201
|
Goenezen S, Rennie MY, Rugonyi S. Biomechanics of early cardiac development. Biomech Model Mechanobiol 2012; 11:1187-204. [PMID: 22760547 DOI: 10.1007/s10237-012-0414-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 06/20/2012] [Indexed: 12/31/2022]
Abstract
Biomechanics affect early cardiac development, from looping to the development of chambers and valves. Hemodynamic forces are essential for proper cardiac development, and their disruption leads to congenital heart defects. A wealth of information already exists on early cardiac adaptations to hemodynamic loading, and new technologies, including high-resolution imaging modalities and computational modeling, are enabling a more thorough understanding of relationships between hemodynamics and cardiac development. Imaging and modeling approaches, used in combination with biological data on cell behavior and adaptation, are paving the road for new discoveries on links between biomechanics and biology and their effect on cardiac development and fetal programming.
Collapse
Affiliation(s)
- Sevan Goenezen
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA.
| | | | | |
Collapse
|
202
|
Qi S, Song Y, Peng Y, Wang H, Long H, Yu X, Li Z, Fang L, Wu A, Luo W, Zhen Y, Zhou Y, Chen Y, Mai C, Liu Z, Fang W. ZEB2 mediates multiple pathways regulating cell proliferation, migration, invasion, and apoptosis in glioma. PLoS One 2012; 7:e38842. [PMID: 22761708 PMCID: PMC3383704 DOI: 10.1371/journal.pone.0038842] [Citation(s) in RCA: 146] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 05/11/2012] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The aim of the present study was to analyze the expression of Zinc finger E-box Binding homeobox 2 (ZEB2) in glioma and to explore the molecular mechanisms of ZEB2 that regulate cell proliferation, migration, invasion, and apoptosis. METHODOLOGY/PRINCIPAL FINDINGS Expression of ZEB2 in 90 clinicopathologically characterized glioma patients was analyzed by immunohistochemistry. Furthermore, siRNA targeting ZEB2 was transfected into U251 and U87 glioma cell lines in vitro and proliferation, migration, invasion, and apoptosis were examined separately by MTT assay, Transwell chamber assay, flow cytometry, and western blot. RESULTS The expression level of ZEB2 protein was significantly increased in glioma tissues compared to normal brain tissues (P<0.001). In addition, high levels of ZEB2 protein were positively correlated with pathology grade classification (P = 0.024) of glioma patients. Knockdown of ZEB2 by siRNA suppressed cell proliferation, migration and invasion, as well as induced cell apoptosis in glioma cells. Furthermore, ZEB2 downregulation was accompanied by decreased expression of CDK4/6, Cyclin D1, Cyclin E, E2F1, and c-myc, while p15 and p21 were upregulated. Lowered expression of ZEB2 enhanced E-cadherin levels but also inhibited β-Catenin, Vimentin, N-cadherin, and Snail expression. Several apoptosis-related regulators such as Caspase-3, Caspase-6, Caspase-9, and Cleaved-PARP were activated while PARP was inhibited after ZEB2 siRNA treatment. CONCLUSION Overexpression of ZEB2 is an unfavorable factor that may facilitate glioma progression. Knockdown ZEB2 expression by siRNA suppressed cell proliferation, migration, invasion and promoted cell apoptosis in glioma cells.
Collapse
Affiliation(s)
- Songtao Qi
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Ye Song
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
- Cancer Research Institute of Southern Medical University, Guangzhou, People’s Republic of China
| | - Yuping Peng
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Hao Wang
- Cancer Research Institute of Southern Medical University, Guangzhou, People’s Republic of China
| | - Hao Long
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Xiaoli Yu
- Cancer Research Institute of Southern Medical University, Guangzhou, People’s Republic of China
| | - Zhiyong Li
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Luxiong Fang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Aibing Wu
- Cancer Research Institute of Southern Medical University, Guangzhou, People’s Republic of China
| | - Weiren Luo
- Cancer Research Institute of Southern Medical University, Guangzhou, People’s Republic of China
| | - Yan Zhen
- Cancer Research Institute of Southern Medical University, Guangzhou, People’s Republic of China
| | - Ying Zhou
- Cancer Research Institute of Southern Medical University, Guangzhou, People’s Republic of China
| | - Yan Chen
- Cancer Research Institute of Southern Medical University, Guangzhou, People’s Republic of China
| | - Chunping Mai
- Cancer Research Institute of Southern Medical University, Guangzhou, People’s Republic of China
| | - Zhen Liu
- Cancer Research Institute of Southern Medical University, Guangzhou, People’s Republic of China
- Department of Pathology, Basic School of Guangzhou Medical College, Guangzhou, People’s Republic of China
| | - Weiyi Fang
- Cancer Research Institute of Southern Medical University, Guangzhou, People’s Republic of China
| |
Collapse
|
203
|
Hagio M, Matsuda Y, Suzuki T, Ishiwata T. Nestin regulates epithelial-mesenchymal transition marker expression in pancreatic ductal adenocarcinoma cell lines. Mol Clin Oncol 2012; 1:83-87. [PMID: 24649127 DOI: 10.3892/mco.2012.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 06/04/2012] [Indexed: 01/15/2023] Open
Abstract
Nestin, a class VI intermediate filament, is a neuronal stem/progenitor cell marker that is also expressed by various types of cancer, including pancreatic ductal adenocarcinoma (PDAC). We previously detected nestin expression in approximately 30% of PDAC cases, and found that nestin promotes the migration, invasion and metastasis of cells. Findings of recent studies have shown that epithelial mesenchymal transition (EMT) is important in the invasion and migration of cancer. In the present study, we investigated whether an altered nestin expression affected the expression levels of EMT markers in PDAC cells. Two human PDAC cell lines, PK-45H and KLM-1, in which nestin was suppressed and overexpressed, respectively, were used. The expression levels of the EMT-related molecules E-cadherin, Snail, Slug and Twist were analyzed using quantitative RT-PCR. Results showed that E-cadherin expression was decreased in nestin-overexpressed KLM-1 cells, and increased in nestin-suppressed PK-45H cells. Snail gene expression in the PDAC cells was altered concomitantly with the changes in nestin expression, while the Slug gene expression was significantly decreased in nestin-overexpressed KLM-1 cells. The Twist gene expression was below the detection limit in the two PDAC cell lines. The present findings indicated that nestin may be involved in the control of cancer behaviors in PDAC via the modulation of EMT-related molecules.
Collapse
Affiliation(s)
- Masahito Hagio
- Departments of Pathology and Integrative Oncological Pathology, Nippon Medical School, Tokyo 113-8602, Japan
| | - Yoko Matsuda
- Departments of Pathology and Integrative Oncological Pathology, Nippon Medical School, Tokyo 113-8602, Japan
| | - Taeko Suzuki
- Departments of Pathology and Integrative Oncological Pathology, Nippon Medical School, Tokyo 113-8602, Japan
| | - Toshiyuki Ishiwata
- Departments of Pathology and Integrative Oncological Pathology, Nippon Medical School, Tokyo 113-8602, Japan
| |
Collapse
|
204
|
Das S, Becker BN, Hoffmann FM, Mertz JE. Reversal of transforming growth factor-β induced epithelial-to-mesenchymal transition and the ZEB proteins. FIBROGENESIS & TISSUE REPAIR 2012; 5:S28. [PMID: 23259633 PMCID: PMC3368790 DOI: 10.1186/1755-1536-5-s1-s28] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background The dynamic process of epithelial-to-mesenchymal transition (EMT) is a causal event in kidney fibrosis. This cellular phenotypic transition involves activation of transcriptional responses and remodeling of cellular structures to change cellular function. The molecular mechanisms that directly contribute to the re-establishment of the epithelial phenotype are poorly understood. Results Here, we discuss recent studies from our group and other laboratories identifying signaling pathways leading to the reversal of EMT in fibrotic models. We also present evidence that transcriptional factors such as the ZEB proteins are important regulators for reversal of EMT. Conclusion These studies provide insights into cellular plasticity and possible targets for therapeutic intervention.
Collapse
Affiliation(s)
- Shreyasi Das
- Sanford-Burnham Medical Research Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Bryan N Becker
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Madison, Wisconsin 53792, USA
| | - F Michael Hoffmann
- Laboratory of Genetics, University of Wisconsin School of Medicine and Public Health, 425-G Henry Mall, Madison, Wisconsin 53706, USA ; McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, 1400 University Ave, Madison, Wisconsin 53706, USA
| | - Janet E Mertz
- McArdle Laboratory for Cancer Research, University of Wisconsin School of Medicine and Public Health, 1400 University Ave, Madison, Wisconsin 53706, USA
| |
Collapse
|
205
|
Shoval I, Kalcheim C. Antagonistic activities of Rho and Rac GTPases underlie the transition from neural crest delamination to migration. Dev Dyn 2012; 241:1155-68. [PMID: 22553120 DOI: 10.1002/dvdy.23799] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2012] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Neural crest progenitors arise as epithelial cells and then undergo a transition into mesenchyme that generates motility. Previously, we showed that active Rho maintains crest cells in the epithelial conformation by keeping stress fibers and membrane-bound N-cadherin. RESULTS While Rho disappears from cell membranes upon delamination, active Rac1 becomes apparent in lamellipodia of mesenchymal cells. Loss of Rac1 function at trunk levels inhibited NC migration but did not prevent cell emigration that is associated with N-cadherin downregulation and G1/S transition. Furthermore, inhibition of Rho stimulated premature Rac1 activity and consequent formation of lamellipodia, leading to NC migration. To examine whether timely migration influences cell fate, Rac1 activity was transiently inhibited to delay dispersion of early NC cells that generate neural derivatives, and its activity was restored by the time of melanoblast migration. Even if confronted with a melanocytic environment, late-dispersing progenitors colonized sensory ganglia where they generated neurons and glia. CONCLUSIONS In the context of crest delamination and migration, activities of Rho and Rac are differential, sequential, and antagonistic. Furthermore, transient inhibition of Rac1 that delays the onset of crest dispersion raises the possibility that the fate of trunk neural progenitors might be restricted prior to migration.
Collapse
Affiliation(s)
- Irit Shoval
- Department of Medical Neurobiology, Hebrew University-Hadassah Medical School, IMRIC and ELSC, Jerusalem, Israel
| | | |
Collapse
|
206
|
Unbalanced expression of CK2 kinase subunits is sufficient to drive epithelial-to-mesenchymal transition by Snail1 induction. Oncogene 2012; 32:1373-83. [PMID: 22562247 DOI: 10.1038/onc.2012.165] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Epithelial-to-mesenchymal transition (EMT) is closely linked to conversion of early-stage tumours into invasive malignancies. Many signalling pathways are involved in EMT, but the key regulatory kinases in this important process have not been clearly identified. Protein kinase CK2 is a multi-subunit protein kinase, which, when overexpressed, has been linked to disease progression and poor prognosis in various cancers. Specifically, overexpression of CK2α in human breast cancers is correlated with metastatic risk. In this article, we show that an imbalance of CK2 subunits reflected by a decrease in the CK2β regulatory subunit in a subset of breast tumour samples is correlated with induction of EMT-related markers. CK2β-depleted epithelial cells displayed EMT-like morphological changes, enhanced migration, and anchorage-independent growth, all of which require Snail1 induction. In epithelial cells, Snail1 stability is negatively regulated by CK2 and GSK3β through synergistic hierarchal phosphorylation. This process depends strongly on CK2β, thus confirming that CK2 functions upstream of Snail1. In primary breast tumours, CK2β underexpression also correlates strongly with expression of EMT markers, emphasizing the link between asymmetric expression of CK2 subunits and EMT in vivo. Our results therefore highlight the importance of CK2β in controlling epithelial cell plasticity. They show that CK2 holoenzyme activity is essential to suppress EMT, and that it contributes to maintaining a normal epithelial morphology. This study also suggests that unbalanced expression of CK2 subunits may drive EMT, thereby contributing to tumour progression.
Collapse
|
207
|
Strobl-Mazzulla PH, Bronner ME. Epithelial to mesenchymal transition: new and old insights from the classical neural crest model. Semin Cancer Biol 2012; 22:411-6. [PMID: 22575214 DOI: 10.1016/j.semcancer.2012.04.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 04/17/2012] [Indexed: 01/11/2023]
Abstract
The epithelial-to-mesenchymal transition (EMT) is an important event converting compact and ordered epithelial cells into migratory mesenchymal cells. Given the molecular and cellular similarities between pathological and developmental EMTs, studying this event during neural crest development offers and excellent in vivo model for understanding the mechanisms underlying this process. Here, we review new and old insight into neural crest EMT in search of commonalities with cancer progression that might aid in the design of specific therapeutic prevention.
Collapse
Affiliation(s)
- Pablo H Strobl-Mazzulla
- Laboratory of Developmental Biology, Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús (CONICET-UNSAM), Chascomús, Argentina.
| | | |
Collapse
|
208
|
Xue HT, Wang HG, Huang XD, Shen P, Ji GZ. Loss of Smad4 expression inhibits epithelial-mesenchymal transition in SMMC-7721 cells. Shijie Huaren Xiaohua Zazhi 2012; 20:923-929. [DOI: 10.11569/wcjd.v20.i11.923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the influence of loss of Smad4 expression on TGF-β1-induced epithelial-mesenchymal transition in the human hepatocellular carcinoma cell line SMMC-7721.
METHODS: The influence of loss of Smad4 expression on the expression of β-catenin and Vimentin mRNAs and proteins was evaluated by RT-PCR and Western blot. Immunofluorescence was used to analyze the location and fluorescence intensity of Smad4, β-catenin, Vimentin in non-transfected SMMC-7721 cells and those transfected with Smad-specific siRNAs (RNAi-Smad4-2 and RNAi-Smad4-12) or unspecific siRNA (RNAi-NC).
RESULTS: Compared to non-transfected SMMC-7721 cells and those tranfected with RNAi-NC, the expression of β-catenin mRNA and protein remarkably increased in SMMC-7721 cells transfected with RNAi-Smad4-2 or RNAi-Smad4-12 (all P < 0.05). Loss of Smad4 expression promoted β-catenin nuclear translocation. Immunofluorescence assay revealed that β-catenin fluorescence was located in the nuclei of non-transfected SMMC-7721 cells and those tranfected with RNAi-NC, but in the cytoplasm of SMMC-7721 cells transfected with RNAi-Smad4-2 or RNAi-Smad4-12. On the other hand, loss of Smad4 expression down-regulated Vimentin protein expression (P < 0.05) and cytoplasmic fluorescence intensity, but had no significant impact on Vimentin mRNA expression in SMMC-7721 cells and those transfected with different siRNAs.
CONCLUSION: Loss of Smad4 expression regulates β-catenin and Vimentin and therefore plays an important role in inhibiting epithelial-mesenchymal transition in SMMC-7721 cells.
Collapse
|
209
|
Shirley SH, Greene VR, Duncan LM, Torres Cabala CA, Grimm EA, Kusewitt DF. Slug expression during melanoma progression. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:2479-89. [PMID: 22503751 DOI: 10.1016/j.ajpath.2012.02.014] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Revised: 02/08/2012] [Accepted: 02/13/2012] [Indexed: 01/05/2023]
Abstract
Slug (Snai2), a member of the Snail family of zinc finger transcription factors, plays a role in the epithelial-to-mesenchymal transformation (EMT) that occurs during melanocyte emigration from the neural crest. A role for Slug in the EMT-like loss of cell adhesion and increased cell motility exhibited during melanoma progression has also been proposed. Our immunohistochemical studies of melanoma arrays, however, revealed that Slug expression was actually higher in nevi than in primary or metastatic melanomas. Moreover, Slug expression in melanomas was not associated with decreased expression of E-cadherin, the canonical Slug target in EMT. Comparisons of endogenous Slug and E-cadherin expression in cultured normal human melanocytes and melanoma cell lines supported our immunohistochemical findings. Expression of exogenous Slug in melanocytes and melanoma cells in vitro, however, suppressed E-cadherin expression, enhanced N-cadherin expression, and stimulated cell migration and invasion. Interestingly, both in tumors and cultured cell lines, there was a clear relationship between expression of Slug and MITF, a transcription factor known to regulate Slug expression during development. Taken together, our findings suggest that Slug expression during melanomagenesis is highest early in the process and that persistent Slug expression is not required for melanoma progression. The precise role of Slug in melanomagenesis remains to be elucidated and may be related to its interactions with other drivers of EMT, such as Snail.
Collapse
Affiliation(s)
- Stephanie H Shirley
- Department of Molecular Carcinogenesis, Science Park, University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA
| | | | | | | | | | | |
Collapse
|
210
|
Zhang K, Zhang M, Zhao H, Yan B, Zhang D, Liang J. S100A4 regulates motility and invasiveness of human esophageal squamous cell carcinoma through modulating the AKT/Slug signal pathway. Dis Esophagus 2012; 25:731-9. [PMID: 22458600 DOI: 10.1111/j.1442-2050.2012.01323.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The involvement of S100A4 in modulating invasiveness of esophageal squamous cell carcinoma (ESCC) cell lines was explored. It was shown that S100A4 expression is positively correlated with the degree of invasiveness in human ESCC cells. The S100A4-rich EC-1 cells displayed higher migratory and invasive cell behavior while ET-1 cells with low S100A4 expression levels displayed lower migratory and invasive cell behavior. S100A4 silencing by small interfering (siRNA) in EC-1 cells induced E-cadherin expression, and overexpression of S100A4 in a lowly invasive TE-1 cells suppressed E-cadherin expression. It is suggested that S100A4 silencing inhibit invasion via E-cadherin upregulation, and overexpression of S100A4 promote invasion via E-cadherin downregulation in ESCC cells. Compared with the vector-transfected cells, S100A4 silencing in EC-1 cells showed reduced ability of migration and invasiveness, and overexpression of S100A4 in TE-1 cells showed increased ability of migration and invasiveness via wound-healing and Transwell assay, and pseudometastatic model assay. Furthermore, re-expression of S100A4 could increase the invasive phenotypes in S100A4 siRNA transfected EC-1 cells, and S100A4 silencing could decrease the invasive phenotypes in S100A4 circular DNA (cDNA) transfected TE-1 cells. It was found that Slug is downregulated in S100A4 siRNA transfected EC-1 cells, and Slug is upregulated in S100A4 cDNA transfected TE-1 cells. It was also discovered S100A4 cDNA induced protein kinase B (AKT) phosphorylation at Serine-473(phospho-AKT [p-AKT]) levels, followed by the Slug upregulation, and S100A4 siRNA decreases the phospho-AKT levels, followed by the Slug downregulation. The data suggested that S100A4 could regulate migratory and invasive behavior of human ESCC cells through modulating AKT/Slug pathway.
Collapse
Affiliation(s)
- K Zhang
- General Surgery, Affiliated Hospital of Medical College, QingDao University, QingDao, Shan Dong Province, China
| | | | | | | | | | | |
Collapse
|
211
|
Myrna KE, Mendonsa R, Russell P, Pot SA, Liliensiek SJ, Jester JV, Nealey PF, Brown D, Murphy CJ. Substratum topography modulates corneal fibroblast to myofibroblast transformation. Invest Ophthalmol Vis Sci 2012; 53:811-6. [PMID: 22232431 DOI: 10.1167/iovs.11-7982] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
PURPOSE The transition of corneal fibroblasts to the myofibroblast phenotype is known to be important in wound healing. The purpose of this study was to determine the effect of topographic cues on TGFβ-induced myofibroblast transformation of corneal cells. METHODS Rabbit corneal fibroblasts were cultured on nanopatterned surfaces having topographic features of varying sizes. Cells were cultured in media containing TGFβ at concentrations ranging from 0 to 10 ng/mL. RNA and protein were collected from cells cultured on topographically patterned and planar substrates and analyzed for the myofibroblast marker α-smooth muscle actin (αSMA) and Smad7 expression by quantitative real time PCR. Western blot and immunocytochemistry analysis for αSMA were also performed. RESULTS Cells grown on patterned surfaces demonstrated significantly reduced levels of αSMA (P < 0.002) compared with planar surfaces when exposed to TGFβ; the greatest reduction was seen on the 1400 nm surface. Smad7 mRNA expression was significantly greater on all patterned surfaces exposed to TGFβ (P < 0.002), whereas cells grown on planar surfaces showed equal or reduced levels of Smad7. Western blot analysis and αSMA immunocytochemical staining demonstrated reduced transition to the myofibroblast phenotype on the 1400 nm surface when compared with cells on a planar surface. CONCLUSIONS These data demonstrate that nanoscale topographic features modulate TGFβ-induced myofibroblast differentiation and αSMA expression, possibly through upregulation of Smad7. It is therefore proposed that in the wound environment, native nanotopographic cues assist in stabilizing the keratocyte/fibroblast phenotype while pathologic microenvironmental alterations may be permissive for increased myofibroblast differentiation and the development of fibrosis and corneal haze.
Collapse
Affiliation(s)
- Kathern E Myrna
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
212
|
Ho CM, Chang SF, Hsiao CC, Chien TY, Shih DTB. Isolation and characterization of stromal progenitor cells from ascites of patients with epithelial ovarian adenocarcinoma. J Biomed Sci 2012; 19:23. [PMID: 22330345 PMCID: PMC3305560 DOI: 10.1186/1423-0127-19-23] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 02/14/2012] [Indexed: 01/06/2023] Open
Abstract
Background At least one-third of epithelial ovarian cancers are associated with the development of ascites containing heterogeneous cell populations, including tumor cells, inflammatory cells, and stromal elements. The components of ascites and their effects on the tumor cell microenvironment remain poorly understood. This study aimed to isolate and characterize stromal progenitor cells from the ascites of patients with epithelial ovarian adenocarcinoma (EOA). Methods Seventeen ascitic fluid samples and 7 fresh tissue samples were collected from 16 patients with EOA. The ascites samples were then cultured in vitro in varying conditions. Flow cytometry and immunocytochemistry were used to isolate and characterize 2 cell populations with different morphologies (epithelial type and mesenchymal type) deriving from the ascites samples. The in vitro cell culture model was established using conditional culture medium. Results The doubling times of the epithelial type and mesenchymal type cells were 36 h and 48 h, respectively, indicating faster growth of the epithelial type cells compared to the mesenchymal type cells. Cultured in vitro, these ascitic cells displayed the potential for self-renewal and long-term proliferation, and expressed the typical cancer stem/progenitor cell markers CD44high, CD24low, and AC133+. These cells also demonstrated high BMP-2, BMP4, TGF-β, Rex-1, and AC133 early gene expression, and expressed EGFR, integrin α2β1, CD146, and Flt-4, which are highly associated with tumorigenesis and metastasis. The epithelial type cells demonstrated higher cytokeratin 18 and E-cadherin expression than the mesenchymal type cells. The mesenchymal type cells, in contrast, demonstrated higher AC133, CD73, CD105, CD117, EGFR, integrin α2β1, and CD146 surface marker expression than the epithelial type cells. Conclusion The established culture system provides an in vitro model for the selection of drugs that target cancer-associated stromal progenitor cells, and for the development of ovarian cancer treatments.
Collapse
Affiliation(s)
- Chih-Ming Ho
- Gynecologic Cancer Center, Department of Obstetrics and GynecologyCathay General Hospital, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
213
|
Abstract
The epicardium, the tissue layer covering the cardiac muscle (myocardium), develops from the proepicardium, a mass of coelomic progenitors located at the venous pole of the embryonic heart. Proepicardium cells attach to and spread over the myocardium to form the primitive epicardial epithelium. The epicardium subsequently undergoes an epithelial-to-mesenchymal transition to give rise to a population of epicardium-derived cells, which in turn invade the heart and progressively differentiate into various cell types, including cells of coronary blood vessels and cardiac interstitial cells. Epicardial cells and epicardium-derived cells signal to the adjacent cardiac muscle in a paracrine fashion, promoting its proliferation and expansion. Recently, high expectations have been raised about the epicardium as a candidate source of cells for the repair of the damaged heart. Because of its developmental importance and therapeutic potential, current research on this topic focuses on the complex signals that control epicardial biology. This review describes the signaling pathways involved in the different stages of epicardial development and discusses the potential of epicardial signals as targets for the development of therapies to repair the diseased heart.
Collapse
|
214
|
Serrano I, McDonald PC, Lock FE, Dedhar S. Role of the integrin-linked kinase (ILK)/Rictor complex in TGFβ-1-induced epithelial-mesenchymal transition (EMT). Oncogene 2012; 32:50-60. [PMID: 22310280 DOI: 10.1038/onc.2012.30] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Epithelial-to-mesenchymal transition (EMT) causes fibrosis, cancer progression and metastasis. Integrin-linked kinase (ILK) is a focal adhesion adaptor and a serine/threonine protein kinase that regulates cell proliferation, survival and EMT. Elucidating the molecular mechanisms necessary for development and progression of human malignancies is critical to predict the most appropriate targets for cancer therapy. Here, we used transforming growth factor beta-1 (TGFβ-1) to promote EMT and migration in mammary epithelial cells. We demonstrate a requirement of ILK activity for TGFβ-1-mediated EMT in mammary epithelial cells. In addition to nuclear translocation of Snail and Slug, TGFβ-1 treatment also induced expression of the mammalian target of rapamycin complex 2 component Rictor and its phosphorylation on Thr1135. Interestingly, TGFβ-1 treatment also induced an interaction between ILK and Rictor. All of these TGFβ-1-induced processes were significantly suppressed by inhibiting ILK activity or by disrupting the ILK/Rictor complex using small-interfering RNA-mediated knockdown. Furthermore, we identified ILK/Rictor complex formation in cancer but not in normal cell types, and this was accompanied by ILK-dependent phosphorylation of Rictor on residue Thr1135. Inhibition of ILK partially reversed the basal mesenchymal phenotype of MDA-MB-231 cells and prevented EMT in MCF10A cells after TGFβ-1 treatment. These data demonstrate a requirement for ILK function in TGFβ-1-induced EMT in mammary epithelial cells and identify the ILK/Rictor complex as a potential molecular target for preventing/reversing EMT.
Collapse
Affiliation(s)
- I Serrano
- Department of Integrative Oncology, British Columbia Cancer Research Centre, BC Cancer Agency, Vancouver, British Columbia, Canada
| | | | | | | |
Collapse
|
215
|
Katz S, Balogh P, Nagy N, Kiss AL. Epithelial-To-Mesenchymal Transition Induced by Freund’s Adjuvant Treatment in Rat Mesothelial Cells: A Morphological and Immunocytochemical Study. Pathol Oncol Res 2012; 18:641-9. [DOI: 10.1007/s12253-011-9489-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Accepted: 12/08/2011] [Indexed: 12/27/2022]
|
216
|
Guadamillas MC, Cerezo A, Del Pozo MA. Overcoming anoikis--pathways to anchorage-independent growth in cancer. J Cell Sci 2012; 124:3189-97. [PMID: 21940791 DOI: 10.1242/jcs.072165] [Citation(s) in RCA: 319] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Anoikis (or cell-detachment-induced apoptosis) is a self-defense strategy that organisms use to eliminate 'misplaced' cells, i.e. cells that are in an inappropriate location. Occasionally, detached or misplaced cells can overcome anoikis and survive for a certain period of time in the absence of the correct signals from the extracellular matrix (ECM). If cells are able to adapt to their new environment, then they have probably become anchorage-independent, which is one of the hallmarks of cancer cells. Anoikis resistance and anchorage-independency allow tumor cells to expand and invade adjacent tissues, and to disseminate through the body, giving rise to metastasis. Thus, overcoming anoikis is a crucial step in a series of changes that a tumor cell undergoes during malignant transformation. Tumor cells have developed a variety of strategies to bypass or overcome anoikis. Some strategies consist of adaptive cellular changes that allow the cells to behave as they would in the correct environment, so that induction of anoikis is aborted. Other strategies aim to counteract the negative effects of anoikis induction by hyperactivating survival and proliferative cascades. The recently discovered processes of autophagy and entosis also highlight the contribution of these mechanisms to rendering the cells in a dormant state until they receive a signal initiated at the ECM, thereby circumventing anoikis. In all situations, the final outcome is the ability of the tumor to grow and metastasize. A better understanding of the mechanisms underlying anoikis resistance could help to counteract tumor progression and prevent metastasis formation.
Collapse
Affiliation(s)
- Marta C Guadamillas
- Integrin Signaling Laboratory, Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | | | | |
Collapse
|
217
|
Xiong H, Hong J, Du W, Lin YW, Ren LL, Wang YC, Su WY, Wang JL, Cui Y, Wang ZH, Fang JY. Roles of STAT3 and ZEB1 proteins in E-cadherin down-regulation and human colorectal cancer epithelial-mesenchymal transition. J Biol Chem 2011; 287:5819-32. [PMID: 22205702 DOI: 10.1074/jbc.m111.295964] [Citation(s) in RCA: 256] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The progression of colorectal carcinoma (CRC) to invasive and metastatic disease may involve localized occurrences of epithelial-mesenchymal transition (EMT). However, mechanisms of the EMT process in CRC progression are not fully understood. We previously showed that knockdown of signal transducer and activator of transcription 3 (STAT3) up-regulated E-cadherin (a key component in EMT progression) in CRC. In this study, we examined the roles of STAT3 in CRC EMT and ZEB1, an EMT inducer, in STAT3-induced down-regulation of E-cadherin. Knockdown of STAT3 significantly increased E-cadherin and decreased N-cadherin and vimentin expressions in highly invasive LoVo CRC cells. Meanwhile, overexpression of STAT3 significantly reduced E-cadherin and enhanced N-cadherin and vimentin expressions in weakly invasive SW1116 CRC cells. Activation of STAT3 significantly increased CRC cell invasiveness and resistance to apoptosis. Knockdown of STAT3 dramatically enhanced chemosensitivity of CRC cells to fluorouracil. STAT3 regulated ZEB1 expression in CRC cells, and the STAT3-induced decrease in E-cadherin and cell invasion depended on activation of ZEB1 in CRC cells. Additionally, pSTAT3(Tyr-705) and ZEB1 expressions were significantly correlated with TNM (tumor, lymph node, and metastasis stages) (p < 0.01). In conclusion, STAT3 may directly mediate EMT progression and regulate ZEB1 expression in CRC. ZEB1 may participate in STAT3-induced cell invasion and E-cadherin down-regulation in CRC cells. The expressions of pSTAT3(Tyr-705) and ZEB1 may be positively associated with CRC metastasis. Our data may provide potential targets to prevent and/or treat CRC invasion and metastasis.
Collapse
Affiliation(s)
- Hua Xiong
- Gastrointestinal Division, Shanghai Jiao-Tong University School of Medicine Renji Hospital, Shanghai Institution of Digestive Disease, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health (Shanghai Jiao-Tong University), 145 Middle Shandong Road, Shanghai 200001, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
218
|
Side population cells in human gallbladder cancer cell line GBC-SD regulated by TGF-β-induced epithelial-mesenchymal transition. ACTA ACUST UNITED AC 2011; 31:749-755. [DOI: 10.1007/s11596-011-0671-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Indexed: 12/31/2022]
|
219
|
Alternative Splicing of Fibroblast Growth Factor Receptor IgIII Loops in Cancer. J Nucleic Acids 2011; 2012:950508. [PMID: 22203889 PMCID: PMC3238399 DOI: 10.1155/2012/950508] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2011] [Revised: 07/27/2011] [Accepted: 08/07/2011] [Indexed: 02/06/2023] Open
Abstract
Alternative splicing
of the IgIII loop of fibroblast growth factor
receptors (FGFRs) 1–3 produces b- and
c-variants of the receptors with distinctly
different biological impact based on their
distinct ligand-binding spectrum. Tissue-specific expression of these splice variants
regulates interactions in embryonic development,
tissue maintenance and repair, and cancer.
Alterations in FGFR2 splicing are involved in
epithelial mesenchymal transition that produces
invasive, metastatic features during tumor
progression.
Recent research has elucidated regulatory factors that determine
the splice choice both on the level of exogenous signaling events
and on the RNA-protein interaction level. Moreover, methodology
has been developed that will enable the in depth analysis of
splicing events during tumorigenesis and provide further insight on
the role of FGFR 1–3 IIIb and IIIc in the pathophysiology of
various malignancies. This paper aims to summarize expression
patterns in various tumor types and outlines possibilities for
further analysis and application.
Collapse
|
220
|
Goulioumis AK, Varakis J, Goumas P, Papadaki H. Androgen receptor in laryngeal carcinoma: could there be an androgen-refractory tumor? ISRN ONCOLOGY 2011; 2011:180518. [PMID: 22191056 PMCID: PMC3236417 DOI: 10.5402/2011/180518] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2011] [Accepted: 10/23/2011] [Indexed: 11/23/2022]
Abstract
Androgen receptors (ARs) which are implicated in the pathogenesis of several malignancies can also be a possible downstream effector in laryngeal cancer. In the present study, 97 invasive squamous laryngeal carcinomas were studied by immunohistochemistry for protein expression of AR. Androgen receptors were expressed in 52.6% of tumor specimens, suggesting their implication in the pathogenesis of this tumor. Our study's aim was to investigate the hypothetical scenario of an androgen refractory laryngeal carcinoma where androgen receptors can be activated by nodal molecules in the course of an Epithelial-to-mesenchymal transition (EMT) phenomenon. In line with this we correlated AR expression with the expression of ILK, p-Akt, E-cadherin, β-catenin in our sample as well as with tumor grade and TNM stage. A reverse correlation between nuclear AR and cytoplasmic ILK expression was evidenced, indicating an interaction of those molecules in laryngeal carcinoma. Finally in our material, in those carcinomas that were expressing ARs, stronger nuclear expression of the receptor was characterized by poorer cell differentiation and correlated with the acquisition of EMT features like E-cadherin loss and β-catenin translocation raising a question whether activated ARs can drive an EMT procedure.
Collapse
Affiliation(s)
- Anastasios K Goulioumis
- Department of Anatomy, School of Medicine, University of Patras, Terpsitheas 61, 26442 Patras, Greece
| | | | | | | |
Collapse
|
221
|
Tian X, Liu Z, Niu B, Zhang J, Tan TK, Lee SR, Zhao Y, Harris DCH, Zheng G. E-cadherin/β-catenin complex and the epithelial barrier. J Biomed Biotechnol 2011; 2011:567305. [PMID: 22007144 PMCID: PMC3191826 DOI: 10.1155/2011/567305] [Citation(s) in RCA: 333] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 07/25/2011] [Indexed: 12/13/2022] Open
Abstract
E-Cadherin/β-catenin complex plays an important role in maintaining epithelial integrity and disrupting this complex affect not only the adhesive repertoire of a cell, but also the Wnt-signaling pathway. Aberrant expression of the complex is associated with a wide variety of human malignancies and disorders of fibrosis resulting from epithelial-mesenchymal transition. These associations provide insights into the complexity that is likely responsible for the fibrosis/tumor suppressive action of E-cadherin/β-catenin.
Collapse
Affiliation(s)
- Xinrui Tian
- Centre for Transplantation and Renal Research, Westmead Millennium Institute, The University of Sydney, NSW 2145, Australia
- Department of Respiratory, Second Hospital of Shanxi Medical University, Shanxi, Taiyuan 030001, China
| | - Zhuola Liu
- Department of Respiratory, Second Hospital of Shanxi Medical University, Shanxi, Taiyuan 030001, China
| | - Bo Niu
- Biotechnology Laboratory, Capital Institute of Pediatrics, Beijing 100020, China
| | - Jianlin Zhang
- Centre for Transplantation and Renal Research, Westmead Millennium Institute, The University of Sydney, NSW 2145, Australia
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, China
| | - Thian Kui Tan
- Centre for Transplantation and Renal Research, Westmead Millennium Institute, The University of Sydney, NSW 2145, Australia
| | - So Ra Lee
- Centre for Transplantation and Renal Research, Westmead Millennium Institute, The University of Sydney, NSW 2145, Australia
| | - Ye Zhao
- Centre for Transplantation and Renal Research, Westmead Millennium Institute, The University of Sydney, NSW 2145, Australia
| | - David C. H. Harris
- Centre for Transplantation and Renal Research, Westmead Millennium Institute, The University of Sydney, NSW 2145, Australia
| | - Guoping Zheng
- Centre for Transplantation and Renal Research, Westmead Millennium Institute, The University of Sydney, NSW 2145, Australia
| |
Collapse
|
222
|
Sossey-Alaoui K, Downs-Kelly E, Das M, Izem L, Tubbs R, Plow EF. WAVE3, an actin remodeling protein, is regulated by the metastasis suppressor microRNA, miR-31, during the invasion-metastasis cascade. Int J Cancer 2011; 129:1331-43. [PMID: 21105030 PMCID: PMC3081370 DOI: 10.1002/ijc.25793] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Accepted: 10/26/2010] [Indexed: 12/21/2022]
Abstract
WAVE3, an actin cytoskeleton remodeling protein, is highly expressed in advanced stages of breast cancer and influences tumor cell invasion. Loss of miR-31 has been associated with cancer progression and metastasis. Here, we show that the activity of WAVE3 to promote cancer cell invasion is regulated by miR-31. An inverse correlation was demonstrated between expression levels of WAVE3 and miR-31 in invasive versus noninvasive breast cancer cell lines. miR-31 directly targeted the 3'-UTR of the WAVE3 mRNA and inhibited its expression in the invasive cancer cells, i.e., miR-31-mediated down-regulation of WAVE3 resulted in a significant reduction in the invasive phenotype of cancer cells. This relationship was specific to the loss of WAVE3 expression because re-expression of a miR-31-resistant form of WAVE3 reversed miR-31-mediated inhibition of cancer cell invasion. Furthermore, expression of miR-31 correlates inversely with breast cancer progression in humans, where an increase in expression of miR-31 target genes was observed as the tumors progressed to more aggressive forms. In conclusion, a novel mechanism for the regulation of WAVE3 expression in cancer cells has been identified, which controls the invasive properties of cancer cells. The study also identifies a critical role for WAVE3, downstream of miR-31, in the invasion-metastasis cascade.
Collapse
Affiliation(s)
- Khalid Sossey-Alaoui
- Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| | | | | | | | | | | |
Collapse
|
223
|
Coll TA, Tito LP, Sobarzo CMA, Cebral E. Embryo developmental disruption during organogenesis produced by CF-1 murine periconceptional alcohol consumption. ACTA ACUST UNITED AC 2011; 92:560-74. [PMID: 21922637 DOI: 10.1002/bdrb.20329] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Accepted: 07/11/2011] [Indexed: 01/09/2023]
Abstract
The aim was to study the control females (CF)-1 mouse embryo differentiation, growth, morphology on embryonic E- and N-cadherin expression at midgestation after periconceptional moderate alcohol ingestion. Adult female mice were exposed to 10% ethanol in drinking water for 17 days previous to and up to day 10 of gestation (ethanol-exposed females, EF) and were compared with nonexposed CF. EF presented reduced quantities of E10 to E10.5 embryos, greater percentage of embryos at stages less than E7.5, reduced implantation site numbers/female, and increased resorptions compared with CF. EF-embryo growth was significantly affected as evidenced by reduced cephalic and body sizes of E10 and E10.5 embryos (scanning electron microscopy) and decreased protein content of E10.5 embryos vs. CF embryos. A significantly higher percentage of EF-E10-10.5 embryos presented abnormal neural tube (NT) closure vs. the percentage of CF. E10 embryos from EF presented elevated tissue disorganization, pyknosis and nuclear condensation in somites, mesenchymal and neuroepithelial tissue. Immunohistochemical E- and N-cadherin distribution patterns were similar in organic structures of E10 embryos between groups. However, western blot revealed that E- and N-cadherin expression levels were significantly increased in EF-derived embryos vs. controls. Perigestational ethanol consumption by CF-1 mice induced significant damage in the organogenic embryogenesis by producing delayed differentiation, growth deficiencies, and increasing the frequency of NT defects. Ethanol exposure may disrupt cell-cell adhesion leading to upregulation of E- and N-cadherin expression suggesting that deregulation of cell adhesion molecules could be involved in the disruption of embryo development at organogenesis in CF-1 mouse.
Collapse
Affiliation(s)
- Tamara A Coll
- Laboratorio de Reproducción y Fisiopatología Materno-Embrionaria, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-CONICET), Departamento de Biodiversidad y Biología Experimental (DBBE), Universidad de Buenos Aires (UBA), Argentina
| | | | | | | |
Collapse
|
224
|
Toiyama Y, Yasuda H, Saigusa S, Matushita K, Fujikawa H, Tanaka K, Mohri Y, Inoue Y, Goel A, Kusunoki M. Co-expression of hepatocyte growth factor and c-Met predicts peritoneal dissemination established by autocrine hepatocyte growth factor/c-Met signaling in gastric cancer. Int J Cancer 2011; 130:2912-21. [PMID: 21796631 DOI: 10.1002/ijc.26330] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Accepted: 06/29/2011] [Indexed: 12/15/2022]
Abstract
Epithelial-mesenchymal transition (EMT) promotes and facilitates migration and invasion of epithelial tumor cells. EMT is induced by factors such as hepatocyte growth factor (HGF). This study aimed to establish whether the HGF/c-Met pathway is associated with gastric cancer metastasis; especially peritoneal dissemination. HGF and c-Met expression and EMT-related molecules were evaluated using real-time PCR and immunohistochemistry. The role of the HGF/c-Met pathway in EMT and anoikis was determined, and kinase inhibitor SU11274 was tested for its ability to block HGF-induced biological effects. In HGF(-) /c-Met(+) gastric cancer cells, recombinant HGF promoted an EMT phenotype that was characterized by morphology, impaired E-cadherin and induction of vimentin. HGF promoted cell growth, invasiveness and migration and inhibition of anoikis. SU11274 blocked HGF-induced EMT and biological effects in vitro. In HGF(+) /c-Met(+) gastric cancer cells, HGF did not affect the biological outcome of EMT and anoikis, but SU11274 exerted the same inhibitory effects as in HGF(-) /c-Met(+) cells. In vivo, HGF(+) /c-Met(+) gastric cancer cells only established peritoneal dissemination and SU11274 inhibited tumor growth. Clinically, HGF expression was significantly correlated with c-Met expression in gastric cancer. Increased HGF and c-Met had a significant association with poor prognosis and predicted peritoneal dissemination. We demonstrated that the HGF/c-Met pathway induces EMT and inhibition of anoikis in gastric cancer cells. Co-expression of HGF and c-Met has the potential to promote peritoneal dissemination in gastric cancer. Blockade of the autocrine HGF/c-Met pathway could be clinically useful for the treatment of peritoneal dissemination in gastric cancer.
Collapse
Affiliation(s)
- Yuji Toiyama
- Department of Gastrointestinal and Pediatric Surgery, Division of Reparative Medicine, Institute of Life Sciences, Graduate School of Medicine, Mie University, Mie, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
225
|
Saini S, Yamamura S, Majid S, Shahryari V, Hirata H, Tanaka Y, Dahiya R. MicroRNA-708 induces apoptosis and suppresses tumorigenicity in renal cancer cells. Cancer Res 2011; 71:6208-19. [PMID: 21852381 DOI: 10.1158/0008-5472.can-11-0073] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancer pathogenesis is restricted by stresses that compromise cell division and survival. In this study, we identify miR-708, a little studied member of a set of microRNAs that have been implicated in stress control, as an important tumor suppressor in renal cell carcinoma (RCC). miR-708 expression was attenuated widely in human RCC specimens. Restoration of miR-708 expression in RCC cell lines decreased cell growth, clonability, invasion, and migration and elicited a dramatic increase in apoptosis. Moreover, intratumoral delivery of miR-708 was sufficient to trigger in vivo regression of established tumors in murine xenograft models of human RCC. Investigation of the targets of miR-708 identified the inhibitor of apoptosis protein survivin as important. siRNA-mediated knockdown of survivin partially phenocopied miR-708 overexpression suggesting that the proapoptotic role of miR-708 may be mediated primarily through survivin regulation. Additionally, we identified the E-cadherin regulators ZEB2 and BMI1 as likely miR-708 targets. Taken together, our findings define a major tumor suppressive role for miR-708, which may offer an attractive new target for prognostic and therapeutic intervention in RCC.
Collapse
Affiliation(s)
- Sharanjot Saini
- Department of Urology, Veterans Affairs Medical Center, San Francisco, California 94121, USA
| | | | | | | | | | | | | |
Collapse
|
226
|
Majumder A, Kirabo A, Karrupiah K, Tsuda S, Caldwell-Busby J, Cardounel AJ, Keseru GM, Sayeski PP. Cell death induced by the Jak2 inhibitor, G6, correlates with cleavage of vimentin filaments. Biochemistry 2011; 50:7774-86. [PMID: 21823612 DOI: 10.1021/bi200847n] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Hyperkinetic Jak2 tyrosine kinase signaling has been implicated in several human diseases including leukemia, lymphoma, myeloma, and the myeloproliferative neoplasms. Using structure-based virtual screening, we previously identified a novel Jak2 inhibitor named G6. We showed that G6 specifically inhibits Jak2 kinase activity and suppresses Jak2-mediated cellular proliferation. To elucidate the molecular and biochemical mechanisms by which G6 inhibits Jak2-mediated cellular proliferation, we treated Jak2-V617F expressing human erythroleukemia (HEL) cells for 12 h with either vehicle control or 25 μM of the drug and compared protein expression profiles using two-dimensional gel electrophoresis. One differentially expressed protein identified by electrospray mass spectroscopy was the intermediate filament protein, vimentin. It was present in DMSO treated cells but absent in G6 treated cells. HEL cells treated with G6 showed both time- and dose-dependent cleavage of vimentin as well as a marked reorganization of vimentin intermediate filaments within intact cells. In a mouse model of Jak2-V617F mediated human erythroleukemia, G6 also decreased the levels of vimentin protein, in vivo. The G6-induced cleavage of vimentin was found to be Jak2-dependent and calpain-mediated. Furthermore, we found that intracellular calcium mobilization is essential and sufficient for the cleavage of vimentin. Finally, we show that the cleavage of vimentin intermediate filaments, per se, is sufficient to reduce HEL cell viability. Collectively, these results suggest that G6-induced inhibition of Jak2-mediated pathogenic cell growth is concomitant with the disruption of intracellular vimentin filaments. As such, this work describes a novel pathway for the targeting of Jak2-mediated pathological cell growth.
Collapse
Affiliation(s)
- Anurima Majumder
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida 32610, United States
| | | | | | | | | | | | | | | |
Collapse
|
227
|
Abstract
OBJECTIVES These studies examined the effect of homozygous deletion of vasoactive intestinal peptide receptor type 1 (VPAC1) on development and function of intestines and pancreas. METHODS Genetically engineered VPAC1-null mutant mice were monitored for growth, development, and glucose homeostasis. Expression of VPAC1 was examined during embryonic development using VPAC1 promoter-driven β-galactosidase transgenic mice. RESULTS Homozygous deletion of VPAC1 resulted in fetal, neonatal, and postweaning death owing to failure to thrive, intestinal obstruction, and hypoglycemia. Histological findings demonstrated disorganized hyperproliferation of intestinal epithelial cells with mucus deposition and bowel wall thickening. The pancreas demonstrated small dysmorphic islets of Langerhans containing α, β, and δ cells. Expression of a VPAC1 promoter-driven transgene was observed in E12.5 and E14.5 intestinal epithelial and pancreatic endocrine cells. Vasoactive intestinal peptide receptor type 1-null mutant animals had lower baseline blood glucose levels compared to both heterozygous and wild-type littermates. Vasoactive intestinal peptide receptor type 1-deficient mice responded to oral glucose challenge with normal rise in blood glucose followed by rapid hypoglycemia and failure to restore baseline glucose levels. Insulin challenge resulted in profound hypoglycemia and inadequate glucose homeostasis in VPAC1-null mutant animals. CONCLUSIONS These observations support a role for VPAC1 during embryonic and neonatal development of intestines and endocrine pancreas.
Collapse
|
228
|
Kevans D, Wang LM, Sheahan K, Hyland J, O'Donoghue D, Mulcahy H, O'Sullivan J. Epithelial-mesenchymal transition (EMT) protein expression in a cohort of stage II colorectal cancer patients with characterized tumor budding and mismatch repair protein status. Int J Surg Pathol 2011; 19:751-60. [PMID: 21791486 DOI: 10.1177/1066896911414566] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The relationship between tumor budding, epithelial-mesenchymal transition (EMT) protein expression, and survival has not been closely examined in stage II colorectal cancer (CRC). This study aimed to assess proteins implicated in EMT and to correlate their expression with tumor budding, microsatellite status, and survival. METHODS A total of 258 stage II CRCs were identified (tumor budding characterized in 122 cases). Immunohistochemistry for LAMC2, E cadherin, cathepsin L, and β catenin using tissue microarrays was performed. EMT and mismatch repair (MMR) protein expression were correlated with tumor budding and survival. RESULTS LAMC2 positivity (P < .001) and low membranous β catenin (P = .056) were associated with tumor budding. In a univariate survival analysis, tumor budding (P < .001), LAMC2 positivity (P < .03), and stromal cytoplasmic cathepsin L (P = .025) predicted poorer prognosis. Multivariate analysis showed tumor budding to be the only variable independently associated with survival: hazard ratio = 7.9 (95% confidence interval = 3-21); P < .001. Tumor budding was more frequent in microsatellite-stable (MSS) versus microsatellite-instable (MSI) tumors: 48% versus 26%, respectively; P = .087. MSS cases exhibited reduced membranous β catenin (P = .002) and increased cytoplasmic and nuclear β catenin (P < .001) compared with MSI cases. CONCLUSION Epithelial mesenchymal protein expression plays a key role in tumor budding and prognosis in early-stage colorectal cancer and requires further evaluation.
Collapse
Affiliation(s)
- David Kevans
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland.
| | | | | | | | | | | | | |
Collapse
|
229
|
Abstract
Lung cancer is the leading cause of cancer death worldwide. Cancer metastasis and resistance to treatment (including radiotherapy, chemotherapy and targeted therapy) are two major causes for the poor survival of lung cancer patients. Epithelial-mesenchymal transition (EMT) is involved in cancer cell invasion, resistance to apoptosis and stem cell features. The process of EMT is controlled by a group of transcriptional factors, zinc finger proteins and basic helix-loop-helix factors. Signaling pathways activated by intrinsic or extrinsic stimuli converge on these transcriptional factors and regulated the phenotypic changes of cancer cells. These EMT regulators may play an important role in cancer progression. In lung cancer, Slug is the most thoroughly investigated EMT regulator. The expression of Slug is associated with lung cancer invasion and resistance to target therapy. In this review, we focus on the current understanding of the role of Slug in the carcinogenesis and progression of lung cancer.
Collapse
Affiliation(s)
- Jin-Yuan Shih
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Chung-Shan South Road, Taipei 100, Taiwan
| | | |
Collapse
|
230
|
Tian M, Neil JR, Schiemann WP. Transforming growth factor-β and the hallmarks of cancer. Cell Signal 2011; 23:951-62. [PMID: 20940046 PMCID: PMC3076078 DOI: 10.1016/j.cellsig.2010.10.015] [Citation(s) in RCA: 208] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 10/01/2010] [Indexed: 02/07/2023]
Abstract
Tumorigenesis is in many respects a process of dysregulated cellular evolution that drives malignant cells to acquire six phenotypic hallmarks of cancer, including their ability to proliferate and replicate autonomously, to resist cytostatic and apoptotic signals, and to induce tissue invasion, metastasis, and angiogenesis. Transforming growth factor-β (TGF-β) is a potent pleiotropic cytokine that functions as a formidable barrier to the development of cancer hallmarks in normal cells and tissues. Paradoxically, tumorigenesis counteracts the tumor suppressing activities of TGF-β, thus enabling TGF-β to stimulate cancer invasion and metastasis. Fundamental gaps exist in our knowledge of how malignant cells overcome the cytostatic actions of TGF-β, and of how TGF-β stimulates the acquisition of cancer hallmarks by developing and progressing human cancers. Here we review the molecular and cellular mechanisms that underlie the ability of TGF-β to mediate tumor suppression in normal cells, and conversely, to facilitate cancer progression and disease dissemination in malignant cells.
Collapse
Affiliation(s)
- Maozhen Tian
- Division of General Medical Sciences–Oncology, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
| | - Jason R. Neil
- Department of Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - William P. Schiemann
- Division of General Medical Sciences–Oncology, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106
| |
Collapse
|
231
|
Ueno NT, Zhang D. Targeting EGFR in Triple Negative Breast Cancer. J Cancer 2011; 2:324-8. [PMID: 21716849 PMCID: PMC3119395 DOI: 10.7150/jca.2.324] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 05/28/2011] [Indexed: 12/22/2022] Open
Abstract
Our preliminary data show that erlotinib inhibits Triple-negative breast cancer (TNBC) in a xenograft model. However, inhibition of metastasis by erlotinib is accompanied by nonspecific effects because erlotinib can inhibit other kinases; thus, more direct targets that regulate TNBC metastasis need to be identified to improve its therapeutic efficacy.
Collapse
Affiliation(s)
- Naoto T Ueno
- 1. Breast Cancer Translational Research Laboratory, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
232
|
Nagaharu K, Zhang X, Yoshida T, Katoh D, Hanamura N, Kozuka Y, Ogawa T, Shiraishi T, Imanaka-Yoshida K. Tenascin C induces epithelial-mesenchymal transition-like change accompanied by SRC activation and focal adhesion kinase phosphorylation in human breast cancer cells. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:754-63. [PMID: 21281808 DOI: 10.1016/j.ajpath.2010.10.015] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2010] [Revised: 10/14/2010] [Accepted: 10/19/2010] [Indexed: 12/20/2022]
Abstract
Tenascin C (TNC) is an extracellular matrix glycoprotein up-regulated in solid tumors. Higher TNC expression is shown in invading fronts of breast cancer, which correlates with poorer patient outcome. We examined whether TNC induces epithelial-mesenchymal transition (EMT) in breast cancer. Immunohistochemical analysis of invasive ductal carcinomas showed that TNC deposition was frequent in stroma with scattered cancer cells in peripheral margins of tumors. The addition of TNC to the medium of the MCF-7 breast cancer cells caused EMT-like change and delocalization of E-cadherin and β-catenin from cell-cell contact. Although amounts of E-cadherin and β-catenin were not changed after EMT in total lysates, they were increased in the Triton X-100-soluble fractions, indicating movement from the membrane into the cytosol. In wound healing assay, cells were scattered from wound edges and showed faster migration after TNC treatment. The EMT phenotype was correlated with SRC activation through phosphorylation at Y418 and phosphorylation of focal adhesion kinase (FAK) at Y861 and Y925 of SRC substrate sites. These phosphorylated proteins colocalized with αv integrin-positive adhesion plaques. A neutralizing antibody against αv or a SRC kinase inhibitor blocked EMT. TNC could induce EMT-like change showing loss of intercellular adhesion and enhanced migration in breast cancer cells, associated with FAK phosphorylation by SRC; this may be responsible for the observed promotion of TNC in breast cancer invasion.
Collapse
Affiliation(s)
- Keiki Nagaharu
- Department of Pathology and Matrix Biology, Graduate School of Medicine, Mie University, Mie, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
233
|
Gordon SR. The effects of soybean agglutinin binding on the corneal endothelium and the re-establishment of an intact monolayer following injury--a short review. J Tissue Viability 2011; 20:20-9. [PMID: 20022250 DOI: 10.1016/j.jtv.2009.11.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Revised: 11/23/2009] [Accepted: 11/24/2009] [Indexed: 12/29/2022]
Abstract
This short review summarizes the localization and effects of the plant lectin soybean agglutinin (SBA) on the injured and non-injured organ-cultured rat corneal endothelium. Although the tissue exists as a non-cycling monolayer on the posterior corneal surface a circular freeze injury promotes wound repair as cells initiate DNA synthesis, mitosis and migration. As a result, by 24 h post-injury, endothelial cells express a surface protein that binds SBA in a diffuse punctate pattern, which by 48 h after injury, becomes confined to the cell periphery. As healing proceeds, SBA binding dramatically declines, such that, only scattered binding is observed by 72 h after wounding. In non-injured organ-cultured endothelia, weak SBA binding appears 24 h after explanation but becomes prominently detected around the cell periphery by 48 h. Incubating injured or non-injured endothelia in SBA leads to alterations in their cellular appearance due to the fact that lectin exposure results in the disruption of the actin cytoskeleton. Although this does not affect migration, treatment with either SBA or N-acetylgalactosamine (the SBA binding sugar) does interfere with the reestablishment of cell-cell contact. It is postulated that the surface protein that binds SBA is expressed during conditions that are stressful to the tissue. During organ-culture the protein's appearance suggests a cellular response to explantation in order to enhance or maintain monolayer integrity. In wound repair its appearance may serve to establish preliminary cell-cell contact during the restoration of the endothelial monolayer.
Collapse
Affiliation(s)
- Sheldon R Gordon
- Department of Biological Sciences, Oakland University, 2200 N. Squirrel Road, Rochester, MI 48309, USA
| |
Collapse
|
234
|
Liao KA, Tsay YG, Huang LC, Huang HY, Li CF, Wu TF. Search for the tumor-associated proteins of oral squamous cell carcinoma collected in Taiwan using proteomics strategy. J Proteome Res 2011; 10:2347-2358. [PMID: 21322651 DOI: 10.1021/pr101146w] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Squamous cell carcinoma (SCC) accounts for more than 90% of malignant tumors of the oral cavity. In Taiwan, oral squamous cell carcinoma (OSCC) is among the most frequent malignancies, largely due to betal quid chewing. Despite the recent improvement in treatment results, the long-term outcome of OSCC generally remains poor, especially for those with advanced diseases. It is therefore desirable to identify potential biomarkers that may aid in risk stratification and perhaps the development of therapeutic targets. In this study, we exploited two-dimensional gel electrophoresis (2-DE) coupled with mass spectrometry to compare the proteome maps of 10 OSCC specimens with their adjacent nontumorous epithelia to identify differentially expressed proteins. Comparative proteomics indicated that 17 proteins were differentially expressed in OSCC with 11 up-regulated and 6 down-regulated proteins. These deregulated proteins participated in cytoskeletal functions, cell signaling, antiapoptosis, angiogenesis, lipid metabolism, drug metabolism, and protein translation/turnover. They were all associated with tumor development in various cancers. Among the dys-regulated proteins, the immunoexpression of three proteins including nicotinamide N-methyltransferase, apolipoprotein AI, and 14-3-3 zeta were evaluated in 38 OSCCs of testing cohort to confirm the proteomics data. Subsequently, the expression of 14-3-3 zeta, as the most relevant to OSCC progression determined by testing cohort, was further assessed in 80 OSCCs of independent validation cohort to identify the clinical relevance of its expression. By this comprehensive study, we identified 14-3-3 zeta as the only prognosticator of local recurrence-free survival (LRFS) and also an independently predicted factor of disease-specific survival (DSS).
Collapse
Affiliation(s)
- Kuo-An Liao
- Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan, 710, Taiwan
| | | | | | | | | | | |
Collapse
|
235
|
Davies D. Cell-extracellular matrix versus cell-cell interactions during the development of the cochlear-vestibular ganglion. J Neurosci Res 2011; 89:1375-87. [PMID: 21557292 DOI: 10.1002/jnr.22664] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 01/31/2011] [Accepted: 03/17/2011] [Indexed: 11/06/2022]
Abstract
Cells destined to become the neurones of the cochlear-vestibular ganglion (CVG) originate within the otic epithelium. Early in development they detach from their neighbors and migrate out of the epithelium, where they coalesce to form the CVG. To accomplish this process, the neuroblasts must modify their interactions with other cells within the epithelium and with proteins in the extracellular matrix to allow for repositioning. The aim of this study was to investigate the roles of the major families of adhesion molecules that mediate cellular interactions with the extracellular matrix, the integrins, and with other cells, the cadherins, in neuroblast segregation from the otic epithelium. The expression of classical cadherins increased in migrating neuroblasts compared with the otic epithelium. Quantitative RT-PCR revealed that this was concomitant with down-regulation of E-cadherin and up-regulation of N-cadherin in the migrating cells. In contrast, the level of β1 integrin expression by the epithelium was maintained in migrating neuroblasts. However, although multiple integrin ligands were expressed within the otic basement membrane at this stage of development, only fibronectin (FN) supported neuroblast migration along the substrate in vitro. Inhibition of β1 integrins resulted in significantly reduced linear migration on FN. Importantly, neuroblasts retained the ability to segregate from the epithelium but remained compacted immediately adjacent to the originating tissue, suggesting dominance of cell-cell over cell-matrix interactions. These data suggest that the balance between cell-cell and cell-substratum interactions directs otic neuroblast migration and gangliogenesis.
Collapse
Affiliation(s)
- Dawn Davies
- School of Physiology and Pharmacology, University of Bristol, Bristol, United Kingdom.
| |
Collapse
|
236
|
Brachyury expression predicts poor prognosis at early stages of colorectal cancer. Eur J Cancer 2011; 47:1080-5. [DOI: 10.1016/j.ejca.2010.11.015] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2010] [Accepted: 11/23/2010] [Indexed: 11/17/2022]
|
237
|
Palena C, Fernando RI, Litzinger MT, Hamilton DH, Huang B, Schlom J. Strategies to target molecules that control the acquisition of a mesenchymal-like phenotype by carcinoma cells. Exp Biol Med (Maywood) 2011; 236:537-45. [PMID: 21427233 PMCID: PMC3508693 DOI: 10.1258/ebm.2011.010367] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The switch of carcinoma cells from an epithelial to a mesenchymal-like phenotype, via a process designated 'epithelial-to-mesenchymal transition (EMT),' has been recognized as a relevant step in the metastasis of solid tumors. Additionally, this phenotypic switch of carcinoma cells has been associated with the acquisition of tumor resistance mechanisms that reduce the antitumor effects of radiation, chemotherapy and some small-molecule-targeted therapies. As multiple signaling pathways and transcriptional regulators that play a role in this phenotypic switch are being identified, novel strategies can be designed to specifically target tumor cells with this metastatic and resistant phenotype. In particular, this review focuses on the potential use of cancer vaccine strategies to target tumor cells that exhibit a mesenchymal-like phenotype, with an emphasis on the characterization of a novel tumor antigen, Brachyury, which we have identified as a critical regulator of EMT in human cancer cells.
Collapse
Affiliation(s)
- Claudia Palena
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
238
|
Tseng CH, Murray KD, Jou MF, Hsu SM, Cheng HJ, Huang PH. Sema3E/plexin-D1 mediated epithelial-to-mesenchymal transition in ovarian endometrioid cancer. PLoS One 2011; 6:e19396. [PMID: 21559368 PMCID: PMC3084850 DOI: 10.1371/journal.pone.0019396] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Accepted: 03/29/2011] [Indexed: 12/20/2022] Open
Abstract
Cancer cells often employ developmental cues for advantageous growth and metastasis. Here, we report that an axon guidance molecule, Sema3E, is highly expressed in human high-grade ovarian endometrioid carcinoma, but not low-grade or other ovarian epithelial tumors, and facilitates tumor progression. Unlike its known angiogenic activity, Sema3E acted through Plexin-D1 receptors to augment cell migratory ability and concomitant epithelial-to-mesenchymal transition (EMT). Sema3E-induced EMT in ovarian endometrioid cancer cells was dependent on nuclear localization of Snail1 through activation of phosphatidylinositol-3-kinase and ERK/MAPK. RNAi-mediated knockdown of Sema3E, Plexin-D1 or Snail1 in Sema3E-expressing tumor cells resulted in compromised cell motility, concurrent reversion of EMT and diminished nuclear localization of Snail1. By contrast, forced retention of Snail1 within the nucleus of Sema3E-negative tumor cells induced EMT and enhanced cell motility. These results show that in addition to the angiogenic effects of Sema3E on tumor vascular endothelium, an EMT strategy could be exploited by Sema3E/Plexin-D1 signaling in tumor cells to promote cellular invasion/migration.
Collapse
Affiliation(s)
- Chun-Hsien Tseng
- Graduate Institute of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Karl D. Murray
- Center for Neuroscience, University of California Davis, Davis, California, United States of America
| | - Mu-Fan Jou
- Graduate Institute of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Su-Ming Hsu
- Graduate Institute of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Hwai-Jong Cheng
- Center for Neuroscience, University of California Davis, Davis, California, United States of America
- * E-mail: (H-JC); (P-HH)
| | - Pei-Hsin Huang
- Graduate Institute of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
- * E-mail: (H-JC); (P-HH)
| |
Collapse
|
239
|
Galoian KA, Temple TH, Galoyan A. Cytostatic effect of novel mTOR inhibitor, PRP-1 (galarmin) in MDA 231 (ER-) breast carcinoma cell line. PRP-1 inhibits mesenchymal tumors. Tumour Biol 2011; 32:745-51. [PMID: 21494810 DOI: 10.1007/s13277-011-0176-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 04/03/2011] [Indexed: 12/18/2022] Open
Abstract
Activation of the PI3K-Akt-mTOR pathway is implicated both in the establishment of tumors and as well as a target for therapy in many types of solid malignancy, its blockade represents an opportunity to improve outcomes in patients with tumors that are associated with poor prognosis. Our experimental data indicates that proline-rich polypeptide-1 (PRP-1, galarmin) is immunomodulator cytokine, produced by hypothalamic neurosecretory cells and exerts its antiproliferative effect on the tumor cells of mesenchymal origin via inhibiting mTOR kinase activity and repressing cell cycle progression. The goal of these investigations was to elucidate the antiproliferative action of PRP-1 on the breast carcinoma cell line MDA 231 (ER-) and to compare PRP-1 action previously reported on other mesenchymal tumors. These experiments confirmed maximum inhibition of cell growth at 0.5 and 1 μg/ml PRP-1 (71% and 63%, respectively) and inhibition at 10 μg/ml of 44%. There was no inhibitory effect observed on luminal T47-D (ER+) cells. Videomicroscopy results demonstrated dividing cells in the cytokine-treated MDA 231 (ER-), suggesting that the cells were not in the state of dormancy. The flow cytometry experiments confirmed that PRP-1-treated cells were accumulated in S phase. No apoptosis, caspase activation, or senescence was detected after treatment with this cytokine. Experiments with mTOR with PRP-1 (10 μg/ml) indicated statistically significant 40% inhibition of mTOR kinase activity in immunoprecipitates of the MDA 231 (ER-) cell line. PRP-1 is a novel mTOR inhibitor with strong antiproliferative action in mesenchymal tumors mostly resistant to radiation and chemotherapy.
Collapse
Affiliation(s)
- Karina A Galoian
- Miller School of Medicine, University of Miami Health System, 1600 N.W 10th avenue, suite 8006 (R-2), Miami, FL 33136, USA.
| | | | | |
Collapse
|
240
|
Activated leukocyte cell adhesion molecule expression and shedding in thyroid tumors. PLoS One 2011; 6:e17141. [PMID: 21364949 PMCID: PMC3043091 DOI: 10.1371/journal.pone.0017141] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Accepted: 01/21/2011] [Indexed: 12/20/2022] Open
Abstract
Activated leukocyte cell adhesion molecule (ALCAM, CD166) is expressed in various tissues, cancers, and cancer-initiating cells. Alterations in expression of ALCAM have been reported in several human tumors, and cell adhesion functions have been proposed to explain its association with cancer. Here we documented high levels of ALCAM expression in human thyroid tumors and cell lines. Through proteomic characterization of ALCAM expression in the human papillary thyroid carcinoma cell line TPC-1, we identified the presence of a full-length membrane-associated isoform in cell lysate and of soluble ALCAM isoforms in conditioned medium. This finding is consistent with proteolytically shed ALCAM ectodomains. Nonspecific agents, such as phorbol myristate acetate (PMA) or ionomycin, provoked increased ectodomain shedding. Epidermal growth factor receptor stimulation also enhanced ALCAM secretion through an ADAM17/TACE-dependent pathway. ADAM17/TACE was expressed in the TPC-1 cell line, and ADAM17/TACE silencing by specific small interfering RNAs reduced ALCAM shedding. In addition, the CGS27023A inhibitor of ADAM17/TACE function reduced ALCAM release in a dose-dependent manner and inhibited cell migration in a wound-healing assay. We also provide evidence for the existence of novel O-glycosylated forms and of a novel 60-kDa soluble form of ALCAM, which is particularly abundant following cell stimulation by PMA. ALCAM expression in papillary and medullary thyroid cancer specimens and in the surrounding non-tumoral component was studied by western blot and immunohistochemistry, with results demonstrating that tumor cells overexpress ALCAM. These findings strongly suggest the possibility that ALCAM may have an important role in thyroid tumor biology.
Collapse
|
241
|
Gauger KJ, Chenausky KL, Murray ME, Schneider SS. SFRP1 reduction results in an increased sensitivity to TGF-β signaling. BMC Cancer 2011; 11:59. [PMID: 21303533 PMCID: PMC3041779 DOI: 10.1186/1471-2407-11-59] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Accepted: 02/08/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Transforming growth factor (TGF)-β plays a dual role during mammary gland development and tumorigenesis and has been shown to stimulate epithelial-mesenchymal transition (EMT) as well as cellular migration. The Wnt/β-catenin pathway is also implicated in EMT and inappropriate activation of the Wnt/β-catenin signaling pathway leads to the development of several human cancers, including breast cancer. Secreted frizzled-related protein 1 (SFRP1) antagonizes this pathway and loss of SFRP1 expression is frequently observed in breast tumors and breast cancer cell lines. We previously showed that when SFRP1 is knocked down in immortalized non-malignant mammary epithelial cells, the cells (TERT-siSFRP1) acquire characteristics associated with breast tumor initiating cells. The phenotypic and genotypic changes that occur in response to SFRP1 loss are consistent with EMT, including a substantial increase in the expression of ZEB2. Considering that ZEB2 has been shown to interact with mediators of TGF-β signaling, we sought to determine whether TGF-β signaling is altered in TERT-siSFRP1 cells. METHODS Luciferase reporter assays and real-time PCR analysis were employed to measure TGF-β transcriptional targets. Western blot analysis was used to evaluate TGF-β-mediated ERK1/2 phosphorylation. Migration chamber assays were utilized to quantify cellular migration. TERT-siSFRP1 cells were transfected with Stealth RNAi™ siRNA in order to knock-down the expression of ZEB2. RESULTS TERT-siSFRP1 cells exhibit a significant increase in both TGF-β-mediated luciferase activity as well as TGF-β transcriptional targets, including Integrin β3 and PAI-1. Phosphorylation of ERK1/2 is increased in TERT-siSFRP1 cells in response to enhanced TGF-β signaling. Furthermore, when the TGF-β pathway is blocked with a TGF-βR antagonist (LY364947), cellular migration is significantly hindered. Finally, we found that when ZEB2 is knocked-down, there is a significant reduction in the expression of exogeneous and endogenous TGF-β transcriptional targets and cellular migration is impeded. CONCLUSIONS We demonstrate that down-regulation of SFRP1 renders mammary epithelial cells more sensitive to TGF-β signaling which can be partially ameliorated by blocking the expression of ZEB2.
Collapse
Affiliation(s)
- Kelly J Gauger
- Pioneer Valley Life Sciences Institute, Baystate Medical Center, Springfield, MA 01199, USA.
| | | | | | | |
Collapse
|
242
|
Richter P, Umbreit C, Franz M, Berndt A, Grimm S, Uecker A, Böhmer FD, Kosmehl H, Berndt A. EGF/TGFβ1 co-stimulation of oral squamous cell carcinoma cells causes an epithelial-mesenchymal transition cell phenotype expressing laminin 332. J Oral Pathol Med 2011; 40:46-54. [PMID: 20819124 DOI: 10.1111/j.1600-0714.2010.00936.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is suggested to be crucial for the development of an invasive and metastatic carcinoma cell phenotype. Therefore, the definition of this phenotype is of great clinical interest. We recently evidenced vimentin positive cells in oral squamous cell carcinoma (OSCC) invasive front expressing laminin γ2 chain mRNA implicating an EMT origin of these cells. To further elucidate the nature of these cells, we have investigated the relation between EMT criteria and laminin-332 expression in a cell culture model of transforming growth factor beta-1 (TGFβ1)/epithelial growth factor (EGF) long time co-stimulation. We demonstrate that in contrast to TGFβ1 or EGF alone, co-stimulation induces phenotype transition in OSCC cells which fulfils the criteria of EMT in terms of vimentin up-regulation and E-cadherin down-regulation on protein level as well as cell scattering. Furthermore, cells displayed a strongly enhanced invasiveness and adhesion to type I-IV collagens. Phenotype transition is accompanied by an enhanced expression of laminin-332, especially of its γ2 chain. We further analyse the expression of extracellular matrix related genes by RT-PCR profiling. With respect to strongly enhanced proteins, data confirm the EMT phenotype of co-stimulated OSCC cells and expression of laminin-332. Furthermore, alpha catenin, collagen type 16, the integrin α7 and β1 chains, and MMP11 are suggested as candidates with potential role in EMT in OSCC. In summary we are able to show that EMT in OSCC is mediated by multiple growth factors and is accompanied by laminin γ2 chain up-regulation evidencing the existence of an intermediate Vim(+) /Ln332(+) EMT phenotype as seen in situ.
Collapse
Affiliation(s)
- Petra Richter
- Institute of Pathology, University Hospital Jena, Jena, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
243
|
Buckley ST, Davies AM, Ehrhardt C. Atomic force microscopy and high-content analysis: two innovative technologies for dissecting the relationship between epithelial-mesenchymal transition-related morphological and structural alterations and cell mechanical properties. Methods Mol Biol 2011; 784:197-208. [PMID: 21898222 DOI: 10.1007/978-1-61779-289-2_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Epithelial-mesenchymal transition (EMT) is a complex series of cellular reprogramming events culminating in striking alterations in morphology towards an invasive mesenchymal phenotype. Increasingly, evidence suggests that EMT exerts a pivotal role in pathophysiological situations including fibrosis and cancer. Core to these dynamical changes in cellular polarity and plasticity is discrete modifications in cytoskeletal structure. In particular, newly established actin-stress fibres supplant a preceding system of highly organised cortical actin. Although cumulative studies have contributed to elucidation of the detailed signalling pathways that underpin this elaborate molecular process, there remains a deficiency regarding its precise contribution to cellular biomechanics. The advent of atomic force microscopy (AFM) and high-content analysis (HCA) provides two innovative technologies for dissecting the relationship between EMT-related morphological and structural alterations and cell mechanical properties. AFM permits acquisition of high resolution topographical images and detailed analysis of cellular viscoelasticity while HCA facilitates a comprehensive and perspicacious assessment of morphological changes. In combination, they offer the possibility of novel insights into the dynamic traits of transitioning cells. Herein, a detailed protocol describing AFM and HCA techniques for evaluation of transforming growth factor-β1-induced EMT of alveolar epithelial cells is provided.
Collapse
Affiliation(s)
- Stephen T Buckley
- School of Pharmacy & Pharmaceutical Sciences, Trinity College Dublin, Ireland.
| | | | | |
Collapse
|
244
|
Abstract
The activity of a variety of extracellular signaling factors is tightly regulated by proteins containing A Disintegrin And a Metalloprotease domain (ADAM) metalloproteases through limited proteolysis. Thus, the identification of ADAM substrates may unveil novel components and mechanisms of cell signaling pathways. We report the identification of the transmembrane protein vasorin (VASN), a transforming growth factor-β (TGFβ) trap, as a substrate of ADAM17. The metalloprotease efficiently generates a soluble fragment encompassing the extracellular domain of VASN. Despite the importance of TGFβ in normal development and tumor progression, the regulation of VASN is completely unknown. Here, we show that only the soluble form of VASN inhibits TGFβ and that the secretion of VASN is tightly controlled by ADAM17. Hence, inhibition of ADAM17 leads to the upregulation of TGFβ signaling. Adding a new level of complexity to the function of ADAM17, we finally show that, through the cleavage of VASN, the metalloprotease controls TGFβ-mediated epithelial-to-mesenchymal transition.
Collapse
|
245
|
Jahn JE, Best DH, Coleman WB. Exogenous expression of synaptotagmin XIII suppresses the neoplastic phenotype of a rat liver tumor cell line through molecular pathways related to mesenchymal to epithelial transition. Exp Mol Pathol 2010; 89:209-16. [DOI: 10.1016/j.yexmp.2010.09.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Accepted: 09/03/2010] [Indexed: 10/19/2022]
|
246
|
Di Meglio F, Castaldo C, Nurzynska D, Romano V, Miraglia R, Bancone C, Langella G, Vosa C, Montagnani S. Epithelial-mesenchymal transition of epicardial mesothelium is a source of cardiac CD117-positive stem cells in adult human heart. J Mol Cell Cardiol 2010; 49:719-727. [PMID: 20566360 DOI: 10.1016/j.yjmcc.2010.05.013] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Revised: 05/17/2010] [Accepted: 05/26/2010] [Indexed: 12/21/2022]
Abstract
Epithelial-mesenchymal transition is implicated in the remodelling of tissues during development and in the adult life. In the heart, it gives origin to progenitors of fibroblasts, coronary endothelium, smooth muscle cells, and cardiomyocytes. Moreover, epicardially-derived cells determine myocardial wall thickness and Purkinje fibre network. Recently, the presence of numerous cardiac stem cells in the subepicardium of the adult human heart has been described and the hypothesis that epicardially-derived cells can contribute to the population of cardiac stem cells in the adult heart has been advanced. In an effort to test this hypothesis and establish a possible link between epicardium, epicardially-derived cells and cardiac stem cells in the adult human heart we have examined epicardial mesothelial cells in the normal and pathological adult human heart with ischemic cardiomyopathy in vivo and we have induced and documented their epithelial-mesenchymal transition in vitro. Noticeably, epicardial cells were missing from the surface of pathological hearts and the cells with the expression of epithelial and mesenchymal markers populated thick subepicardial space. When the fragments of epicardium from the normal hearts were cultured on the specific substrate formed by extracellular matrix derived from cardiac fibroblasts, we obtained the outgrowth of the epithelial sheet with the mRNA and protein expression characteristic of epicardium. TGFβ induced cellular and molecular changes typical of epithelial-mesenchymal transition. Moreover, the epicardially-derived cells expressed CD117 antigen. Thus, this study provides evidence that cardiac stem cells can originate from epithelial-mesenchymal transition of the epicardial cells in the adult human heart.
Collapse
Affiliation(s)
- Franca Di Meglio
- Department of Biomorphological and Functional Sciences, University of Naples Federico II, Naples, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
247
|
Gil D, Ciołczyk-Wierzbicka D, Dulińska-Litewka J, Zwawa K, McCubrey JA, Laidler P. The mechanism of contribution of integrin linked kinase (ILK) to epithelial-mesenchymal transition (EMT). ACTA ACUST UNITED AC 2010; 51:195-207. [PMID: 21035499 DOI: 10.1016/j.advenzreg.2010.09.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 09/05/2010] [Indexed: 12/22/2022]
Abstract
Integrin linked kinase (ILK) is ubiquitously expressed serine/threonine protein kinase, a binding partner of β1 and β3 integrin subunit as a cytoplasmic effector of integrin receptors that functionally links them to the actin cytoskeleton.We postulate that ILK is important enzyme involved in epithelial-mesenchymal transition (EMT) a critical event in the process of cancer progression. Commonly used EMT molecular markers include among others increased expression of N-cadherin and vimentin, nuclear localization of β-catenin, and the decrease of E-cadherin synthesis. In this study we were able to show that N-cadherin expression in melanoma cells is dependent on ILK signaling and the translocation of β-catenin to the nucleus. Silencing of ILK expression by siRNA significantly inhibited the stabilization and subsequent nuclear translocation of β-catenin and the expression of N-cadherin, a crucial molecule in the EMT, which facilitates association with fibroblast and endothelial cells during invasion of various cancers. The results allow to cautiously speculate on the important role of ILK in the cross-talk between integrins and cadherins accompanying EMT in melanoma.
Collapse
Affiliation(s)
- Dorota Gil
- Chair of Medical Biochemistry Jagiellonian University Medical College, Kraków, Poland.
| | | | | | | | | | | |
Collapse
|
248
|
Abstract
The zinc finger transcription factor Slug (Snai2) serves a wide variety of functions in the epidermis, with roles in skin development, hair growth, wound healing, skin cancer, and sunburn. Slug is expressed in basal keratinocytes and hair follicles where it is important in maintaining epidermal homeostasis. Slug also helps coordinate the skin response to exogenous stimuli. Slug is rapidly induced by a variety of growth factors and injurious agents and Slug controls, directly or indirectly, a variety of keratinocyte responses, including changes in differentiation, adhesion, motility, and production of inflammatory mediators. Slug thus modulates the interactions of the keratinocyte with its environment and with surrounding cells. The function of Slug in the epidermis appears to be distinct from that of the closely related Snail transcription factor.
Collapse
Affiliation(s)
- Stephanie H Shirley
- Department of Carcinogenesis, University of Texas MD Anderson Cancer Center, Science Park Research Division, Smithville, Texas 78957, USA
| | | | | | | |
Collapse
|
249
|
Ørom UA, Derrien T, Beringer M, Gumireddy K, Gardini A, Bussotti G, Lai F, Zytnicki M, Notredame C, Huang Q, Guigo R, Shiekhattar R. Long noncoding RNAs with enhancer-like function in human cells. Cell 2010; 143:46-58. [PMID: 20887892 PMCID: PMC4108080 DOI: 10.1016/j.cell.2010.09.001] [Citation(s) in RCA: 1445] [Impact Index Per Article: 96.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Revised: 07/01/2010] [Accepted: 08/13/2010] [Indexed: 12/21/2022]
Abstract
While the long noncoding RNAs (ncRNAs) constitute a large portion of the mammalian transcriptome, their biological functions has remained elusive. A few long ncRNAs that have been studied in any detail silence gene expression in processes such as X-inactivation and imprinting. We used a GENCODE annotation of the human genome to characterize over a thousand long ncRNAs that are expressed in multiple cell lines. Unexpectedly, we found an enhancer-like function for a set of these long ncRNAs in human cell lines. Depletion of a number of ncRNAs led to decreased expression of their neighboring protein-coding genes, including the master regulator of hematopoiesis, SCL (also called TAL1), Snai1 and Snai2. Using heterologous transcription assays we demonstrated a requirement for the ncRNAs in activation of gene expression. These results reveal an unanticipated role for a class of long ncRNAs in activation of critical regulators of development and differentiation.
Collapse
Affiliation(s)
| | - Thomas Derrien
- Centre for Genomic Regulation (CRG), UPF, Barcelona, Spain
| | - Malte Beringer
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104
| | | | | | | | - Fan Lai
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104
| | | | | | - Qihong Huang
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104
| | - Roderic Guigo
- Centre for Genomic Regulation (CRG), UPF, Barcelona, Spain
| | - Ramin Shiekhattar
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104
- Centre for Genomic Regulation (CRG), UPF, Barcelona, Spain
| |
Collapse
|
250
|
Abstract
The epithelial-mesenchymal transition (EMT) describes a rapid and often reversible modulation of phenotype by epithelial cells. EMT was originally defined in the context of developmental stages, including heart morphogenesis, mesoderm and neural crest formation. Epithelial cells loosen cell-cell adhesion structures throughout EMT. They modulate their polarity, cytoskeleton organization and typically express vimentin filaments and downregulate cytokeratins. They become isolated, mobile and resistant to anoikis. The EMT at least superficially resembles the evolution from normal to transformed cell phenotype during carcinoma progression. The relevance of the concept of EMT in this context was indicated by in vitro models using transformed epithelial cells. Transduction pathways typical of embryogenic EMT in vivo were also found to be activated during cancer progression. More recently, it has been found that such pathways indicate an increased plasticity linked to cellular stemness and ability to generate tumors. However, in the absence of direct evidence, a number of oncologists and pathologists remain skeptical about applying the EMT concept to human tumor progression. Typically in the cancer field, EMT concept appears to be fully relevant in some situations, but the concept has to be adjusted in other situations to reflect tumor cell renewal and plasticity during carcinoma progression and metastasis.
Collapse
Affiliation(s)
- P Savagner
- IRCM U896 INSERM, CRLC Val d'Aurelle-Paul Lamarque, Montpellier, France.
| |
Collapse
|