pathology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | | |
Collapse
|
361
|
Lindsay AJ, McCaffrey MW. Rab coupling protein mediated endosomal recycling of N-cadherin influences cell motility. Oncotarget 2016; 8:104717-104732. [PMID: 29285208 PMCID: PMC5739595 DOI: 10.18632/oncotarget.10513] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 05/13/2016] [Indexed: 12/18/2022] Open
Abstract
Rab coupling protein (RCP) is a Rab GTPase effector that functions in endosomal recycling. The RCP gene is frequently amplified in breast cancer, leading to increased cancer aggressiveness. Furthermore, RCP enhances the motility of ovarian cancer cells by coordinating the recycling of α5β1 integrin and EGF receptor to the leading edge of migrating cells. Here we report that RCP also influences the motility of lung adenocarcinoma cells. Knockdown of RCP inhibits the motility of A549 cells in 2D and 3D migration assays, while its overexpression enhances migration in these assays. Depletion of RCP leads to a reduction in N-cadherin protein levels, which could be restored with lysosomal inhibitors. Trafficking assays revealed that RCP knockdown inhibits the return of endocytosed N-cadherin to the cell surface. We propose that RCP regulates the endosomal recycling of N-cadherin, and in its absence N-cadherin is diverted to the degradative pathway. The increased aggressiveness of tumour cells that overexpress RCP may be due to biased recycling of N-cadherin in metastatic cancer cells.
Collapse
Affiliation(s)
- Andrew J Lindsay
- Molecular Cell Biology Laboratory, School of Biochemistry and Cell Biology, Biosciences Institute, University College Cork, Cork, Ireland
| | - Mary W McCaffrey
- Molecular Cell Biology Laboratory, School of Biochemistry and Cell Biology, Biosciences Institute, University College Cork, Cork, Ireland
| |
Collapse
|
362
|
Abstract
During cardiac trabeculation, cardiomyocytes delaminate from the outermost (compact) layer to form complex muscular structures known as trabeculae. As these cardiomyocytes delaminate, the remodeling of adhesion junctions must be tightly coordinated so cells can extrude from the compact layer while remaining in tight contact with their neighbors. In this study, we examined the distribution of N-cadherin (Cdh2) during cardiac trabeculation in zebrafish. By analyzing the localization of a Cdh2-EGFP fusion protein expressed under the control of the zebrafish cdh2 promoter, we initially observed Cdh2-EGFP expression along the lateral sides of embryonic cardiomyocytes, in an evenly distributed pattern, and with the occasional appearance of punctae. Within a few hours, Cdh2-EGFP distribution on the lateral sides of cardiomyocytes evolves into a clear punctate pattern as Cdh2-EGFP molecules outside the punctae cluster to increase the size of these aggregates. In addition, Cdh2-EGFP molecules also appear on the basal side of cardiomyocytes that remain in the compact layer. Delaminating cardiomyocytes accumulate Cdh2-EGFP on the surface facing the basal side of compact layer cardiomyocytes, thereby allowing tight adhesion between these layers. Importantly, we find that blood flow/cardiac contractility is required for the transition from an even distribution of Cdh2-EGFP to the formation of punctae. Furthermore, using time-lapse imaging of beating hearts in conjunction with a Cdh2 tandem fluorescent protein timer transgenic line, we observed that Cdh2-EGFP molecules appear to move from the lateral to the basal side of cardiomyocytes along the cell membrane, and that Erb-b2 receptor tyrosine kinase 2 (Erbb2) function is required for this relocalization.
Collapse
|
363
|
Ninsontia C, Phiboonchaiyanan PP, Chanvorachote P. Zinc induces epithelial to mesenchymal transition in human lung cancer H460 cells via superoxide anion-dependent mechanism. Cancer Cell Int 2016; 16:48. [PMID: 27330411 PMCID: PMC4912812 DOI: 10.1186/s12935-016-0323-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 06/06/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Epithelial to mesenchymal transition (EMT) has been shown to be a crucial enhancing mechanism in the process of cancer metastasis, as it increases cancer cell capabilities to migrate, invade and survive in circulating systems. This study aimed to investigate the effect of essential element zinc on EMT characteristics in lung cancer cells. METHODS The effect of zinc on EMT was evaluated by determining the EMT behaviors using migration, invasion and colony formation assay. EMT markers were examined by western blot analysis. Reactive oxygen species (ROS) were detected by specific fluorescence dyes and flow cytometry. All results were analyzed by ANOVA, followed by individual comparisons with post hoc test. RESULTS The present study has revealed for the first time that the zinc could induce EMT and related metastatic behaviors in lung cancer cells. Results showed that treatment of the cells with zinc resulted in the significant increase of EMT markers N-cadherin, vimentin, snail and slug and decrease of E-cadherin proteins. Zinc-treated cells exhibited the mesenchymal-like morphology and increased cancer cell motility with significant increase of activated FAK, Rac1, and RhoA. Also, tumorigenic abilities of lung cancer cells could be enhanced by zinc. Importantly, the underlying mechanism was found to be caused by the ability of zinc to generate intracellular superoxide anion. Zinc was shown to induce cellular superoxide anion generation and the up-regulation of EMT markers and the induced cell migration and invasion in zinc-treated cells could be attenuated by the treatment of MnTBAP, a specific superoxide anion inhibitor. CONCLUSION Knowledge gains from this study may highlight the roles of this important element in the regulation of EMT and cancer metastasis and fulfill the understanding in the area of cancer cell biology.
Collapse
Affiliation(s)
- Chuanpit Ninsontia
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Pathumwan, Bangkok, 10330 Thailand ; Cell-based Drug and Health Products Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Preeyaporn Plaimee Phiboonchaiyanan
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Pathumwan, Bangkok, 10330 Thailand ; Cell-based Drug and Health Products Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Pithi Chanvorachote
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Pathumwan, Bangkok, 10330 Thailand ; Cell-based Drug and Health Products Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
364
|
Smithers CC, Overduin M. Structural Mechanisms and Drug Discovery Prospects of Rho GTPases. Cells 2016; 5:E26. [PMID: 27304967 PMCID: PMC4931675 DOI: 10.3390/cells5020026] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 05/28/2016] [Accepted: 06/07/2016] [Indexed: 12/25/2022] Open
Abstract
Rho GTPases regulate cellular morphology and dynamics, and some are key drivers of cancer progression. This superfamily offers attractive potential targets for therapeutic intervention, with RhoA, Rac1 and Cdc42 being prime examples. The challenges in developing agents that act on these signaling enzymes include the lack of obvious druggable pockets and their membrane-bound activities. However, progress in targeting the similar Ras protein is illuminating new strategies for specifically inhibiting oncogenic GTPases. The structures of multiple signaling and regulatory states of Rho proteins have been determined, and the post-translational modifications including acylation and phosphorylation points have been mapped and their functional effects examined. The development of inhibitors to probe the significance of overexpression and mutational hyperactivation of these GTPases underscores their importance in cancer progression. The ability to integrate in silico, in vitro, and in vivo investigations of drug-like molecules indicates the growing tractability of GTPase systems for lead optimization. Although no Rho-targeted drug molecules have yet been clinically approved, this family is clearly showing increasing promise for the development of precision medicine and combination cancer therapies.
Collapse
Affiliation(s)
- Cameron C Smithers
- Department of Biochemistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada.
| | - Michael Overduin
- Department of Biochemistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada.
| |
Collapse
|
365
|
Yoshida T, Okuyama H, Nakayama M, Endo H, Tomita Y, Nonomura N, Nishimura K, Inoue M. Dynamic Change in p63 Protein Expression during Implantation of Urothelial Cancer Clusters. Neoplasia 2016; 17:574-85. [PMID: 26297435 PMCID: PMC4547408 DOI: 10.1016/j.neo.2015.07.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 07/10/2015] [Accepted: 07/13/2015] [Indexed: 01/12/2023] Open
Abstract
Although the dissemination of urothelial cancer cells is supposed to be a major cause of the multicentricity of urothelial tumors, the mechanism of implantation has not been well investigated. Here, we found that cancer cell clusters from the urine of patients with urothelial cancer retain the ability to survive, grow, and adhere. By using cell lines and primary cells collected from multiple patients, we demonstrate that △ Np63α protein in cancer cell clusters was rapidly decreased through proteasomal degradation when clusters were attached to the matrix, leading to downregulation of E-cadherin and upregulation of N-cadherin. Decreased △ Np63α protein level in urothelial cancer cell clusters was involved in the clearance of the urothelium. Our data provide the first evidence that clusters of urothelial cancer cells exhibit dynamic changes in △ Np63α expression during attachment to the matrix, and decreased △ Np63α protein plays a critical role in the interaction between cancer cell clusters and the urothelium. Thus, because △ Np63α might be involved in the process of intraluminal dissemination of urothelial cancer cells, blocking the degradation of △ Np63α could be a target of therapy to prevent the dissemination of urothelial cancer.
Collapse
Affiliation(s)
- Takahiro Yoshida
- Department of Biochemistry, Osaka Medical Center for Cancer and Cardiovascular Diseases; Department of Urology, Osaka University Graduate School of Medicine
| | - Hiroaki Okuyama
- Department of Biochemistry, Osaka Medical Center for Cancer and Cardiovascular Diseases
| | - Masashi Nakayama
- Department of Urology, Osaka Medical Center for Cancer and Cardiovascular Diseases
| | - Hiroko Endo
- Department of Biochemistry, Osaka Medical Center for Cancer and Cardiovascular Diseases
| | - Yasuhiko Tomita
- Department of Pathology, Osaka Medical Center for Cancer and Cardiovascular Diseases
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine
| | - Kazuo Nishimura
- Department of Urology, Osaka Medical Center for Cancer and Cardiovascular Diseases
| | - Masahiro Inoue
- Department of Biochemistry, Osaka Medical Center for Cancer and Cardiovascular Diseases; Department of Clinical and Experimental Pathophysiology, Osaka University Graduate School of Pharmaceutical Sciences.
| |
Collapse
|
366
|
Li B, Shi H, Wang F, Hong D, Lv W, Xie X, Cheng X. Expression of E-, P- and N-Cadherin and Its Clinical Significance in Cervical Squamous Cell Carcinoma and Precancerous Lesions. PLoS One 2016; 11:e0155910. [PMID: 27223886 PMCID: PMC4880319 DOI: 10.1371/journal.pone.0155910] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/08/2016] [Indexed: 01/08/2023] Open
Abstract
Aberrant expression of classical cadherins has been observed in tumor invasion and metastasis, but its involvement in cervical carcinogenesis and cancer progression is not clear. We investigated E-, P- and N-cadherin expression and its significance in cervical squamous cell carcinoma (SCC) and cervical intraepithelial neoplasia (CIN). This retrospective study enrolled 508 patients admitted to Women's Hospital, School of Medicine, Zhejiang University with cervical lesions between January 2006 and December 2010. Immunochemical staining was performed in 98 samples of normal cervical epithelium (NC), 283 of CIN, and 127 of early-stage SCC. The association of cadherin staining with clinical characteristics and survival of the patients was evaluated by univariate and multivariate analysis. We found gradients of decreasing E-cadherin expression and increasing P-cadherin expression from NC through CIN to SCC. Aberrant E-cadherin and P-cadherin expression were significantly associated with clinical parameters indicating poor prognosis and shorter patient survival. Interestingly, we found very low levels of positive N-cadherin expression in CIN and SCC tissues that were not related to CIN or cancer. Pearson chi-square tests showed that E-cadherin expression in SCC was inversely correlated with P-cadherin expression (E-P switch), and was not correlated with N-cadherin expression. More important, patients with tissues exhibiting an E-P switch in expression had highly aggressive phenotypes and poorer prognosis than those without E-P switch expression. Our findings suggest that E-cadherin and P-cadherin, but not N-cadherin staining, might be useful in diagnosing CIN and for predicting prognosis in patients with early-stage SCC.
Collapse
Affiliation(s)
- Baohua Li
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haiyan Shi
- Department of Pathology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Fenfen Wang
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Die Hong
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Weiguo Lv
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Women’s Reproductive Health Key Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xing Xie
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Women’s Reproductive Health Key Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaodong Cheng
- Department of Gynecologic Oncology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Women’s Reproductive Health Key Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- * E-mail:
| |
Collapse
|
367
|
Belmonte JM, Clendenon SG, Oliveira GM, Swat MH, Greene EV, Jeyaraman S, Glazier JA, Bacallao RL. Virtual-tissue computer simulations define the roles of cell adhesion and proliferation in the onset of kidney cystic disease. Mol Biol Cell 2016; 27:3673-3685. [PMID: 27193300 PMCID: PMC5221598 DOI: 10.1091/mbc.e16-01-0059] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 05/10/2016] [Indexed: 12/15/2022] Open
Abstract
Virtual-tissue modeling is used to model emergent cyst growth in polycystic kidney disease. Model predictions, confirmed experimentally, show that decreased cell adhesion is necessary to produce stalk cysts, and loss of contact inhibition causes saccular cysts. Virtual-tissue modeling can be used to fully explore cell- and tissue-based behaviors. In autosomal dominant polycystic kidney disease (ADPKD), cysts accumulate and progressively impair renal function. Mutations in PKD1 and PKD2 genes are causally linked to ADPKD, but how these mutations drive cell behaviors that underlie ADPKD pathogenesis is unknown. Human ADPKD cysts frequently express cadherin-8 (cad8), and expression of cad8 ectopically in vitro suffices to initiate cystogenesis. To explore cell behavioral mechanisms of cad8-driven cyst initiation, we developed a virtual-tissue computer model. Our simulations predicted that either reduced cell–cell adhesion or reduced contact inhibition of proliferation triggers cyst induction. To reproduce the full range of cyst morphologies observed in vivo, changes in both cell adhesion and proliferation are required. However, only loss-of-adhesion simulations produced morphologies matching in vitro cad8-induced cysts. Conversely, the saccular cysts described by others arise predominantly by decreased contact inhibition, that is, increased proliferation. In vitro experiments confirmed that cell–cell adhesion was reduced and proliferation was increased by ectopic cad8 expression. We conclude that adhesion loss due to cadherin type switching in ADPKD suffices to drive cystogenesis. Thus, control of cadherin type switching provides a new target for therapeutic intervention.
Collapse
Affiliation(s)
- Julio M Belmonte
- Biocomplexity Institute, Physics Department, Indiana University, Bloomington, IN 47405
| | - Sherry G Clendenon
- Biocomplexity Institute, Physics Department, Indiana University, Bloomington, IN 47405
| | - Guilherme M Oliveira
- Biocomplexity Institute, Physics Department, Indiana University, Bloomington, IN 47405
| | - Maciej H Swat
- Biocomplexity Institute, Physics Department, Indiana University, Bloomington, IN 47405
| | - Evan V Greene
- Division of Nephrology, Richard L. Roudebush VA Medical Center, and Indiana University School of Medicine, Indianapolis, IN 46202
| | - Srividhya Jeyaraman
- Biocomplexity Institute, Physics Department, Indiana University, Bloomington, IN 47405
| | - James A Glazier
- Biocomplexity Institute, Physics Department, Indiana University, Bloomington, IN 47405
| | - Robert L Bacallao
- Division of Nephrology, Richard L. Roudebush VA Medical Center, and Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
368
|
Plutoni C, Bazellières E, Gauthier-Rouvière C. P-cadherin-mediated Rho GTPase regulation during collective cell migration. Small GTPases 2016; 7:156-63. [PMID: 27152729 PMCID: PMC5003545 DOI: 10.1080/21541248.2016.1173772] [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] [Indexed: 10/29/2022] Open
Abstract
This commentary addresses the role of P-cadherin in collective cell migration (CCM), a cooperative and coordinated migration mode, used by cells during normal and pathological migration processes. We discuss how cadherin-mediated cell-cell junctions (CCJs) play a critical role in CCM through their ability to regulate Rho GTPase-dependent pathways and how this leads to the generation and orientation of mechanical forces. We will also highlight the key function of P-cadherin (a poor prognostic marker in several tumors) in promoting collective cell movement in epithelial and mesenchymal cells.
Collapse
Affiliation(s)
- Cédric Plutoni
- a Institute for Research in Immunology and Cancer, Université de Montréal , Montréal , Québec , Canada
| | | | | |
Collapse
|
369
|
Alaee M, Danesh G, Pasdar M. Plakoglobin Reduces the in vitro Growth, Migration and Invasion of Ovarian Cancer Cells Expressing N-Cadherin and Mutant p53. PLoS One 2016; 11:e0154323. [PMID: 27144941 PMCID: PMC4856367 DOI: 10.1371/journal.pone.0154323] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 04/12/2016] [Indexed: 12/20/2022] Open
Abstract
Aberrant expression of cadherins and catenins plays pivotal roles in ovarian cancer development and progression. Plakoglobin (PG, γ-catenin) is a paralog of β-catenin with dual adhesive and signaling functions. While β-catenin has known oncogenic function, PG generally acts as a tumor/metastasis suppressor. We recently showed that PG interacted with p53 and that its growth/metastasis inhibitory function may be mediated by this interaction. Very little is known about the role of PG in ovarian cancer. Here, we investigated the in vitro tumor/metastasis suppressor effects of PG in ovarian cancer cell lines with mutant p53 expression and different cadherin profiles. We showed that the N-cadherin expressing and E-cadherin and PG deficient ES-2 cells were highly migratory and invasive, whereas OV-90 cells that express E-cadherin, PG and very little/no N-cadherin were not. Exogenous expression of PG or E-cadherin or N-cadherin knockdown in ES-2 cells (ES-2-E-cad, ES-2-PG and ES-2-shN-cad) significantly reduced their migration and invasion. Also, PG expression or N-cadherin knockdown significantly decreased ES-2 cells growth. Furthermore, PG interacted with both cadherins and with wild type and mutant p53 in normal ovarian and ES-2-PG cell lines, respectively.
Collapse
Affiliation(s)
- Mahsa Alaee
- Department of Oncology, University of Alberta, Edmonton, AB, T6G1Z2, Canada
| | - Ghazal Danesh
- Department of Oncology, University of Alberta, Edmonton, AB, T6G1Z2, Canada
| | - Manijeh Pasdar
- Department of Oncology, University of Alberta, Edmonton, AB, T6G1Z2, Canada
- * E-mail:
| |
Collapse
|
370
|
Frismantiene A, Dasen B, Pfaff D, Erne P, Resink TJ, Philippova M. T-cadherin promotes vascular smooth muscle cell dedifferentiation via a GSK3β-inactivation dependent mechanism. Cell Signal 2016; 28:516-530. [DOI: 10.1016/j.cellsig.2016.02.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 02/12/2016] [Accepted: 02/18/2016] [Indexed: 11/24/2022]
|
371
|
Zhao L, Jiang R, Xu M, Zhu P, Mo XM, Wang N, Chen GG, Liu ZM. Concomitant high expression of BRAFV600E, P-cadherin and cadherin 6 is associated with High TNM stage and lymph node metastasis in conventional papillary thyroid carcinoma. Clin Endocrinol (Oxf) 2016; 84:748-55. [PMID: 26285159 DOI: 10.1111/cen.12878] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 08/10/2015] [Accepted: 08/12/2015] [Indexed: 11/28/2022]
Abstract
CONTEXT AND OBJECTIVE BRAFV600E mutation is the most common activating mutation associated with aggressive behaviours in human tumours including conventional papillary thyroid carcinoma (cPTC). P-cadherin and cadherin 6 have been shown to be mesenchymal-associated cadherins and promote cancer cell invasion and metastasis. The purpose of this study was to examine BRAFV600E, P-cadherin and cadherin 6 expressions in cPTC and to assess the association of their expression with clinicopathological indicators. METHODS BRAFV600E, P-cadherin and cadherin 6 protein expressions in 80 cPTCs, 61 nodular hyperplasia and 76 normal thyroid tissues were examined by immunohistochemistry. The correlation of their protein expression with clinicopathological indicators of cPTC was statistically analysed. RESULTS Protein expression of BRAFV600E, P-cadherin and cadherin 6 was upregulated in cPTC. High protein expression of BRAFV600E, P-cadherin and cadherin 6 was significantly correlated with high TNM stage and lymph node metastasis (LNM) (P < 0·001). Furthermore, BRAFV600E, P-cadherin and cadherin 6 protein expressions were correlated with one another. BRAFV600E high expression combined with both P-cadherin and cadherin-6 high expressions had stronger correlation with high TNM stage and LNM when compared with BRAFV600E high expression combined with either P-cadherin or cadherin-6 high expression (P = 0·042, 0·017 for TNM stage and P = 0·003, 0·006 for LNM, respectively) and only BRAFV600E high expression (P < 0·001 for both TNM stage and LNM). CONCLUSIONS Concomitant high expression of BRAFV600E, P-cadherin and cadherin 6 is strongly associated with high TNM stage and LNM in cPTC.
Collapse
Affiliation(s)
- Le Zhao
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Rong Jiang
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Man Xu
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Ping Zhu
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Xiao-Mei Mo
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Ni Wang
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - George G Chen
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin N.T., Hong Kong, China
| | - Zhi-Min Liu
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| |
Collapse
|
372
|
Lowe A, Harris R, Bhansali P, Cvekl A, Liu W. Intercellular Adhesion-Dependent Cell Survival and ROCK-Regulated Actomyosin-Driven Forces Mediate Self-Formation of a Retinal Organoid. Stem Cell Reports 2016; 6:743-756. [PMID: 27132890 PMCID: PMC4939656 DOI: 10.1016/j.stemcr.2016.03.011] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 03/30/2016] [Accepted: 03/31/2016] [Indexed: 02/06/2023] Open
Abstract
In this study we dissected retinal organoid morphogenesis in human embryonic stem cell (hESC)-derived cultures and established a convenient method for isolating large quantities of retinal organoids for modeling human retinal development and disease. Epithelialized cysts were generated via floating culture of clumps of Matrigel/hESCs. Upon spontaneous attachment and spreading of the cysts, patterned retinal monolayers with tight junctions formed. Dispase-mediated detachment of the monolayers and subsequent floating culture led to self-formation of retinal organoids comprising patterned neuroretina, ciliary margin, and retinal pigment epithelium. Intercellular adhesion-dependent cell survival and ROCK-regulated actomyosin-driven forces are required for the self-organization. Our data supports a hypothesis that newly specified neuroretina progenitors form characteristic structures in equilibrium through minimization of cell surface tension. In long-term culture, the retinal organoids autonomously generated stratified retinal tissues, including photoreceptors with ultrastructure of outer segments. Our system requires minimal manual manipulation, has been validated in two lines of human pluripotent stem cells, and provides insight into optic cup invagination in vivo. Established a method for isolating large amounts of retinal organoids from hESCs Dispase-mediated cell detachment led to self-formation of the retinal organoids Intercellular adhesions in the floating cultures are required for cell survival ROCK-regulated actomyosin-driven forces are required for the self-organization
Collapse
Affiliation(s)
- Albert Lowe
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Raven Harris
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Punita Bhansali
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ales Cvekl
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Wei Liu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
373
|
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
|
374
|
Guan X, Xu M, Millar SE, Bartlett JD. Beta-catenin is essential for ameloblast movement during enamel development. Eur J Oral Sci 2016; 124:221-7. [PMID: 26957367 DOI: 10.1111/eos.12261] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2016] [Indexed: 12/21/2022]
Abstract
Beta-catenin is a multifunctional protein that plays key roles in cadherin-based cell adherens junctions and in the Wnt signaling pathway. The canonical Wnt/β-catenin pathway can regulate transcription factors that control cell movement/invasion. We investigated whether β-catenin regulates ameloblast movement through canonical Wnt signaling. The morphological and physical properties of enamel were assessed in enamel from control and β-catenin conditional knockout (cKO) mice. Ameloblast-lineage cells (ALC) were used to investigate the potential roles of β-catenin in cell migration and in E-cadherin expression. Compared with controls, incisors from β-catenin cKO mice were short, blunt, and where enamel was present, it was soft and malformed. Scanning electron microscopy revealed a dysplastic rod pattern within the enamel of incisors from β-catenin cKO mice, and Vickers microhardness measurements confirmed that mice with β-catenin ablated from their enamel organ had enamel that was significantly softer than normal. Amelogenesis was disrupted in the absence of β-catenin and the ameloblasts did not differentiate properly. We further demonstrated that migration of ALCs was inhibited in vitro and that E-cadherin expression was significantly up-regulated when ALCs were treated with the β-catenin inhibitor, ICG-001. Beta-catenin ablation causes enamel malformation in mice and this phenotype may occur, in part, by a lack of ameloblast differentiation and/or movement necessary to form the decussating enamel rod structure.
Collapse
Affiliation(s)
- Xiaomu Guan
- Department of Mineralized Tissue Biology and Harvard School of Dental Medicine, Forsyth Institute, Cambridge, MA, USA
| | - Mingang Xu
- Departments of Dermatology and Cell & Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah E Millar
- Departments of Dermatology and Cell & Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John D Bartlett
- Division of Biosciences, College of Dentistry, Ohio State University, Columbus, OH, USA
| |
Collapse
|
375
|
The interplay between microRNAs and Twist1 transcription factor: a systematic review. Tumour Biol 2016; 37:7007-19. [PMID: 26880587 DOI: 10.1007/s13277-016-4960-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 02/02/2016] [Indexed: 12/11/2022] Open
Abstract
Twist1 (also known as Twist) is a transcription factor that belongs to the family of basic helix-loop-helix (bHLH) proteins. It functions as a negative regulator of epithelial gene expression and a positive regulator of mesenchymal gene expression, thereby leading to induction of the epithelial mesenchymal transition (EMT), a process in which epithelial cells acquire the motile and migratory characteristics of mesenchymal cells. In addition to regulating the expression of protein-coding genes, Twist1 regulates the expression of microRNAs (miRNAs), adding a regulatory layer to EMT induction. Interestingly, the mRNA of Twist1 represents a downstream target of miRNAs, indicating an intricate network between miRNAs and Twist1. This network was shown to play multiple roles in cancer cell migration, invasion, and metastasis. The network can induce angiogenesis, protect cells from oncogene-induced apoptosis and senescence, enhance cancer cell resistance to conventional therapies, and increase cancer stem cell (CSC) populations. Recently, miRNAs have attracted considerable attention as potential promising tools in cancer therapies. Thus, this systematic review was conducted to clarify the reciprocal link between Twist1 and miRNAs in order to provide potential candidate miRNAs for diagnostic and therapeutic approaches in cancer treatment.
Collapse
|
376
|
Iwata E, Hasegawa T, Takeda D, Ueha T, Kawamoto T, Akisue T, Sakai Y, Komori T. Transcutaneous carbon dioxide suppresses epithelial-mesenchymal transition in oral squamous cell carcinoma. Int J Oncol 2016; 48:1493-8. [PMID: 26846904 DOI: 10.3892/ijo.2016.3380] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 01/05/2016] [Indexed: 11/06/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most common form of oral cancers. Recent studies have shown that the malignant transformation of various carcinomas, including OSCC, is associated with epithelial-mesenchymal transition (EMT), and that expression of the EMT factors are significantly associated with tumor invasion, tumor metastasis, and survival rates in OSCC patients. Hence, there is a possibility that EMT suppression may improve the prognosis of OSCC patients. Hypoxia inducible factor-1α (HIF-1α) is a crucial microenvironmental factor in tumor progression, which induces the expression of EMT factors. We previously reported that transcutaneous CO2 suppresses both human OSCC tumor growth and metastasis to the regional lymph nodes by improving hypoxia in treated tissue. According to this background, we hypothesized that increased EMT with HIF-1α expression may increase the progression and the metastatic potential of OSCC, and that decreased hypoxia by transcutaneous CO2 could suppress EMT. In the present study, in vitro studies showed that hypoxic conditions increased the expression of HIF-1α and EMT factors in OSCC cells. In addition, in vivo studies revealed that transcutaneous CO2 increased E-cadherin expression with the decreased expression of HIF-1α, Snail, Slug, N-cadherin, and Vimentin in tumor treatment. These results suggest that transcutaneous CO2 could suppress EMT by improving hypoxia, resulting in the reduction of metastatic potential of OSCC. The findings indicate that transcutaneous CO2 may be able to improve the prognosis of OSCC patients through the suppression of EMT.
Collapse
Affiliation(s)
- Eiji Iwata
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takumi Hasegawa
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Daisuke Takeda
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takeshi Ueha
- Division of Rehabilitation Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Teruya Kawamoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Toshihiro Akisue
- Division of Rehabilitation Medicine, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Yoshitada Sakai
- Division of Rehabilitation Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takahide Komori
- Department of Oral and Maxillofacial Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
377
|
Chauvet N, Romanò N, Meunier AC, Galibert E, Fontanaud P, Mathieu MN, Osterstock G, Osterstock P, Baccino E, Rigau V, Loiseau H, Bouillot-Eimer S, Barlier A, Mollard P, Coutry N. Combining Cadherin Expression with Molecular Markers Discriminates Invasiveness in Growth Hormone and Prolactin Pituitary Adenomas. J Neuroendocrinol 2016; 28:12352. [PMID: 26686489 DOI: 10.1111/jne.12352] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 11/24/2015] [Accepted: 12/15/2015] [Indexed: 01/06/2023]
Abstract
Although growth hormone (GH)- and prolactin (PRL)-secreting pituitary adenomas are considered benign, in many patients, tumour growth and/or invasion constitute a particular challenge. In other tumours, progression relies in part on dysfunction of intercellular adhesion mediated by the large family of cadherins. In the present study, we have explored the contribution of cadherins in GH and PRL adenoma pathogenesis, and evaluated whether this class of adherence molecules was related to tumour invasiveness. We have first established, by quantitative polymerase chain reaction and immunohistochemistry, the expression profile of classical cadherins in the normal human pituitary gland. We show that the cadherin repertoire is restricted and cell-type specific. Somatotrophs and lactotrophs express mainly E-cadherin and cadherin 18, whereas N-cadherin is present in the other endocrine cell types. This repertoire undergoes major differential modification in GH and PRL tumours: E-cadherin is significantly reduced in invasive GH adenomas, and this loss is associated with a cytoplasmic relocalisation of cadherin 18 and catenins. In invasive prolactinomas, E-cadherin distribution is altered and is accompanied by a mislocalisation of cadherin 18, β-catenin and p120 catenin. Strikingly, de novo expression of N-cadherin is present in a subset of adenomas and cells exhibit a mesenchymal phenotype exclusively in invasive tumours. Binary tree analysis, performed by combining the cadherin repertoire with the expression of a subset of known molecular markers, shows that cadherin/catenin complexes play a significant role in discrimination of tumour invasion.
Collapse
Affiliation(s)
- N Chauvet
- UMR-5203, CNRS, Institut de Génomique Fonctionnelle, Montpellier, France
- U1191, INSERM, Montpellier, France
- UMR-5203, Université de Montpellier, Montpellier, France
| | - N Romanò
- UMR-5203, CNRS, Institut de Génomique Fonctionnelle, Montpellier, France
- U1191, INSERM, Montpellier, France
- UMR-5203, Université de Montpellier, Montpellier, France
| | - A-C Meunier
- UMR-5203, CNRS, Institut de Génomique Fonctionnelle, Montpellier, France
- U1191, INSERM, Montpellier, France
- UMR-5203, Université de Montpellier, Montpellier, France
| | - E Galibert
- UMR-5203, CNRS, Institut de Génomique Fonctionnelle, Montpellier, France
- U1191, INSERM, Montpellier, France
- UMR-5203, Université de Montpellier, Montpellier, France
| | - P Fontanaud
- UMR-5203, CNRS, Institut de Génomique Fonctionnelle, Montpellier, France
- U1191, INSERM, Montpellier, France
- UMR-5203, Université de Montpellier, Montpellier, France
| | - M-N Mathieu
- UMR-5203, CNRS, Institut de Génomique Fonctionnelle, Montpellier, France
- U1191, INSERM, Montpellier, France
- UMR-5203, Université de Montpellier, Montpellier, France
| | - G Osterstock
- UMR-5203, CNRS, Institut de Génomique Fonctionnelle, Montpellier, France
- U1191, INSERM, Montpellier, France
- UMR-5203, Université de Montpellier, Montpellier, France
| | - P Osterstock
- Service de Médecine Légale, Hôpital Lapeyronie, CHU Montpellier, Montpellier, France
| | - E Baccino
- Service de Médecine Légale, Hôpital Lapeyronie, CHU Montpellier, Montpellier, France
| | - V Rigau
- Laboratoire d'Anatomie et Cytologie Pathologiques, Hôpital Gui de Chauliac, CHU Montpellier, Montpellier, France
| | - H Loiseau
- Service de Neurochirurgie, CHU Bordeaux, Site Pellegrin, Université de Bordeaux, Bordeaux, France
| | - S Bouillot-Eimer
- Service de Pathologie, CHU Bordeaux, Site Pellegrin, Université de Bordeaux, Bordeaux, France
| | - A Barlier
- Faculté de Médecine, CRN2M-UMR 7286, CNRS, Université Aix-Marseille, Marseille, France
- Laboratoire de Biologie Moléculaire, AP-HM, Hôpital de la Conception, Marseille, France
| | - P Mollard
- UMR-5203, CNRS, Institut de Génomique Fonctionnelle, Montpellier, France
- U1191, INSERM, Montpellier, France
- UMR-5203, Université de Montpellier, Montpellier, France
| | - N Coutry
- UMR-5203, CNRS, Institut de Génomique Fonctionnelle, Montpellier, France
- U1191, INSERM, Montpellier, France
- UMR-5203, Université de Montpellier, Montpellier, France
| |
Collapse
|
378
|
Ortiz A, Fuchs SY. Anti-metastatic functions of type 1 interferons: Foundation for the adjuvant therapy of cancer. Cytokine 2016; 89:4-11. [PMID: 26822709 DOI: 10.1016/j.cyto.2016.01.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 01/19/2016] [Accepted: 01/20/2016] [Indexed: 01/08/2023]
Abstract
The anti-tumorigenic effects that type 1 interferons (IFN1) elicited in the in vitro studies prompted consideration of IFN1 as a potent candidate for clinical treatment. Though not all patients responded to IFN1, clinical trials have shown that patients with high risk melanoma, a highly refractory solid malignancy, benefit greatly from intermediate IFN1 treatment in regards to relapse-free and distant-metastasis-free survival. The mechanisms by which IFN1 treatment at early stages of disease suppress tumor recurrence or metastatic incidence are not fully understood. Intracellular IFN1 signaling is known to affect cell differentiation, proliferation, and apoptosis. Moreover, recent studies have revealed specific IFN1-regulated genes that may contribute to IFN1-mediated suppression of cancer progression and metastasis. In concert, expression of these different IFN1 stimulated genes may impede numerous mechanisms that mediate metastatic process. Though, IFN1 treatment is still utilized as part of standard care for metastatic melanoma (alone or in combination with other therapies), cancers find the ways to develop insensitivity to IFN1 treatment allowing for unconstrained disease progression. To determine how and when IFN1 treatment would be most efficacious during disease progression, we must understand how IFN1 signaling affects different metastasis steps. Here, we specifically focus on the anti-metastatic role of endogenous IFN1 and parameters that may help to use pharmaceutical IFN1 in the adjuvant treatment to prevent cancer recurrence and metastatic disease.
Collapse
Affiliation(s)
- Angélica Ortiz
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Serge Y Fuchs
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
379
|
Xiong J, Yang H, Luo W, Shan E, Liu J, Zhang F, Xi T, Yang J. The anti-metastatic effect of 8-MOP on hepatocellular carcinoma is potentiated by the down-regulation of bHLH transcription factor DEC1. Pharmacol Res 2016; 105:121-33. [PMID: 26808085 DOI: 10.1016/j.phrs.2016.01.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 01/08/2016] [Accepted: 01/19/2016] [Indexed: 01/22/2023]
Abstract
Despite progress in diagnostics and treatment of hepatocellular carcinoma (HCC), its prognosis remains poor. 8-Methoxypsoralen (8-MOP), a formerly considered photosensitizing agent, has been reported to induce cell apoptosis in HepG2 cells in a modest way when used alone. In this study, it was demonstrated that 8-MOP inhibited HCC HepG2 cells and SMMC-7721 cells migratory and invasive potentiality, as well as modulated the expression of various EMT-associated genes such as enhancing E-cadherin and reducing N-cadherin, vimentin, α-SMA and MMP9 in a concentration-dependent way. Differentiated embryonic chondrocyte-expressed gene 1, DEC1 (BHLHE40/Stra13/Sharp2), is a basic helix-loop-helix (bHLH) transcription factor that regulates cell growth, differentiation, apoptosis and tumorigenesis. 8-MOP suppressed the expression of DEC1 in a concentration- and time-dependent manner. Overexpression of DEC1 endorsed the HepG2 cells a higher metastatic phenotype, while totally abolished 8-MOP-repressed metastatic capability. In the meanwhile, overexpression of DEC1 promoted EMT process by suppressing expression of epithelial protein and enhancing expression of mesenchymal proteins, while potently antagonized the regulation of EMT-associated genes by 8-MOP. In vivo experiments revealed that the treatment of 8-MOP (5 or 20mg/kg) resulted in a dose-dependent decreases in the lung metastasis of hepatoma H22-transplanted mice without any obvious toxicity to the organs, as well as increased expression of E-cadherin in lung tissues. Consistently, 8-MOP down-regulated the expression of DEC1 in the lungs of tumor-bearing mice, which further confirms that DEC1 was correlated with 8-MOP-induced anti-metastatic effect. The present findings establish a function for DEC1 in HCC metastatic progression and suggest its candidacy as a novel target for the anti-metastasis effect of 8-MOP.
Collapse
Affiliation(s)
- Jing Xiong
- Department of Pharmacology, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, China
| | - Huan Yang
- Research Center of Biotechnology, School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu 210009, China
| | - Wenjing Luo
- Research Center of Biotechnology, School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu 210009, China
| | - Enfang Shan
- Department of Pharmacology, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, China
| | - Jie Liu
- Department of Pharmacology, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, China
| | - Feng Zhang
- Research Center of Biotechnology, School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu 210009, China
| | - Tao Xi
- Research Center of Biotechnology, School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, Jiangsu 210009, China.
| | - Jian Yang
- Department of Pharmacology, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, China.
| |
Collapse
|
380
|
Reginato A, Girolami D, Menchetti L, Foiani G, Mandara M. E-cadherin, N-cadherin Expression and Histologic Characterization of Canine Choroid Plexus Tumors. Vet Pathol 2016; 53:788-91. [DOI: 10.1177/0300985815620844] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Choroid plexus tumors (CPTs) are reported with an increasing incidence in dogs, and they call for a reexamination of histologic features and criteria of classification corresponding to their biological behavior. In this study, the human World Health Organization classification was applied to 16 canine CPTs, and the expression of molecules involved in neoplastic cell adhesion (E-cadherin, N-cadherin), invasion (doublecortin), and proliferation (Ki-67) was investigated. Mitotic index was found to be the main criterion for grading CPTs. Cell density and multilayering of papillae were also statistically associated with histologic grade. Intraventricular spread and parenchymal invasion was observed for tumors showing histologic benign features. E-cadherin was expressed in all CPT grades, independent of tumor invasion. N-cadherin immunolabeling was more expressed in grade I than high-grade CPTs, whereas doublecortin expression was not detected in CPTs. An increasing proliferative activity was observed in relation with histologic grade.
Collapse
Affiliation(s)
- A. Reginato
- Department of Veterinary Medicine, University of Perugia, Italy
| | - D. Girolami
- Department of Veterinary Medicine, University of Perugia, Italy
| | - L. Menchetti
- Department of Veterinary Medicine, University of Perugia, Italy
| | - G. Foiani
- Department of Veterinary Medicine, University of Perugia, Italy
| | - M.T. Mandara
- Department of Veterinary Medicine, University of Perugia, Italy
| |
Collapse
|
381
|
Ma J, Zhao J, Lu J, Wang P, Feng H, Zong Y, Ou B, Zheng M, Lu A. Cadherin-12 enhances proliferation in colorectal cancer cells and increases progression by promoting EMT. Tumour Biol 2016; 37:9077-88. [PMID: 26762412 PMCID: PMC4990612 DOI: 10.1007/s13277-015-4555-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Accepted: 11/30/2015] [Indexed: 12/17/2022] Open
Abstract
Cadherin-12 (CDH12) is a subtype of N-cadherin family. In this study, we investigated the expression of CDH12 and the role of CDH12 in prognosis of colorectal cancer (CRC) patients. In addition, we observed the influence of CDH12 on proliferation and progression of CRC cell lines. By using immunohistochemical staining, we analyzed CRC samples and adjacent non-tumor tissues collected from 78 patients who underwent laparoscopic surgery in Shanghai Minimally Invasive Center, China. Statistical analyses were used to analyze relationship between CDH12 and tumor features. Kaplan-Meier method was used to analyze patients' survival. Proliferation ability of CRC cells was tested by CCK-8 assay, and transwell assays were performed to detect migration and invasion ability. Western blot assay was performed to investigate epithelial-mesenchymal transition (EMT) variants. We found that expression of CDH12 in tumor tissue was higher than in adjacent normal tissue. High expression of CDH12 was associated with tumor invasion depth and predicts poor prognosis of CRC patients. Ectopic/repressing expression of CDH12 increased/decreased the proliferation and migration ability of CRC cells. CDH12 is able to increase cancer cell migration and invasion via promoting EMT by targeting transcriptional factor Snail. These findings may conclude that CDH12 may act as a predictor in CRC patients' prognosis and an oncogene in CRC cell proliferation and migration. CDH12 may influence CRC cell progression through promoting EMT by targeting Snail. In addition, CDH12 is promoted by MCP1 through induction of MCPIP.
Collapse
Affiliation(s)
- Junjun Ma
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Jingkun Zhao
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Institute of Digestive Surgery, Shanghai, People's Republic of China
| | - Jun Lu
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China.,Department of General Surgery, Hangzhou First People's Hospital, Hangzhou, Zhejiang, People's Republic of China
| | - Puxiongzhi Wang
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Institute of Digestive Surgery, Shanghai, People's Republic of China
| | - Hao Feng
- Department of Surgery, Munich University, Munich, Germany
| | - Yaping Zong
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China
| | - Baochi Ou
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China.,Shanghai Institute of Digestive Surgery, Shanghai, People's Republic of China
| | - Minhua Zheng
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China.
| | - Aiguo Lu
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
382
|
Mandal M, Ghosh B, Anura A, Mitra P, Pathak T, Chatterjee J. Modeling continuum of epithelial mesenchymal transition plasticity. Integr Biol (Camb) 2016; 8:167-76. [PMID: 26762753 DOI: 10.1039/c5ib00219b] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Living systems respond to ambient pathophysiological changes by altering their phenotype, a phenomenon called 'phenotypic plasticity'. This program contains information about adaptive biological dynamism. Epithelial-mesenchymal transition (EMT) is one such process found to be crucial in development, wound healing, and cancer wherein the epithelial cells with restricted migratory potential develop motile functions by acquiring mesenchymal characteristics. In the present study, phase contrast microscopy images of EMT induced HaCaT cells were acquired at 24 h intervals for 96 h. The expression study of relevant pivotal molecules viz. F-actin, vimentin, fibronectin and N-cadherin was carried out to confirm the EMT process. Cells were intuitively categorized into five distinct morphological phenotypes. A population of 500 cells for each temporal point was selected to quantify their frequency of occurrence. The plastic interplay of cell phenotypes from the observations was described as a Markovian process. A model was formulated empirically using simple linear algebra, to depict the possible mechanisms of cellular transformation among the five phenotypes. This work employed qualitative, semi-quantitative and quantitative tools towards illustration and establishment of the EMT continuum. Thus, it provides a newer perspective to understand the embedded plasticity across the EMT spectrum.
Collapse
Affiliation(s)
- Mousumi Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | | | | | | | | | | |
Collapse
|
383
|
Abba ML, Patil N, Leupold JH, Allgayer H. MicroRNA Regulation of Epithelial to Mesenchymal Transition. J Clin Med 2016; 5:jcm5010008. [PMID: 26784241 PMCID: PMC4730133 DOI: 10.3390/jcm5010008] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 12/18/2015] [Accepted: 01/05/2016] [Indexed: 02/07/2023] Open
Abstract
Epithelial to mesenchymal transition (EMT) is a central regulatory program that is similar in many aspects to several steps of embryonic morphogenesis. In addition to its physiological role in tissue repair and wound healing, EMT contributes to chemo resistance, metastatic dissemination and fibrosis, amongst others. Classically, the morphological change from epithelial to mesenchymal phenotype is characterized by the appearance or loss of a group of proteins which have come to be recognized as markers of the EMT process. As with all proteins, these molecules are controlled at the transcriptional and translational level by transcription factors and microRNAs, respectively. A group of developmental transcription factors form the backbone of the EMT cascade and a large body of evidence shows that microRNAs are heavily involved in the successful coordination of mesenchymal transformation and vice versa, either by suppressing the expression of different groups of transcription factors, or otherwise acting as their functional mediators in orchestrating EMT. This article dissects the contribution of microRNAs to EMT and analyzes the molecular basis for their roles in this cellular process. Here, we emphasize their interaction with core transcription factors like the zinc finger enhancer (E)-box binding homeobox (ZEB), Snail and Twist families as well as some pluripotency transcription factors.
Collapse
Affiliation(s)
- Mohammed L Abba
- Department of Experimental Surgery, Center for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Ruprecht Karl University of Heidelberg, Ludolf-Krehl-Str. 6, 68135 Mannheim, Germany.
| | - Nitin Patil
- Department of Experimental Surgery, Center for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Ruprecht Karl University of Heidelberg, Ludolf-Krehl-Str. 6, 68135 Mannheim, Germany.
| | - Jörg Hendrik Leupold
- Department of Experimental Surgery, Center for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Ruprecht Karl University of Heidelberg, Ludolf-Krehl-Str. 6, 68135 Mannheim, Germany.
| | - Heike Allgayer
- Department of Experimental Surgery, Center for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, Ruprecht Karl University of Heidelberg, Ludolf-Krehl-Str. 6, 68135 Mannheim, Germany.
| |
Collapse
|
384
|
Seo JH, Hirata M, Kakinoki S, Yamaoka T, Yui N. Dynamic polyrotaxane-coated surface for effective differentiation of mouse induced pluripotent stem cells into cardiomyocytes. RSC Adv 2016. [DOI: 10.1039/c6ra03967g] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Increasing molecular mobility of hydrated polyrotaxane (PRX)-coated surfaces was effective to promote the differentiation of mouse induced pluripotent stem cells (iPS cells) into cardiomyocytes.
Collapse
Affiliation(s)
- Ji-Hun Seo
- Institute of Biomaterials and Bioengineering
- Tokyo Medical and Dental University
- Tokyo 101-0062
- Japan
- Department of Materials Science and Engineering
| | - Mitsuhi Hirata
- Department of Biomedical Engineering
- National Cerebral and Cardiovascular Center Research Institute
- Osaka 565-8565
- Japan
- JST-CREST
| | - Sachiro Kakinoki
- Department of Biomedical Engineering
- National Cerebral and Cardiovascular Center Research Institute
- Osaka 565-8565
- Japan
- JST-CREST
| | - Tetsuji Yamaoka
- Department of Biomedical Engineering
- National Cerebral and Cardiovascular Center Research Institute
- Osaka 565-8565
- Japan
- JST-CREST
| | - Nobuhiko Yui
- Institute of Biomaterials and Bioengineering
- Tokyo Medical and Dental University
- Tokyo 101-0062
- Japan
- JST-CREST
| |
Collapse
|
385
|
Bryan RT. Cell adhesion and urothelial bladder cancer: the role of cadherin switching and related phenomena. Philos Trans R Soc Lond B Biol Sci 2015; 370:20140042. [PMID: 25533099 DOI: 10.1098/rstb.2014.0042] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cadherins are mediators of cell-cell adhesion in epithelial tissues. E-cadherin is a known tumour suppressor and plays a central role in suppressing the invasive phenotype of cancer cells. However, the abnormal expression of N- and P-cadherin ('cadherin switching', CS) has been shown to promote a more invasive and m̀alignant phenotype of cancer, with P-cadherin possibly acting as a key mediator of invasion and metastasis in bladder cancer. Cadherins are also implicated in numerous signalling events related to embryonic development, tissue morphogenesis and homeostasis. It is these wide ranging effects and the serious implications of CS that make the cadherin cell adhesion molecules and their related pathways strong candidate targets for the inhibition of cancer progression, including bladder cancer. This review focuses on CS in the context of bladder cancer and in particular the switch to P-cadherin expression, and discusses other related molecules and phenomena, including EpCAM and the development of the cancer stem cell phenotype.
Collapse
Affiliation(s)
- Richard T Bryan
- School of Cancer Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
386
|
Blaschuk OW. N-cadherin antagonists as oncology therapeutics. Philos Trans R Soc Lond B Biol Sci 2015; 370:20140039. [PMID: 25533096 DOI: 10.1098/rstb.2014.0039] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The cell adhesion molecule (CAM), N-cadherin, has emerged as an important oncology therapeutic target. N-cadherin is a transmembrane glycoprotein mediating the formation and structural integrity of blood vessels. Its expression has also been documented in numerous types of poorly differentiated tumours. This CAM is involved in regulating the proliferation, survival, invasiveness and metastasis of cancer cells. Disruption of N-cadherin homophilic intercellular interactions using peptide or small molecule antagonists is a promising novel strategy for anti-cancer therapies. This review discusses: the discovery of N-cadherin, the mechanism by which N-cadherin promotes cell adhesion, the role of N-cadherin in blood vessel formation and maintenance, participation of N-cadherin in cancer progression, the different types of N-cadherin antagonists and the use of N-cadherin antagonists as anti-cancer drugs.
Collapse
Affiliation(s)
- Orest W Blaschuk
- Division of Urology, Department of Surgery, McGill University, Montreal, Quebec, Canada H3A 1A1
| |
Collapse
|
387
|
Liu H, Gu Y, Qi J, Han C, Zhang X, Bi C, Yu W. Inhibition of E-cadherin/catenin complex formation by O-linked N-acetylglucosamine transferase is partially independent of its catalytic activity. Mol Med Rep 2015; 13:1851-60. [PMID: 26707622 DOI: 10.3892/mmr.2015.4718] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 11/24/2015] [Indexed: 11/06/2022] Open
Abstract
p120-catenin (p120) contains a large central armadillo repeat domain, via which it binds to E‑cadherin to stabilize the latter, thereby regulating cell‑to‑cell adhesion. A previous study by our group demonstrated that O‑linked N‑acetylglucosamine (O‑GlcNAc) is involved in the regulation of the interaction between p120 and E‑cadherin. As O‑GlcNAc transferase (OGT) is able to directly bind to the majority of its target proteins, the present study hypothesized that OGT may additionally regulate the formation of the E‑cadherin/catenin complex independent of its catalytic activity. To verify this hypothesis, a catalytically inactive OGT mutant was expressed in H1299 cells, and its effects on the formation of the E‑cadherin/catenin complex were assessed. A cytoskeleton‑binding protein extraction assay confirmed that OGT inhibited the formation of the E‑cadherin/catenin complex independent of its catalytic activity. In addition, co‑immunoprecipitation and pull‑down assays were used to evaluate the interaction between OGT and p120. Immunoblotting indicated that OGT was able to directly bind to p120. To determine the domain of p120 involved in binding to OGT, a series of deletion mutants of p120 were constructed and subjected to protein binding assays by pull‑down assays. Immunoblotting showed that OGT bound to the regulatory and armadillo domains of p120, which might interfere with the interaction between p120 and E‑cadherin. Finally, OGT, p120 and E‑cadherin cytoplasmic domains (ECD) were recombinantly expressed in BL21 (DE3) recombinant E. coli cells, and a glutathione S‑transferase (GST) pull‑down assay was performed to assess the interactions among the purified recombinant proteins. Immunoblotting indicated that maltose‑binding protein (MBP)‑OGT inhibited the binding of His‑p120 to GST‑ECD in a dose‑dependent manner. All of these results suggested that OGT inhibited the formation of the E‑cadherin/catenin complex through reducing the interaction between p120 and E‑cadherin. The present study provided a novel underlying mechanism of the regulation of the interaction between p120 and E‑cadherin, and thus E‑cadherin‑mediated cell‑cell adhesion, which has essential roles in cancer development and progression.
Collapse
Affiliation(s)
- Haiyan Liu
- Department of Glycobiology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, P.R. China
| | - Yuchao Gu
- Department of Glycobiology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, P.R. China
| | - Jieqiong Qi
- Department of Glycobiology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, P.R. China
| | - Cuifang Han
- Department of Glycobiology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, P.R. China
| | - Xinling Zhang
- Department of Glycobiology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, P.R. China
| | - Chuanlin Bi
- Department of Glycobiology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, P.R. China
| | - Wengong Yu
- Department of Glycobiology, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
388
|
Fei F, Zhang D, Yang Z, Wang S, Wang X, Wu Z, Wu Q, Zhang S. The number of polyploid giant cancer cells and epithelial-mesenchymal transition-related proteins are associated with invasion and metastasis in human breast cancer. J Exp Clin Cancer Res 2015; 34:158. [PMID: 26702618 PMCID: PMC4690326 DOI: 10.1186/s13046-015-0277-8] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 12/18/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Previously, we reported that polyploid giant cancer cells (PGCCs) induced by cobalt chloride (CoCl2) could have generated daughter cells with strong invasiveness and migration capabilities via asymmetric divisions. This study compared the expression of epithelial-mesenchymal transition (EMT)-related proteins, including E-cadherin, N-cadherin, and vimentin, between PGCCs and their daughter cells, and control breast cancer cell lines MCF-7 and MDA-MB-231. The clinicopathological significance of EMT-related protein expression in human breast cancer was analyzed. METHODS Western blot was used to compare the expression levels of E-cadherin, N-cadherin, and vimentin in breast cancer lines MCF-7 and MDA-MB-231, between PGCCs with budding daughter cells and control breast cancer cells. Furthermore, 167 paraffin-embedded breast tumor tissue samples were analyzed, including samples obtained from 52 patients with primary breast cancer with lymph node metastasis (group I) and their corresponding lymph node metastatic tumors (group II), 52 patients with primary breast cancer without metastasis (group III), and 11 patients with benign breast lesions (group IV). The number of PGCCs was compared among these four groups. RESULTS The number of PGCCs increased with the malignant grade of breast tumor. Group IIhad the highest number of PGCCs and the differences among group I, II, III and IV had statistically significance (P =0.000). In addition, the expression of E-cadherin (P = 0.000), N-cadherin (P = 0.000), and vimentin (P = 0.000) was significantly different among the four groups. Group II exhibited the highest expression levels of N-cadherin and vimentin and the lowest expression levels of E-cadherin. CONCLUSIONS These data suggest that the number of PGCCs and the EMT-related proteins E-cadherin, N-cadherin, and vimentin may be valuable biomarkers to assess metastasis in patients with breast cancer.
Collapse
Affiliation(s)
- Fei Fei
- Department of Pathology, Anhui Medical University, Hefei, Anhui, 230032, People's Republic of China.
| | - Dan Zhang
- Department of Pathology, Tianjin Union Medicine Center, Tianjin, 300121, P.R China.
| | - Zhengduo Yang
- Department of Pathology, Tianjin Union Medicine Center, Tianjin, 300121, P.R China.
| | - Shujing Wang
- Department of Pathology, Anhui Medical University, Hefei, Anhui, 230032, People's Republic of China.
| | - Xian Wang
- Department of Pathology, Anhui Medical University, Hefei, Anhui, 230032, People's Republic of China.
| | - Zhengsheng Wu
- Department of Pathology, Anhui Medical University, Hefei, Anhui, 230032, People's Republic of China.
| | - Qiang Wu
- Department of Pathology, Anhui Medical University, Hefei, Anhui, 230032, People's Republic of China.
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medicine Center, Tianjin, 300121, P.R China.
| |
Collapse
|
389
|
Abstract
High-mobility group nucleosome-binding domain 5 (HMGN5) is a new member of the high-mobility group N (HMGN) protein family that is involved in nucleosomal binding and transcriptional activation. It was first discovered in mouse, and recent studies found that the expressions of HMGN5 in many human cancers were also highly regulated, such as prostate, bladder, breast, and lung and clear cell renal cell carcinoma. Numerous reports have demonstrated that HMGN5 plays significant roles in many biological and pathological conditions, such as in developmental defects, hypersensitivity to stress, embryonic stem cell differentiation, and tumor progression. Importantly, deficiency of HMGN5 has been shown to be linked to cancer cell growth, cell cycle regulation, migration, invasion, and clinical outcomes, and it represents a promising therapeutic target for many malignant tumors. In the present review, we provide an overview of the current knowledge concerning the role of HMGN5 in cancer development and progression.
Collapse
|
390
|
Fridriksdottir AJ, Kim J, Villadsen R, Klitgaard MC, Hopkinson BM, Petersen OW, Rønnov-Jessen L. Propagation of oestrogen receptor-positive and oestrogen-responsive normal human breast cells in culture. Nat Commun 2015; 6:8786. [PMID: 26564780 PMCID: PMC4660059 DOI: 10.1038/ncomms9786] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 10/04/2015] [Indexed: 02/07/2023] Open
Abstract
Investigating the susceptibility of oestrogen receptor-positive (ERpos) normal human breast epithelial cells (HBECs) for clinical purposes or basic research awaits a proficient cell-based assay. Here we set out to identify markers for isolating ERpos cells and to expand what appear to be post-mitotic primary cells into exponentially growing cultures. We report a robust technique for isolating ERpos HBECs from reduction mammoplasties by FACS using two cell surface markers, CD166 and CD117, and an intracellular cytokeratin marker, Ks20.8, for further tracking single cells in culture. We show that ERpos HBECs are released from growth restraint by small molecule inhibitors of TGFβ signalling, and that growth is augmented further in response to oestrogen. Importantly, ER signalling is functionally active in ERpos cells in extended culture. These findings open a new avenue of experimentation with normal ERpos HBECs and provide a basis for understanding the evolution of human breast cancer. Culturing normal primary breast cells that express the oestrogen receptor is difficult. Here, the authors isolate oestrogen receptor positive normal breast cells using the cell surface markers CD166 and CD117, and show that the cultures can be repeatedly passaged and retain oestrogen receptor protein expression.
Collapse
Affiliation(s)
- Agla J Fridriksdottir
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark.,Danish Stem Cell Centre, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Jiyoung Kim
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark.,Danish Stem Cell Centre, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - René Villadsen
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark.,Danish Stem Cell Centre, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Marie Christine Klitgaard
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark.,Danish Stem Cell Centre, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark.,Department of Biology, University of Copenhagen, DK-2100 Copenhagen Ø, Denmark
| | - Branden M Hopkinson
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark.,Danish Stem Cell Centre, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Ole William Petersen
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark.,Danish Stem Cell Centre, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Lone Rønnov-Jessen
- Department of Biology, University of Copenhagen, DK-2100 Copenhagen Ø, Denmark
| |
Collapse
|
391
|
Tong GH, Tong WW, Qin XS, Lu LP, Liu Y. DBD-F induces apoptosis in gastric cancer-derived cells through suppressing HIF2α expression. Cell Oncol (Dordr) 2015; 38:479-84. [PMID: 26526811 DOI: 10.1007/s13402-015-0253-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2015] [Indexed: 01/29/2023] Open
Abstract
PURPOSE Gastric cancer is the third leading cause of cancer-related death in China. Accumulating evidence indicates that HIF2α may affect the aggressiveness of gastric cancer. It has also been found that HIF2α C-terminal PAS domains can form complexes with inactive benzoxadiazole antagonists. Here, the anti-tumor effect of 4-(N,Ndimethylaminosulphonyl)-7-fluoro-1,2,3-benzoxadiazole (DBD-F) on human gastric cancer cells was examined using both in vitro and in vivo assays. METHODS AND RESULTS We found that DBD-F can induce apoptosis and inhibit the mobility of MKN28 and MKN45 gastric cancer-derived cells in vitro. We also found that DBD-F can suppress tumor growth in established gastric cancer-derived xenograft models in vivo. Finally, we found that DBD-F can inhibit HIF2α expression in gastric cancer-derived cells. CONCLUSIONS From our findings we conclude that DBD-F (i) is cytotoxic to gastric cancer-derived cells and (ii) can induce apoptosis in these cells via the MEK/ERK signaling pathway. In addition, our findings strongly indicate that DBD-F can inhibit HIF2α expression by affecting the phosphorylation status of MEK/ERK in gastric cancer-derived cells.
Collapse
Affiliation(s)
- Guang-Hui Tong
- Department of Laboratory Medicine, ShengJing Affiliated Hospital, China Medical University, Shenyang, 110004, China
| | - Wei-Wei Tong
- Department of Laboratory Medicine, ShengJing Affiliated Hospital, China Medical University, Shenyang, 110004, China
| | - Xiao-Song Qin
- Department of Laboratory Medicine, ShengJing Affiliated Hospital, China Medical University, Shenyang, 110004, China
| | - Li-Ping Lu
- Department of Laboratory Medicine, ShengJing Affiliated Hospital, China Medical University, Shenyang, 110004, China
| | - Yong Liu
- Department of Laboratory Medicine, ShengJing Affiliated Hospital, China Medical University, Shenyang, 110004, China.
| |
Collapse
|
392
|
Giarnieri E, Bellipanni G, Macaluso M, Mancini R, Holstein AC, Milanese C, Giovagnoli MR, Giordano A, Russo G. Review: Cell Dynamics in Malignant Pleural Effusions. J Cell Physiol 2015; 230:272-7. [PMID: 25205557 DOI: 10.1002/jcp.24806] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 09/05/2014] [Indexed: 12/29/2022]
Abstract
Malignant pleural effusions (MPEs) are a common manifestation found in patients with lung cancer. After cytological and histological confirmation of malignancy, talc pleurodesis still remains the treatment of choice in patients with MPEs resistant to chemotherapy. Despite this, primary challenges include reduced quality of life and life expectancy in general. Therefore, a better understanding of the cell biology of MPEs, along with improvements in treatment is greatly needed. It has recently been demonstrated that MPEs may represent an excellent source for identification of molecular mechanisms within the tumor and its environment. The present review summarizes the current understanding of MPEs cells and tumor microenvironment, and particularly focuses on dissecting the cross-talk between MPEs and epithelial to mesenchymal transition (EMT), inflammation and cancer stem cells.
Collapse
Affiliation(s)
- Enrico Giarnieri
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, University La Sapienza, Rome, Italy
| | - Gianfranco Bellipanni
- Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, BioLife Science Bldg., Philadelphia, Pennsylvania
| | - Marcella Macaluso
- Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, BioLife Science Bldg., Philadelphia, Pennsylvania
| | - Rita Mancini
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, University La Sapienza, Rome, Italy
| | - Adam Carl Holstein
- Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, BioLife Science Bldg., Philadelphia, Pennsylvania
| | - Carla Milanese
- Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, BioLife Science Bldg., Philadelphia, Pennsylvania
| | - Maria Rosaria Giovagnoli
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, University La Sapienza, Rome, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, BioLife Science Bldg., Philadelphia, Pennsylvania.,INT-CROM, "Pascale Foundation" National Cancer Institute-Cancer Research Center, Mercogliano (AV), Italy.,Department of Medical and Surgical Sciences and Neurosciences, University of Siena, Siena, Italy
| | - Giuseppe Russo
- Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, BioLife Science Bldg., Philadelphia, Pennsylvania
| |
Collapse
|
393
|
Nguyen AH, Wang Y, White DE, Platt MO, McDevitt TC. MMP-mediated mesenchymal morphogenesis of pluripotent stem cell aggregates stimulated by gelatin methacrylate microparticle incorporation. Biomaterials 2015; 76:66-75. [PMID: 26519649 DOI: 10.1016/j.biomaterials.2015.10.043] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 10/15/2015] [Accepted: 10/18/2015] [Indexed: 01/03/2023]
Abstract
Matrix metalloproteinases (MMPs) remodel the extracellular matrix (ECM) to facilitate epithelial-to-mesenchymal transitions (EMTs) and promote cell specification during embryonic development. In this study, we hypothesized that introducing degradable ECM-based biomaterials to pluripotent stem cell (PSC) aggregates would modulate endogenous proteolytic activity and consequently enhance the differentiation and morphogenesis within 3D PSC aggregates. Gelatin methacrylate (GMA) microparticles (MPs) of low (∼20%) or high (∼90%) cross-linking densities were incorporated into mouse embryonic stem cell (ESC) aggregates, and the effects on MMP activity and cell differentiation were examined with or without MMP inhibition. ESC aggregates containing GMA MPs expressed significantly higher levels of total MMP and MMP-2 than aggregates without MPs. GMA MP incorporation increased expression of EMT markers and enhanced mesenchymal morphogenesis of PSC aggregates. MMP inhibition completely abrogated these effects, and GMA MP-induced MMP activation within ESC aggregates was partially reduced by pSMAD 1/5/8 inhibition. These results suggest that GMA particles activate MMPs by protease-substrate interactions to promote EMT and mesenchymal morphogenesis of ESC aggregates in an MMP-dependent manner. We speculate that controlling protease activity via the introduction of ECM-based materials may offer a novel route to engineer the ECM microenvironment to modulate stem cell differentiation.
Collapse
Affiliation(s)
- Anh H Nguyen
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Yun Wang
- The Gladstone Institute for Cardiovascular Disease, San Francisco, CA, USA
| | - Douglas E White
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Manu O Platt
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA; The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Todd C McDevitt
- The Gladstone Institute for Cardiovascular Disease, San Francisco, CA, USA; Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
394
|
Shen J, Lu J, Sui L, Wang D, Yin M, Hoffmann I, Legler A, Pflugfelder GO. The orthologous Tbx transcription factors Omb and TBX2 induce epithelial cell migration and extrusion in vivo without involvement of matrix metalloproteinases. Oncotarget 2015; 5:11998-2015. [PMID: 25344916 PMCID: PMC4322970 DOI: 10.18632/oncotarget.2426] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 09/02/2014] [Indexed: 01/06/2023] Open
Abstract
The transcription factors TBX2 and TBX3 are overexpressed in various human cancers. Here, we investigated the effect of overexpressing the orthologous Tbx genes Drosophila optomotor-blind (omb) and human TBX2 in the epithelium of the Drosophila wing imaginal disc and observed two types of cell motility. Omb/TBX2 overexpressing cells could move within the plane of the epithelium. Invasive cells migrated long-distance as single cells retaining or regaining normal cell shape and apico-basal polarity in spite of attenuated apical DE-cadherin concentration. Inappropriate levels of DE-cadherin were sufficient to drive cell migration in the wing disc epithelium. Omb/TBX2 overexpression and reduced DE-cadherin-dependent adhesion caused the formation of actin-rich lateral cell protrusions. Omb/TBX2 overexpressing cells could also delaminate basally, penetratingthe basal lamina, however, without degradation of extracellular matrix. Expression of Timp, an inhibitor of matrix metalloproteases, blocked neither intraepithelial motility nor basal extrusion. Our results reveal an MMP-independent mechanism of cell invasion and suggest a conserved role of Tbx2-related proteins in cell invasion and metastasis-related processes.
Collapse
Affiliation(s)
- Jie Shen
- Department of Entomology, China Agricultural University, Beijing, China
| | - Juan Lu
- Department of Entomology, China Agricultural University, Beijing, China
| | - Liyuan Sui
- Department of Entomology, China Agricultural University, Beijing, China
| | - Dan Wang
- Department of Entomology, China Agricultural University, Beijing, China
| | - Meizhen Yin
- Key Laboratory of Carbon Fiber and Functional Polymers, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, China
| | - Inka Hoffmann
- Institute of Genetics, Johannes Gutenberg-University, Mainz, Germany
| | - Anne Legler
- Institute of Genetics, Johannes Gutenberg-University, Mainz, Germany
| | | |
Collapse
|
395
|
N-cadherin functions as a growth suppressor in a model of K-ras-induced PanIN. Oncogene 2015; 35:3335-41. [PMID: 26477318 PMCID: PMC4837100 DOI: 10.1038/onc.2015.382] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Revised: 08/31/2015] [Accepted: 09/04/2015] [Indexed: 12/21/2022]
Abstract
Cadherin subtype switching from E-cadherin to N-cadherin is associated with the epithelial-to-mesenchymal transition (EMT), a process required for invasion and dissemination of carcinoma cells. We found N-cadherin is expressed in human and mouse pancreatic intraepithelial neoplasia (PanIN), suggesting that N-cadherin may also play a role in early stage pancreatic cancer. To investigate the role of N-cadherin in mouse PanIN (mPanIN), we simultaneously activated oncogenic K-rasG12D and deleted the N-cadherin (Cdh2) gene in the murine pancreas. Genetic ablation of N-cadherin (N-cad KO) caused hyperproliferation, accelerated mPanIN progression, and early tumor development in K-rasG12D mice. Decreased E-cadherin and redistribution of β-catenin accompanied the loss of N-cadherin in pancreatic ductal epithelial cells (PDEC). Nuclear accumulation of β-catenin and its transcription co-activator Tcf4 led to activation of Wnt/β-catenin target genes. Unexpectedly, loss of N-cadherin in the K-rasG12D model resulted in increased mPanIN progression and tumor incidence. These in vivo results demonstrate for the first time that N-cadherin functions as a growth suppressor in the context of oncogenic K-ras.
Collapse
|
396
|
Takehara T, Teramura T, Onodera Y, Frampton J, Fukuda K. Cdh2 stabilizes FGFR1 and contributes to primed-state pluripotency in mouse epiblast stem cells. Sci Rep 2015; 5:14722. [PMID: 26420260 PMCID: PMC4588589 DOI: 10.1038/srep14722] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 09/03/2015] [Indexed: 12/13/2022] Open
Abstract
The cell adhesion molecule Cadherin 2 (Cdh2) plays important roles in somatic cell adhesion, proliferation and migration. Cdh2 is also highly expressed in mouse epiblast stem cells (mEpiSCs), but its function in these cells is unknown. To understand the function of Cdh2 in mEpiSCs, we compared the expression of pluripotency-related genes in mEpiSCs and mouse embryonic stem cells (mESCs) after either Cdh2 knockdown or Cdh2 over-expression. Introduction of specific siRNA against Cdh2 led to attenuation of pluripotency-related genes. Pluripotent gene expression was not recovered by over-expression of Cdh1 following Cdh2 knockdown. Western blot analysis and co-immunoprecipitation assays revealed that Cdh2 stabilizes FGFR1 in mEpiSCs. Furthermore, stable transfection of mESCs with Cdh2 cDNA followed by FGF2 supplementation accelerated cell differentiation. Thus, Cdh2 contributes to the establishment and maintenance of FGF signaling-dependent self-renewal in mEpiSCs through stabilization of FGFR1.
Collapse
Affiliation(s)
- Toshiyuki Takehara
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine, 377-2 Ohnohigashi, Osaka-sayama, Osaka, Japan 5898511
| | - Takeshi Teramura
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine, 377-2 Ohnohigashi, Osaka-sayama, Osaka, Japan 5898511
| | - Yuta Onodera
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine, 377-2 Ohnohigashi, Osaka-sayama, Osaka, Japan 5898511
| | - John Frampton
- School of Biomedical Engineering, Dalhousie University, Halifax, Nova Scotia, Canada B3H 4R2 1-902-494-4175
| | - Kanji Fukuda
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine, 377-2 Ohnohigashi, Osaka-sayama, Osaka, Japan 5898511
| |
Collapse
|
397
|
Garg S, Fischer SC, Schuman EM, Stelzer EHK. Lateral assembly of N-cadherin drives tissue integrity by stabilizing adherens junctions. J R Soc Interface 2015; 12:20141055. [PMID: 25589573 DOI: 10.1098/rsif.2014.1055] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cadherin interactions ensure the correct registry and anchorage of cells during tissue formation. Along the plasma membrane, cadherins form inter-junctional lattices via cis- and trans-dimerization. While structural studies have provided models for cadherin interactions, the molecular nature of cadherin binding in vivo remains unexplored. We undertook a multi-disciplinary approach combining live cell imaging of three-dimensional cell assemblies (spheroids) with a computational model to study the dynamics of N-cadherin interactions. Using a loss-of-function strategy, we demonstrate that each N-cadherin interface plays a distinct role in spheroid formation. We found that cis-dimerization is not a prerequisite for trans-interactions, but rather modulates trans-interfaces to ensure tissue stability. Using a model of N-cadherin junction dynamics, we show that the absence of cis-interactions results in low junction stability and loss of tissue integrity. By quantifying the binding and unbinding dynamics of the N-cadherin binding interfaces, we determined that mutating either interface results in a 10-fold increase in the dissociation constant. These findings provide new quantitative information on the steps driving cadherin intercellular adhesion and demonstrate the role of cis-interactions in junction stability.
Collapse
Affiliation(s)
- S Garg
- Department of Synaptic Plasticity, Max Planck Institute for Brain Research, 60438 Frankfurt am Main, Germany
| | - S C Fischer
- Department of Physical Biology (IZN, FB 15), Buchmann Institute for Molecular Life Sciences (BMLS), Cluster of Excellence Frankfurt Macromolecular Complexes (CEF MC), Goethe Universität Frankfurt am Main, 60438 Frankfurt am Main, Germany
| | - E M Schuman
- Department of Synaptic Plasticity, Max Planck Institute for Brain Research, 60438 Frankfurt am Main, Germany
| | - E H K Stelzer
- Department of Physical Biology (IZN, FB 15), Buchmann Institute for Molecular Life Sciences (BMLS), Cluster of Excellence Frankfurt Macromolecular Complexes (CEF MC), Goethe Universität Frankfurt am Main, 60438 Frankfurt am Main, Germany
| |
Collapse
|
398
|
Wang P, Yang AT, Cong M, Liu TH, Zhang D, Huang J, Tong XF, Zhu ST, Xu Y, Tang SZ, Wang BE, Ma H, Jia JD, You H. EGF Suppresses the Initiation and Drives the Reversion of TGF-β1-induced Transition in Hepatic Oval Cells Showing the Plasticity of Progenitor Cells. J Cell Physiol 2015; 230:2362-70. [PMID: 25739869 DOI: 10.1002/jcp.24962] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Accepted: 02/17/2015] [Indexed: 12/15/2022]
Abstract
Transforming growth factor-β1 (TGF-β1) induces hepatic progenitors to tumor initiating cells through epithelial-mesenchymal transition (EMT), thus raising an important drawback for stem cell-based therapy. How to block and reverse TGF-β1-induced transition is crucial for progenitors' clinical application and carcinogenic prevention. Rat adult hepatic progenitors, hepatic oval cells, experienced E-cadherin to N-cadherin switch and changed to α-smooth muscle actin (α-SMA) positive cells after TGF-β1 incubation, indicating EMT. When TGF-β1 plus EGF were co-administrated to these cells, EGF dose-dependently suppressed the cadherin switch and α-SMA expression. Interestingly, if EGF was applied to TGF-β1-pretreated cells, the cells that have experienced EMT could return to their epithelial phenotype. Abruption of EGF receptor revealed that EGF exerted its blockage and reversal effects through phosphorylation of ERK1/2 and Akt. These findings suggest an important attribute of EGF on opposing and reversing TGF-β1 effects, indicating the plasticity of hepatic progenitors.
Collapse
Affiliation(s)
- Ping Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, China.,Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | - Ai-Ting Yang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Min Cong
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Tian-Hui Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Dong Zhang
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Jian Huang
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Xiao-Fei Tong
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Sheng-Tao Zhu
- Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Yong Xu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Shu-Zhen Tang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Bao-En Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Hong Ma
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Ji-Dong Jia
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Hong You
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis & National Clinical Research Center of Digestive Diseases, Beijing, China
| |
Collapse
|
399
|
Busaranon K, Plaimee P, Sritularak B, Chanvorachote P. Moscatilin inhibits epithelial-to-mesenchymal transition and sensitizes anoikis in human lung cancer H460 cells. J Nat Med 2015; 70:18-27. [PMID: 26384689 DOI: 10.1007/s11418-015-0931-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 07/21/2015] [Indexed: 12/18/2022]
Abstract
Metastasis in lung cancer has been recognized as an important cause of high mortality. Resistance to anoikis and the epithelial-to-mesenchymal transition (EMT) are critical factors for the successful spread of cancer cells. Compounds that suppress these features of cancer cells should be potentially active for anti-metastasis approaches. We have demonstrated for the first time that moscatilin, at its non-toxic concentrations to lung cancer cells and human normal keratinocytes, significantly decreases lung cancer cell survival in the detached condition, and suppresses the formation of tumors in an anchorage-independent growth assay. Furthermore, we found that moscatilin significantly decreased the activated level of survival proteins, namely ERK and Akt. In addition, moscatilin down-regulated cavelolin-1 (Cav-1), leading to a reduction in anti-apoptotic Mcl-1 protein. In terms of EMT, treatment of the cells with moscatilin significantly suppressed mesenchymal cell markers, namely vimentin, Slug, and Snail. These results indicate that moscatilin inhibited anoikis resistance in lung cancer cells via survival suppression, Cav-1 down-regulation, and inhibition of EMT. The compound could therefore be beneficial for the treatment and prevention of lung cancer metastasis.
Collapse
Affiliation(s)
- Kesarin Busaranon
- Faculty of Pharmacy, Rangsit University, Pathum Thani, 12000, Thailand
| | - Preeyaporn Plaimee
- Cell-Based Drug and Health Product Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Boonchoo Sritularak
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Pithi Chanvorachote
- Cell-Based Drug and Health Product Development Research Unit, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand. .,Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
400
|
Yang Y, Park SY, Nguyen TT, Yu YH, Nguyen TV, Sun EG, Udeni J, Jeong MH, Pereira I, Moon C, Ha HH, Kim KK, Hur JS, Kim H. Lichen Secondary Metabolite, Physciosporin, Inhibits Lung Cancer Cell Motility. PLoS One 2015; 10:e0137889. [PMID: 26371759 PMCID: PMC4570789 DOI: 10.1371/journal.pone.0137889] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 08/24/2015] [Indexed: 01/28/2023] Open
Abstract
Lichens produce various unique chemicals that can be used for pharmaceutical purposes. To screen for novel lichen secondary metabolites showing inhibitory activity against lung cancer cell motility, we tested acetone extracts of 13 lichen samples collected in Chile. Physciosporin, isolated from Pseudocyphellaria coriacea (Hook f. & Taylor) D.J. Galloway & P. James, was identified as an effective compound and showed significant inhibitory activity in migration and invasion assays against human lung cancer cells. Physciosporin treatment reduced both protein and mRNA levels of N-cadherin with concomitant decreases in the levels of epithelial-mesenchymal transition markers such as snail and twist. Physciosporin also suppressed KITENIN (KAI1 C-terminal interacting tetraspanin)-mediated AP-1 activity in both the absence and presence of epidermal growth factor stimulation. Quantitative real-time PCR analysis showed that the expression of the metastasis suppressor gene, KAI1, was increased while that of the metastasis enhancer gene, KITENIN, was dramatically decreased by physciosporin. Particularly, the activity of 3’-untranslated region of KITENIN was decreased by physciosporin. Moreover, Cdc42 and Rac1 activities were decreased by physciosporin. These results demonstrated that the lichen secondary metabolite, physciosporin, inhibits lung cancer cell motility through novel mechanisms of action.
Collapse
Affiliation(s)
- Yi Yang
- Korean Lichen Research Institute, Sunchon National University, Sunchon 540–950, Republic of Korea
| | - So-Yeon Park
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Sunchon 540–950, Republic of Korea
| | - Thanh Thi Nguyen
- Korean Lichen Research Institute, Sunchon National University, Sunchon 540–950, Republic of Korea
- Faculty of Natural Science and Technology, Tay Nguyen University, Buon Ma Thuot, Vietnam
| | - Young Hyun Yu
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Sunchon 540–950, Republic of Korea
| | - Tru Van Nguyen
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Sunchon 540–950, Republic of Korea
| | - Eun Gene Sun
- Medical Research Center for Gene Regulation, Chonnam National University Medical School, Gwangju 500–872, Republic of Korea
| | - Jayalal Udeni
- Korean Lichen Research Institute, Sunchon National University, Sunchon 540–950, Republic of Korea
| | - Min-Hye Jeong
- Korean Lichen Research Institute, Sunchon National University, Sunchon 540–950, Republic of Korea
| | - Iris Pereira
- Institute of Biological Sciences, Universidad de Talca, Talca 747–721, Chile
| | - Cheol Moon
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Sunchon 540–950, Republic of Korea
| | - Hyung-Ho Ha
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Sunchon 540–950, Republic of Korea
| | - Kyung Keun Kim
- Medical Research Center for Gene Regulation, Chonnam National University Medical School, Gwangju 500–872, Republic of Korea
| | - Jae-Seoun Hur
- Korean Lichen Research Institute, Sunchon National University, Sunchon 540–950, Republic of Korea
- * E-mail: (HK); (J-SH)
| | - Hangun Kim
- College of Pharmacy and Research Institute of Life and Pharmaceutical Sciences, Sunchon National University, Sunchon 540–950, Republic of Korea
- * E-mail: (HK); (J-SH)
| |
Collapse
|