151
|
He Z, Deng R, Huang X, Ni Y, Yang X, Wang Z, Hu Q. Lipopolysaccharide enhances OSCC migration by promoting epithelial-mesenchymal transition. J Oral Pathol Med 2014; 44:685-92. [PMID: 25367215 DOI: 10.1111/jop.12285] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2014] [Indexed: 01/19/2023]
Abstract
BACKGROUND This study was performed to examine whether lipopolysaccharide can influence cell migration and epithelial-mesenchymal transition of oral squamous cell carcinoma. METHODS Three oral squamous cell carcinoma cell lines (HSC3, CAL27, and SCC4) were obtained for the study. TLR4 expression in three cell lines was analyzed by Q-PCR and Western blot. After cells treated with LPS, cell migration was analyzed by wound-healing and chemotaxis cell migration assay. Changes of E-cadherin and vimentin expression were tested by Western blot and immunofluorescence staining. To examine NF-κB activation, NF-κB nuclear translocation was investigated. RESULTS TLR4 was expressed in all three cell lines and was highest in HSC3 while lowest in SCC4. TLR4 ligand lipopolysaccharide accelerated wound healing and enhanced cell migration. Also, it stimulated epithelial-mesenchymal transition demonstrated by decreased E-cadherin and increased vimentin expression. Lipopolysaccharide also provoked NF-κB nuclear translocation. Either TLR4 or NF-κB blocking reverted these effects. CONCLUSIONS Lipopolysaccharide can induce TLR4-mediated epithelial-mesenchymal transition and cell migration in oral squamous cell carcinoma. These responses could further affect tumor progressing by inducing tumor cell metastasis.
Collapse
Affiliation(s)
- Zhifeng He
- Central Laboratory of Stomatology, Institute and Hospital of Stomatology, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Runzhi Deng
- Department of Oral and Maxillofacial Surgery, Institute and Hospital of Stomatology, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Xiaofeng Huang
- Central Laboratory of Stomatology, Institute and Hospital of Stomatology, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Yanhong Ni
- Central Laboratory of Stomatology, Institute and Hospital of Stomatology, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Xudong Yang
- Department of Oral and Maxillofacial Surgery, Institute and Hospital of Stomatology, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Zhiyong Wang
- Department of Oral and Maxillofacial Surgery, Institute and Hospital of Stomatology, Nanjing University Medical School, Nanjing University, Nanjing, China
| | - Qingang Hu
- Department of Oral and Maxillofacial Surgery, Institute and Hospital of Stomatology, Nanjing University Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
152
|
McLane JS, Rivet CJ, Gilbert RJ, Ligon LA. A biomaterial model of tumor stromal microenvironment promotes mesenchymal morphology but not epithelial to mesenchymal transition in epithelial cells. Acta Biomater 2014; 10:4811-4821. [PMID: 25058401 DOI: 10.1016/j.actbio.2014.07.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 07/11/2014] [Accepted: 07/14/2014] [Indexed: 12/13/2022]
Abstract
The stromal tissue surrounding most carcinomas is comprised of an extracellular matrix densely packed with collagen-I fibers, which are often highly aligned in metastatic disease. Here we developed an in vitro model to test the effect of an aligned fibrous environment on cancer cell morphology and behavior, independent of collagen ligand presentation. We grew cells on a biomimetic surface of aligned electrospun poly-l-lactic acid (PLLA) fibers and then examined the effect of this environment on growth rate, morphology, cytoskeletal organization, biochemical and genetic markers of epithelial to mesenchymal transition (EMT), cell surface adhesion, and cell migration. We grew a phenotypically normal breast epithelial cell line (MCF10A) and an invasive breast cancer cell line (MDA-MB-231) on three different substrates: typical flat culture surface (glass or plastic), flat PLLA (glass coated with PLLA) or electrospun PLLA fibers. Cells of both types adopted a more mesenchymal morphology when grown on PLLA fibers, and this effect was exaggerated in the more metastatic-like MDA-MB-231 cells. However, neither cell type underwent the changes in gene expression indicative of EMT despite the changes in cell shape, nor did they exhibit the decreased adhesive strength or increased migration typical of metastatic cells. These results suggest that changes in cell morphology alone do not promote a more mesenchymal phenotype and consequently that the aligned fibrous environment surrounding epithelial cancers may not promote EMT solely through topographical cues.
Collapse
Affiliation(s)
- Joshua S McLane
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180-3590, USA; Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY 12180-3590, USA
| | - Christopher J Rivet
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180-3590, USA; Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180-3590, USA
| | - Ryan J Gilbert
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180-3590, USA; Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180-3590, USA
| | - Lee A Ligon
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180-3590, USA; Department of Biological Sciences, Rensselaer Polytechnic Institute, Troy, NY 12180-3590, USA.
| |
Collapse
|
153
|
Neelakantan D, Drasin DJ, Ford HL. Intratumoral heterogeneity: Clonal cooperation in epithelial-to-mesenchymal transition and metastasis. Cell Adh Migr 2014; 9:265-76. [PMID: 25482627 DOI: 10.4161/19336918.2014.972761] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Although phenotypic intratumoral heterogeneity was first described many decades ago, the advent of next-generation sequencing has provided conclusive evidence that in addition to phenotypic diversity, significant genotypic diversity exists within tumors. Tumor heterogeneity likely arises both from clonal expansions, as well as from differentiation hierarchies existent in the tumor, such as that established by cancer stem cells (CSCs) and non-CSCs. These differentiation hierarchies may arise due to genetic mutations, epigenetic alterations, or microenvironmental influences. An additional differentiation hierarchy within epithelial tumors may arise when only a few tumor cells trans-differentiate into mesenchymal-like cells, a process known as epithelial-to-mesenchymal transition (EMT). Again, this process can be influenced by both genetic and non-genetic factors. In this review we discuss the evidence for clonal interaction and cooperation for tumor maintenance and progression, particularly with respect to EMT, and further address the far-reaching effects that tumor heterogeneity may have on cancer therapy.
Collapse
Key Words
- CLL, chronic lymphoblastic leukemia
- CSC, cancer stem cell
- EMP, epithelial-mesenchymal plasticity
- EMT, epithelial-to-mesenchymal transition
- GFP, green fluorescent protein.
- MET, mesenchymal-to-epithelial transition
- MMTV, mouse mammary tumor virus
- NGS, next generation sequencing
- OxR, oxaliplatin resistant
- SCLC, small cell lung cancer
- TGF-β, transforming growth factor-β
- cancer stem cells/CSCs
- clonal evolution
- epithelial-mesenchymal transition (EMT)
- hPDGF human platelet-derived growth factor
- intratumoral heterogeneity
- metastasis
- miRNA, microRNA
- non-cell autonomous
- tumor microenvironment
Collapse
Affiliation(s)
- Deepika Neelakantan
- a Department of Pharmacology ; University of Colorado; School of Medicine ; Aurora, CO USA
| | | | | |
Collapse
|
154
|
Increased expression of CCN2, epithelial membrane antigen, and fibroblast activation protein in hepatocellular carcinoma with fibrous stroma showing aggressive behavior. PLoS One 2014; 9:e105094. [PMID: 25126747 PMCID: PMC4134271 DOI: 10.1371/journal.pone.0105094] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 07/21/2014] [Indexed: 02/06/2023] Open
Abstract
Tumor behavior is affected by the tumor microenvironment, composed of cancer-associated fibroblasts (CAFs). Meanwhile, hepatocellular carcinomas (HCC) with fibrous stroma reportedly exhibit aggressive behavior suggestive of tumor-stroma interaction. However, evidence of the crosstalk remains unclear. In this study, CCN2, epithelial membrane antigen (EMA), fibroblast activation protein (FAP), and keratin 19 (K19) expression was studied in 314 HCCs (cohort 1), 42 scirrhous HCCs (cohort 2), and 36 chronic hepatitis/cirrhosis specimens by immunohistochemistry. Clinicopathological parameters were analyzed according to the expressions of these markers. In tumor epithelial cells from cohort 1, CCN2 and EMA were expressed in 15.3% and 17.2%, respectively, and their expressions were more frequent in HCCs with fibrous stroma (≥5% of tumor area) than those without (P<0.05 for all); CCN2 expression was well correlated with K19 and EMA expression. In tumor stromal cells, FAP expression was found in 6.7%. In cohort 2, CCN2, EMA, and FAP expression was noted in 40.5%, 40.5%, and 66.7%, respectively, which was more frequent than that in cohort 1 (P<0.05 for all). Additionally, EMA expression was associated with the expression of K19, CCN2, and FAP (P<0.05 for all); EMA expressing tumor epithelial cells showed a topographic closeness to FAP-expressing CAFs. Analysis of disease-free survival revealed CCN2 expression to be a worse prognostic factor in both cohort 1 (P = 0.005) and cohort 2 (P = 0.023), as well as EMA as a worse prognostic factor in cohort 2 (P = 0.048). In conclusion, expression of CCN2, EMA, and FAP may be involved in the activation of CAFs in HCC, giving rise to aggressive behavior. Significant correlation between EMA-expressing tumor cells and FAP-expressing CAFs and their topographic closeness suggests possible cross-talk between tumor epithelial cells and stromal cells in the tumor microenvironment of HCC.
Collapse
|
155
|
Duangkumpha K, Techasen A, Loilome W, Namwat N, Thanan R, Khuntikeo N, Yongvanit P. BMP-7 blocks the effects of TGF-β-induced EMT in cholangiocarcinoma. Tumour Biol 2014; 35:9667-76. [PMID: 24969562 DOI: 10.1007/s13277-014-2246-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 06/18/2014] [Indexed: 01/12/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) is characterized by the loss of epithelial markers and the gain of mesenchymal markers. EMT is believed to be a major mechanism supporting cancer cell metastasis. The activation of EMT can be induced by various types of inflammatory cytokines including transforming growth factor β (TGF-β) whereas bone morphogenetic protein-7 (BMP-7) can inhibit this process. In this study, the up-regulation of Twist transcription factor and N-cadherin, mesenchymal marker in CCA tissues, has been demonstrated and it has been found that the high expression of Twist was significantly associated with poor prognosis of CCA patients (P = 0.010). Moreover, CCA samples showing Twist nuclear expression were significantly correlated with the up-regulation of N-cadherin (P = 0.024). These results also showed that the inflammatory mediator TGF-β induces CCA cell migration, one of the metastatic processes possibly via stimulation of Twist, N-cadherin and vimentin expression. Additionally, it has been shown that BMP-7 inhibits TGF-β-induced CCA cell migration, through inhibition of TGF-β-mediated Twist and N-cadherin expressions. These data reinforce the rationale to use BMP-7 as an EMT inhibitor to suppress the progression of CCA and might be a therapeutic approach to improve efficiency for CCA treatment.
Collapse
Affiliation(s)
- Kassaporn Duangkumpha
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | | | | | | | | | | | | |
Collapse
|
156
|
Fazilaty H, Mehdipour P. Genetics of breast cancer bone metastasis: a sequential multistep pattern. Clin Exp Metastasis 2014; 31:595-612. [PMID: 24493024 DOI: 10.1007/s10585-014-9642-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 01/26/2014] [Indexed: 02/05/2023]
|
157
|
Schiavone M, Rampazzo E, Casari A, Battilana G, Persano L, Moro E, Liu S, Leach SD, Tiso N, Argenton F. Zebrafish reporter lines reveal in vivo signaling pathway activities involved in pancreatic cancer. Dis Model Mech 2014; 7:883-94. [PMID: 24878567 PMCID: PMC4073277 DOI: 10.1242/dmm.014969] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pancreatic adenocarcinoma, one of the worst malignancies of the exocrine pancreas, is a solid tumor with increasing incidence and mortality in industrialized countries. This condition is usually driven by oncogenic KRAS point mutations and evolves into a highly aggressive metastatic carcinoma due to secondary gene mutations and unbalanced expression of genes involved in the specific signaling pathways. To examine in vivo the effects of KRASG12D during pancreatic cancer progression and time correlation with cancer signaling pathway activities, we have generated a zebrafish model of pancreatic adenocarcinoma in which eGFP-KRASG12D expression was specifically driven to the pancreatic tissue by using the GAL4/UAS conditional expression system. Outcrossing the inducible oncogenic KRASG12D line with transgenic zebrafish reporters, harboring specific signaling responsive elements of transcriptional effectors, we were able to follow TGFβ, Notch, Bmp and Shh activities during tumor development. Zebrafish transgenic lines expressing eGFP-KRASG12D showed normal exocrine pancreas development until 3 weeks post fertilization (wpf). From 4 to 24 wpf we observed several degrees of acinar lesions, characterized by an increase in mesenchymal cells and mixed acinar/ductal features, followed by progressive bowel and liver infiltrations and, finally, highly aggressive carcinoma. Moreover, live imaging analysis of the exocrine pancreatic tissue revealed an increasing number of KRAS-positive cells and progressive activation of TGFβ and Notch pathways. Increase in TGFβ, following KRASG12D activation, was confirmed in a concomitant model of medulloblastoma (MDB). Notch and Shh signaling activities during tumor onset were different between MDB and pancreatic adenocarcinoma, indicating a tissue-specific regulation of cell signaling pathways. Moreover, our results show that a living model of pancreatic adenocarcinoma joined with cell signaling reporters is a suitable tool for describing in vivo the signaling cascades and molecular mechanisms involved in tumor development and a potential platform to screen for novel oncostatic drugs.
Collapse
Affiliation(s)
- Marco Schiavone
- Department of Biology, University of Padua, 35131 Padua, Italy
| | - Elena Rampazzo
- Department of Molecular Medicine, University of Padua, 35131 Padua, Italy
| | | | - Giusy Battilana
- Department of Molecular Medicine, University of Padua, 35131 Padua, Italy
| | - Luca Persano
- Department of Woman and Child Health, University of Padua, 35131 Padua, Italy
| | - Enrico Moro
- Department of Molecular Medicine, University of Padua, 35131 Padua, Italy
| | - Shu Liu
- Department of Surgery and The McKusick-Nathans Institute of Genetic Medicine Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Steve D Leach
- Department of Surgery and The McKusick-Nathans Institute of Genetic Medicine Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Natascia Tiso
- Department of Biology, University of Padua, 35131 Padua, Italy
| | | |
Collapse
|
158
|
Wutka A, Palagani V, Barat S, Chen X, El Khatib M, Götze J, Belahmer H, Zender S, Bozko P, Malek NP, Plentz RR. Capsaicin treatment attenuates cholangiocarcinoma carcinogenesis. PLoS One 2014; 9:e95605. [PMID: 24748170 PMCID: PMC3991659 DOI: 10.1371/journal.pone.0095605] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 03/28/2014] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED Capsaicin, the most abundant pungent molecule produced by pepper plants, represents an important ingredient in spicy foods consumed throughout the world. Studies have shown that capsaicin can relieve inflammation and has anti-proliferative effects on various human malignancies. Cholangiocarcinoma (CC) is a cancer disease with rising incidence. The prognosis remains dismal with little advance in treatment. The aim of the present study is to explore the anti-tumor activity of capsaicin in cultured human CC cell lines. Capsaicin effectively impaired cell proliferation, migration, invasion, epithelial to mesenchymal transition and growth of softagar colonies. Further, we show that capsaicin treatment of CC cells regulates the Hedgehog signaling pathway. CONCLUSION Our results provide a basis for capsaicin to improve the prognosis of CCs in vivo and present new insights into the effectiveness and mode of action of capsaicin.
Collapse
Affiliation(s)
- Annika Wutka
- Department of Internal Medicine I, Medical University Hospital, Tuebingen, Germany
| | - Vindhya Palagani
- Department of Internal Medicine I, Medical University Hospital, Tuebingen, Germany
| | - Samarpita Barat
- Department of Internal Medicine I, Medical University Hospital, Tuebingen, Germany
| | - Xi Chen
- Department of Internal Medicine I, Medical University Hospital, Tuebingen, Germany
| | - Mona El Khatib
- Department of Internal Medicine I, Medical University Hospital, Tuebingen, Germany
| | - Julian Götze
- Department of Internal Medicine I, Medical University Hospital, Tuebingen, Germany
| | - Hanane Belahmer
- Department of Internal Medicine I, Medical University Hospital, Tuebingen, Germany
| | - Steffen Zender
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover, Germany
| | - Przemyslaw Bozko
- Department of Internal Medicine I, Medical University Hospital, Tuebingen, Germany
| | - Nisar P. Malek
- Department of Internal Medicine I, Medical University Hospital, Tuebingen, Germany
| | - Ruben R. Plentz
- Department of Internal Medicine I, Medical University Hospital, Tuebingen, Germany
| |
Collapse
|
159
|
Abstract
The transdifferentiation of epithelial cells into motile mesenchymal cells, a process known as epithelial-mesenchymal transition (EMT), is integral in development, wound healing and stem cell behaviour, and contributes pathologically to fibrosis and cancer progression. This switch in cell differentiation and behaviour is mediated by key transcription factors, including SNAIL, zinc-finger E-box-binding (ZEB) and basic helix-loop-helix transcription factors, the functions of which are finely regulated at the transcriptional, translational and post-translational levels. The reprogramming of gene expression during EMT, as well as non-transcriptional changes, are initiated and controlled by signalling pathways that respond to extracellular cues. Among these, transforming growth factor-β (TGFβ) family signalling has a predominant role; however, the convergence of signalling pathways is essential for EMT.
Collapse
|
160
|
Yan H, Sun Y. Evaluation of the mechanism of epithelial-mesenchymal transition in human ovarian cancer stem cells transfected with a WW domain-containing oxidoreductase gene. Oncol Lett 2014; 8:426-430. [PMID: 24959289 PMCID: PMC4063641 DOI: 10.3892/ol.2014.2063] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 03/27/2014] [Indexed: 01/06/2023] Open
Abstract
The aim of the present study was to investigate the impact of the WW domain-containing oxidoreductase (WWOX) gene on the mechanisms underlying epithelial-mesenchymal transition (EMT) in human ovarian cancer stem cells. Western blot analysis was performed to detect the differences in the expression of the EMT markers, E-cadherin, β-catenin, N-cadherin, vimentin and fibronectin, between human ovarian cancer stem cells and the human epithelial ovarian carcinoma cell line, HO-8910. A pcDNA3.1-WWOX eukaryotic expression vector was subsequently transfected into the ovarian cancer stem cells (recombinant plasmid group) or an empty plasmid (empty plasmid group) and non-transfected ovarian cancer stem cells (blank control group) served as the controls. Following the transfection of the WWOX gene, methyl thiazolyl tetrazolium cell viability and Transwell® invasion assays, and western blot analysis were performed to detect changes in the proliferative capability and invasive capacity of ovarian cancer stem cells, as well as the expression of EMT markers and regulatory factors, Elf5 and Snail. The expression levels of E-cadherin and β-catenin in the ovarian cancer stem cells were identified to be significantly lower than those in the HO-8910 cells, whereas the expression levels of N-cadherin, vimentin and fibronectin in the ovarian cancer stem cells were found to be significantly higher than those in the HO-8910 cells. At each time point, the cellular proliferative capacity of the recombinant plasmid group was observed to be significantly lower than that of the empty plasmid or blank control groups (P<0.05 vs. the controls). The number of penetrating cells in the recombinant plasmid, empty plasmid and the blank control groups were 105.5±3.1, 199.7±3.4 and 191.4±4.1, respectively (mean ± standard error of the mean; P<0.05 vs. the controls). In addition, the protein expression of E-cadherin, β-catenin and Elf5 in the recombinant plasmid group was found to be significantly higher than that in the other two groups, whereas the protein expression of N-cadherin, vimentin, fibronectin and Snail in the recombinant plasmid group was significantly lower than that in the other two groups. An EMT exists in ovarian cancer stem cells, and the WWOX gene inhibits the cellular proliferation of ovarian cancer stem cells and reduces their invasive capability. Therefore, the WWOX gene may reverse the EMT in ovarian cancer stem cells by regulating the expression of the EMT regulatory factors, Elf5 and Snail.
Collapse
Affiliation(s)
- Hongchao Yan
- Department of Oncology, Shandong University School Of Medicine, Jinan, Shandong 250012, P.R. China
| | - Yuping Sun
- Department of Medical Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, P.R. China
| |
Collapse
|
161
|
Pagoulatos D, Pharmakakis N, Lakoumentas J, Assimakopoulou M. Ηypoxia-inducible factor-1α, von Hippel-Lindau protein, and heat shock protein expression in ophthalmic pterygium and normal conjunctiva. Mol Vis 2014; 20:441-57. [PMID: 24715760 PMCID: PMC3976686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 03/28/2014] [Indexed: 10/28/2022] Open
Abstract
PURPOSE Εnhanced expression of transcription factor hypoxia inducible factor HIF-1α is known to play a critical role in the modulation of cell metabolism and survival pathways as well as having stem-cell-like properties. Furthermore, accumulated data reveal the existence of cross-regulation between the oxygen-sensing and heat shock pathways contributing to the adaptation of cells under stressful conditions. Pterygium, a stem cell disorder with premalignant features, has been reported to demonstrate hypoxia. The purpose of this study was to investigate the co-expression patterns of transcription factor HIF-1α and von Hippel Lindau protein (pVHL)--which normally acts to keep levels of HIF-1α activity low under normoxic conditions--in pterygium and normal conjunctival human samples. Additionally, expression of HIF-1α compared to the activation of heat shock proteins (Hsp90, Hsp70, and Hsp27) was studied. Emphasis was placed on the detection of HIF-1α and Hsp90, which associates with and stabilizes HIF-1α to promote its transcriptional activity. METHODS Semi-serial paraffin-embedded sections and tissue extracts from pterygium and normal conjunctival samples were studied by immunohistochemistry and western blot analysis, respectively, with the use of specific antibodies. Double labeling immunofluorescence studies on cryostat sections were also included. RESULTS Statistically significant increased expression of HIF-1α and Hsps (Hsp90, Hsp70, and Hsp27) in pterygia compared to normal conjunctiva was demonstrated (p<0.05). In contrast, no significant difference was detected for pVHL expression (p>0.05). Immunohistochemical findings revealed nuclear HIF-1α immunoreactivity in all the epithelial layers of 23/32 (71.8%) pterygium tissues. Furthermore, all epithelial layers of the majority (75%) of pterygium samples showed strong cytoplasmic immunoreactivity for Hsp27 while Hsp27 expression was detected in all pterygia (100%) examined. Hsp27 expression was not observed in the superficial layer of goblet cells. In some samples, focal basal epithelial cells exhibited weak Hsp27 expression or were Hsp27 immunonegative. Ιmmunoreactivity of phopsho-Hsp27 showed the same distribution pattern as Hsp27 did. Epithelium of all pterygia (100%) displayed moderate to strong Hsp90 cytoplasmic immunoreactivity. Furthermore, the majority of pterygia, specifically, 30/32 (93.7%) and 27/32 (84.3%) demonstrated, respectively, Hsp70 and pVHL cytoplasmic immunoreactivity. Hsp90, Hsp70, and pVHL immunoreactivity was mainly detected in basal and suprabasal epithelial layers even though strong immunoreactivity in all epithelial layers was also observed in some pterygia. Stroma vessels were immunopositive for Hsps (Hsp90, Hsp70, and Hsp27) and pVHL. A statistically significant correlation between the expression of HIF-1α and the activation status of Hsps (Hsp90, Hsp70, and Hsp27; p<0.05) was observed whereas HIF-1α expression did not correlate with pVHL expression (p>0.05). Double labeling immunofluorescence studies showed nuclear HIF-1α co-localization with cytoplasmic Hsp90 expression in cells distributed in the entire epithelium of pterygia, in contrast to, normal conjunctiva, which exhibited only a few scattered epithelial cells with cytoplasmic HIF-1α expression and basal epithelial cells with Hsp90 expression. CONCLUSIONS The upregulation of coordinated activation of HIF-1α and Hsps in pterygium may represent an adaptive process for the survival of cells under stressful conditions. The significance of the association of HIF-1α with Hsp90 with respect to the therapeutic approach of pterygium requires further evaluation.
Collapse
Affiliation(s)
- Dionysios Pagoulatos
- Department of Anatomy-Histology and Embryology, School of Medicine, University of Patras, Rion, Greece
| | - Nikolaos Pharmakakis
- Department of Ophthalmology, School of Medicine, University of Patras, Rion, Greece
| | - John Lakoumentas
- Department of Medical Physics, School of Medicine, University of Patras, Rion, Greece
| | - Martha Assimakopoulou
- Department of Anatomy-Histology and Embryology, School of Medicine, University of Patras, Rion, Greece
| |
Collapse
|
162
|
Bouyssou JMC, Manier S, Huynh D, Issa S, Roccaro AM, Ghobrial IM. Regulation of microRNAs in cancer metastasis. Biochim Biophys Acta Rev Cancer 2014; 1845:255-65. [PMID: 24569228 DOI: 10.1016/j.bbcan.2014.02.002] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Revised: 01/15/2014] [Accepted: 02/12/2014] [Indexed: 12/13/2022]
Abstract
Metastasis is a phenomenon of crucial importance in defining prognosis in patients with cancer and is often responsible for cancer-related mortality. It is known that several steps are necessary for clonal cells to disseminate from their primary tumor site and colonize distant tissues, thus originating metastatic lesions. Therefore, investigating the molecular actors regulating this process may provide helpful insights in the development of efficient therapeutic responses. Recent evidences have indicated the role of microRNAs (miRNAs) in modulating the metastatic process in solid tumors. miRNAs are small regulatory non-coding RNAs that bind to specific target mRNAs, leading to translational repression. miRNAs are known to act as negative regulators of gene expression and are involved in the regulation of biological processes, including cell growth, differentiation and apoptosis, both in physiological conditions and during diseases, such as tumors. In the specific field of tumorigenesis, miRNAs play an important role in mediating oncogenesis and favoring tumor progression, as a result of their ability to modulate epithelial-to-mesenchymal transition (EMT) and other series of events facilitating the formation of metastasis. The role of miRNAs in cancer development has been widely studied and has helped elucidate events such as the change in expression of oncogenes, tumor-suppressors and cancer-related proteins. This review focuses on the mechanisms underlying the role of miRNAs as part of the metastatic process.
Collapse
Affiliation(s)
- Juliette M C Bouyssou
- Dana-Farber Cancer Institute, Department of Medical Oncology, Harvard Medical School, 450 Brookline Avenue, HIM 246, Boston, MA 02215, USA; Ecole de Biologie Industrielle, 32 Boulevard du port, 95094 Cergy-Pontoise cedex, France
| | - Salomon Manier
- Dana-Farber Cancer Institute, Department of Medical Oncology, Harvard Medical School, 450 Brookline Avenue, HIM 246, Boston, MA 02215, USA
| | - Daisy Huynh
- Dana-Farber Cancer Institute, Department of Medical Oncology, Harvard Medical School, 450 Brookline Avenue, HIM 246, Boston, MA 02215, USA
| | - Samar Issa
- Ecole de Biologie Industrielle, 32 Boulevard du port, 95094 Cergy-Pontoise cedex, France
| | - Aldo M Roccaro
- Dana-Farber Cancer Institute, Department of Medical Oncology, Harvard Medical School, 450 Brookline Avenue, HIM 246, Boston, MA 02215, USA
| | - Irene M Ghobrial
- Dana-Farber Cancer Institute, Department of Medical Oncology, Harvard Medical School, 450 Brookline Avenue, HIM 246, Boston, MA 02215, USA.
| |
Collapse
|
163
|
Brosseau JP, Lucier JF, Nwilati H, Thibault P, Garneau D, Gendron D, Durand M, Couture S, Lapointe E, Prinos P, Klinck R, Perreault JP, Chabot B, Abou-Elela S. Tumor microenvironment-associated modifications of alternative splicing. RNA (NEW YORK, N.Y.) 2014; 20:189-201. [PMID: 24335142 PMCID: PMC3895271 DOI: 10.1261/rna.042168.113] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Pre-mRNA alternative splicing is modified in cancer, but the origin and specificity of these changes remain unclear. Here, we probed ovarian tumors to identify cancer-associated splicing isoforms and define the mechanism by which splicing is modified in cancer cells. Using high-throughput quantitative PCR, we monitored the expression of splice variants in laser-dissected tissues from ovarian tumors. Surprisingly, changes in alternative splicing were not limited to the tumor tissues but were also found in the tumor microenvironment. Changes in the tumor-associated splicing events were found to be regulated by splicing factors that are differentially expressed in cancer tissues. Overall, ∼20% of the alternative splicing events affected by the down-regulation of the splicing factors QKI and RBFOX2 were altered in the microenvironment of ovarian tumors. Together, our results indicate that the tumor microenvironment undergoes specific changes in alternative splicing orchestrated by a limited number of splicing factors.
Collapse
Affiliation(s)
- Jean-Philippe Brosseau
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
- Département de Biochimie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Jean-François Lucier
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
- Département de Microbiologie et d'Infectiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Hanad Nwilati
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Philippe Thibault
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Daniel Garneau
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Daniel Gendron
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Mathieu Durand
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Sonia Couture
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Elvy Lapointe
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Panagiotis Prinos
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Roscoe Klinck
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
- Département de Microbiologie et d'Infectiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Jean-Pierre Perreault
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
- Département de Biochimie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Benoit Chabot
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
- Département de Microbiologie et d'Infectiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
| | - Sherif Abou-Elela
- Laboratoire de Génomique Fonctionnelle de l'Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
- Département de Microbiologie et d'Infectiologie, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, Québec, Canada J1E 4K8
- Corresponding authorE-mail
| |
Collapse
|
164
|
Yang WN, Ai ZH, Wang J, Xu YL, Teng YC. Correlation between the overexpression of epidermal growth factor receptor and mesenchymal makers in endometrial carcinoma. J Gynecol Oncol 2014; 25:36-42. [PMID: 24459579 PMCID: PMC3893673 DOI: 10.3802/jgo.2014.25.1.36] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Revised: 09/17/2013] [Accepted: 10/02/2013] [Indexed: 01/12/2023] Open
Abstract
Objective The objective of this study was to evaluate the effect of overexpression of epidermal growth factor receptor (EGFR) on the expression of epithelial cell markers (E-cadherin and α-catenin) and mesenchymal cell markers (N-cadherin and vimentin) in endometrial carcinoma. Methods The expression of all 4 markers was evaluated in EGFR overexpressing Ishikawa cells, control Ishikawa cells, and KLE cells using reverse transcription polymerase chain reaction (RT-PCR) and Western blotting. The expression of these 4 markers was also determined in cancerous tissues of patients with endometrial carcinoma using immunohistochemical staining. Results Ishikawa cells transfected with EGFR showed decreased expression of E-cadherin and α-catenin and increased expression of N-cadherin and vimentin compared with control Ishikawa cells (p<0.01 for all). The expression of N-cadherin and vimentin was higher and the expression of E-cadherin and α-catenin was lower in stage II-III than stage I and in grade II-III than grade I endometrial carcinoma tissue (p<0.01 for all). Conclusion Decreased expression of epithelial markers (E-cadherin and α-catenin) and increased expression of mesenchymal markers (N-cadherin and vimentin) were observed in human endometrial carcinoma tissue. These findings correlate with high EGFR expression in cultured endometrial carcinoma cells.
Collapse
Affiliation(s)
- Wei-Ning Yang
- Department of Obstetrics and Gynecology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Zhi-Hong Ai
- Department of Obstetrics and Gynecology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Juan Wang
- Department of Obstetrics and Gynecology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yan-Li Xu
- Department of Obstetrics and Gynecology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yin-Cheng Teng
- Department of Obstetrics and Gynecology, Shanghai Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
165
|
Maryáš J, Faktor J, Dvořáková M, Struhárová I, Grell P, Bouchal P. Proteomics in investigation of cancer metastasis: Functional and clinical consequences and methodological challenges. Proteomics 2014; 14:426-40. [DOI: 10.1002/pmic.201300264] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2013] [Revised: 09/16/2013] [Accepted: 10/04/2013] [Indexed: 01/05/2023]
Affiliation(s)
- Josef Maryáš
- Department of Biochemistry; Faculty of Science; Masaryk University; Brno Czech Republic
| | - Jakub Faktor
- Department of Biochemistry; Faculty of Science; Masaryk University; Brno Czech Republic
- Regional Centre for Applied Molecular Oncology; Masaryk Memorial Cancer Institute; Brno Czech Republic
| | - Monika Dvořáková
- Department of Biochemistry; Faculty of Science; Masaryk University; Brno Czech Republic
- Regional Centre for Applied Molecular Oncology; Masaryk Memorial Cancer Institute; Brno Czech Republic
| | - Iva Struhárová
- Department of Biochemistry; Faculty of Science; Masaryk University; Brno Czech Republic
- Regional Centre for Applied Molecular Oncology; Masaryk Memorial Cancer Institute; Brno Czech Republic
| | - Peter Grell
- Department of Comprehensive Cancer Care; Masaryk Memorial Cancer Institute; Brno Czech Republic
| | - Pavel Bouchal
- Department of Biochemistry; Faculty of Science; Masaryk University; Brno Czech Republic
- Regional Centre for Applied Molecular Oncology; Masaryk Memorial Cancer Institute; Brno Czech Republic
| |
Collapse
|
166
|
Bao B, Azmi AS, Li Y, Ahmad A, Ali S, Banerjee S, Kong D, Sarkar FH. Targeting CSCs in tumor microenvironment: the potential role of ROS-associated miRNAs in tumor aggressiveness. Curr Stem Cell Res Ther 2014; 9:22-35. [PMID: 23957937 PMCID: PMC4493722 DOI: 10.2174/1574888x113089990053] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 08/01/2013] [Accepted: 08/06/2013] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) have been widely considered as critical cellular signaling molecules involving in various biological processes such as cell growth, differentiation, proliferation, apoptosis, and angiogenesis. The homeostasis of ROS is critical to maintain normal biological processes. Increased production of ROS, namely oxidative stress, due to either endogenous or exogenous sources causes irreversible damage of bio-molecules such as DNA, proteins, lipids, and sugars, leading to genomic instability, genetic mutation, and altered gene expression, eventually contributing to tumorigenesis. A great amount of experimental studies in vitro and in vivo have produced solid evidence supporting that oxidative stress is strongly associated with increased tumor cell growth, treatment resistance, and metastasis, and all of which contribute to tumor aggressiveness. More recently, the data have indicated that altered production of ROS is also associated with cancer stem cells (CSCs), epithelial-to-mesenchymal transition (EMT), and hypoxia, the most common features or phenomena in tumorigenesis and tumor progression. However, the exact mechanism by which ROS is involved in the regulation of CSC and EMT characteristics as well as hypoxia- and, especially, HIF-mediated pathways is not well known. Emerging evidence suggests the role of miRNAs in tumorigenesis and progression of human tumors. Recently, the data have indicated that altered productions of ROS are associated with deregulated expression of miRNAs, suggesting their potential roles in the regulation of ROS production. Therefore, targeting ROS mediated through the deregulation of miRNAs by novel approaches or by naturally occurring anti-oxidant agents such as genistein could provide a new therapeutic approach for the prevention and/or treatment of human malignancies. In this article, we will discuss the potential role of miRNAs in the regulation of ROS production during tumorigenesis. Finally, we will discuss the role of genistein, as a potent anti-tumor agent in the regulation of ROS production during tumorigenesis and tumor development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Fazlul H Sarkar
- Departments of Pathology and Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, 740 HWCRC, 4100 John R Street, Detroit, MI 48201, USA.
| |
Collapse
|
167
|
MUC1 drives epithelial-mesenchymal transition in renal carcinoma through Wnt/β-catenin pathway and interaction with SNAIL promoter. Cancer Lett 2013; 346:225-36. [PMID: 24384091 DOI: 10.1016/j.canlet.2013.12.029] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 12/18/2013] [Accepted: 12/20/2013] [Indexed: 01/11/2023]
Abstract
MUC1 is overexpressed in human carcinomas. The transcription factor SNAIL can activate epithelial-mesenchymal transition (EMT) in cancer cells. In this study, in renal carcinoma, we demonstrate that (i) MUC1 and SNAIL were overexpressed in human sarcomatoid carcinomas, (ii) SNAIL increased indirectly MUC1 expression, (iii) MUC1 overexpression induced EMT, (iv) MUC1 C-terminal domain (MUC1-C) and β-catenin increased SNAIL transcriptional activity by interaction with its promoter and (v) blocking MUC1-C nuclear localization decreased Wnt/β-catenin signaling pathway activation and SNAIL expression. Altogether, our findings demonstrate that MUC1 is an actor in EMT and appears as a new therapeutic target.
Collapse
|
168
|
Guan Z, Ding C, Du Y, Zhang K, Zhu JN, Zhang T, He D, Xu S, Wang X, Fan J. HAF drives the switch of HIF-1α to HIF-2α by activating the NF-κB pathway, leading to malignant behavior of T24 bladder cancer cells. Int J Oncol 2013; 44:393-402. [PMID: 24316875 PMCID: PMC3898811 DOI: 10.3892/ijo.2013.2210] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 11/19/2013] [Indexed: 12/14/2022] Open
Abstract
Hypoxia is a characteristic feature of solid tumors, leading to malignant behavior. During this process, HIF family members (HIFs) and the NF-κB pathway are activated. In addition, the hypoxia-associated factor (HAF) is reported to participate in the regulation of HIFs. However, the precise relationship among HIFs, HAF and the NF-κB pathway in bladder cancer (BC) remains unknown. In the current investigation, T24 BC cells were exposed to hypoxia, or by plasmid transfection to overexpress HAF or RelA (P65) to demonstrate their roles. The results indicate that hypoxia leads to the elevation of HAF plus activation of the NF-κB pathway, accompanied by the switch of HIF-1α to HIF-2α, resulting in the enhanced ability of malignancy in T24 cells. In order to further demonstrate the significance of this switch, HIF-1α and HIF-2α were co-transfected into T24 cells with HIF-β, respectively. The following results indicate that the T24hif-2α/β cells show enhanced ability of malignancy, accompanied by the maintenance of stem-cell markers, but the T24hif-1α/β cells show higher expression of metabolism-related genes. Boyden assays and wound-healing assays indicate the enhanced ability of malignancy for T24hif-2α/β. Thus, we conclude that on the hypoxic microenvironment, the switching of HIF-1α to HIF-2α, which is driven by HAF through activating the NF-κB pathway, contributes to the malignancy of T24 cells, accompanied by the maintenance of stem-cell markers. This provides us an avenue for understanding the progression of bladder cancer.
Collapse
Affiliation(s)
- Zhenfeng Guan
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, P.R. China
| | - Chen Ding
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, P.R. China
| | - Yiqing Du
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, P.R. China
| | - Kai Zhang
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, P.R. China
| | - Jian Ning Zhu
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, P.R. China
| | - Tingting Zhang
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, P.R. China
| | - Dalin He
- Department of Urology, The First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Shan Xu
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, P.R. China
| | - Xinyang Wang
- Oncology Research Lab, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, P.R. China
| | - Jinhai Fan
- Department of Urology, The First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, P.R. China
| |
Collapse
|
169
|
Ho CS, Yap SH, Phuah NH, In LLA, Hasima N. MicroRNAs associated with tumour migration, invasion and angiogenic properties in A549 and SK-Lu1 human lung adenocarcinoma cells. Lung Cancer 2013; 83:154-62. [PMID: 24360396 DOI: 10.1016/j.lungcan.2013.11.024] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 11/20/2013] [Accepted: 11/27/2013] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Dysregulation in miRNA expression contributes towards the initiation and progression of metastasis by regulating multiple target genes. In this study, variations in miRNA expression profiles were investigated between high and low invasive NSCLC cell lines followed by identification of miRNAs with targets governing NSCLC's metastatic potential. MATERIALS AND METHODS Two NSCLC sub-cell lines possessing opposing migration and invasion properties were established using serial transwell invasion assays. Global miRNA expression profiles were obtained using microarray followed by RT-qPCR validation. Target prediction and pathway enrichment analyses were conducted on dysregulated miRNAs using DIANA-mirPath, DIANA-microT 4.0 and TargetScan 5.2 softwares. Metastatic effects of dysregulated miRNAs were evaluated using wound healing assay, invasion assay and HUVEC angiogenesis assay following transfection with mimics and inhibitors. RESULTS A total of eleven differentially expressed miRNAs were revealed from microarray analyses, with four miRNAs validated through RT-qPCR. Three of these miRNAs were further selected for biological function validations, with only two modulating metastasis. A pathway model describing interactions between miRNAs and metastasis highlighted four major pathways: non-canonical Wnt/PCP, TGF-β, MAPK and integrin-FAK-Src signalling cascade. CONCLUSION These results provide a list of potential candidate metastatic markers during the classification of NSCLCs and a platform for the development of bio-therapeutics targeting these miRNA control elements.
Collapse
Affiliation(s)
- Chai San Ho
- Institute of Biological Sciences, Division of Genetics and Molecular Biology, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Seow Hui Yap
- Institute of Biological Sciences, Division of Genetics and Molecular Biology, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Neoh Hun Phuah
- Institute of Biological Sciences, Division of Genetics and Molecular Biology, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Lionel L A In
- Institute of Biological Sciences, Division of Genetics and Molecular Biology, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Noor Hasima
- Institute of Biological Sciences, Division of Genetics and Molecular Biology, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia; Centre for Research in Biotechnology for Agriculture (CEBAR), University of Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
170
|
RANDLE DIANDRAD, CLARKE SHINEKA, HENDERSON VERONICA, ODERO-MARAH VALERIEA. Snail mediates invasion through uPA/uPAR and the MAPK signaling pathway in prostate cancer cells. Oncol Lett 2013; 6:1767-1773. [PMID: 26889270 PMCID: PMC4738196 DOI: 10.3892/ol.2013.1635] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Accepted: 09/17/2013] [Indexed: 01/17/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a process by which cancer cells acquire mesenchymal properties, such as induction of vimentin, while epithelial-associated genes like E-cadherin are lost. This enables cells to be more metastatic. Factors that are able to induce EMT include growth factors such as transforming growth factor-β (TGF-β) and epidermal growth factor, and transcription factors such as Snail. Snail-induced EMT promotes migration and invasion and we hypothesized that this may be mediated by the activity of urokinase-type plasminogen activator (uPA) and its receptor (uPAR). LNCaP, 22Rv1 and ARCaP human prostate cancer (CaP) cells stably transfected with empty vector control (Neo) or constitutively active Snail exhibited increased cell invasion. Superarray analysis revealed an upregulation in uPA and uPAR RNA expression in Snail-transfected ARCaP cells compared with that of a Neo control. In addition, the protein expression levels of Snail, uPA and uPAR were measured by western blot analysis which showed that overexpression of Snail increased uPA and uPAR protein levels. The activity of uPA in conditioned media was measured using an ELISA which revealed that uPA activity was elevated in LNCaP, 22Rv1 and ARCaP cells overexpressing Snail. Additionally, transient silencing of uPAR in ARCaP cells overexpressing Snail using short interfering RNA resulted in abrogation of Snail-mediated invasion. Snail overexpression was associated with increased extracellular-signal-regulated kinase activity, and antagonism of this activity with mitogen-activated protein (MAPK) inhibitor, UO126, inhibited cell invasion and decreased uPA activity. Therefore, Snail-mediated cell invasion in human CaP cells may occur via the regulation of uPA/uPAR and the MAPK signaling pathway.
Collapse
Affiliation(s)
- DIANDRA D. RANDLE
- Department of Biological Sciences, Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
| | - SHINEKA CLARKE
- Department of Biological Sciences, Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
| | - VERONICA HENDERSON
- Department of Biological Sciences, Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
| | - VALERIE A. ODERO-MARAH
- Department of Biological Sciences, Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
| |
Collapse
|
171
|
Qi Y, Wang CC, He YL, Zou H, Liu CX, Pang LJ, Hu JM, Jiang JF, Zhang WJ, Li F. The correlation between morphology and the expression of TGF-β signaling pathway proteins and epithelial-mesenchymal transition-related proteins in synovial sarcomas. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2013; 6:2787-99. [PMID: 24294365 PMCID: PMC3843259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Accepted: 10/31/2013] [Indexed: 06/02/2023]
Abstract
Synovial sarcoma (SS) is a malignant tumor of soft tissue and is noted for late local recurrence and metastasis. Aberrant epithelial-mesenchymal transition (EMT) has been implicated in the pathogenesis of diverse human malignancies. Immunohistochemical techniques were used to assess EMT-related proteins (E-cadherin, N-cadherin, β-catenin, Snail, and Slug) and the TGF-β1 pathway (TGF-β1 and Smad2/3) proteins expression in different histological subtypes and epithelial mesenchymal compositions of SS. The expression of cell-surface (E-cadherin) and cytoskeletal proteins (β-catenin) were higher significantly in biphasic SSs (BSSs) (70.4%, 51.9%) than MFSSs (both for 10%). Among monophasic fibrous SSs (MFSSs) samples, E-cadherin protein expression was negatively correlated with expression Snail, Slug, TGF-β1, and Smad2/3. The expression levels of Snail and Smad2/3 were correlated with the pTNM stage (I-II vs. III-IV; P=0.047, P=0.021) and TGF-β1 exhibited a tendency toward a positive correlation with pTNM stage (I-II vs. III-IV; P=0.052), but did not correlate with the histological grade (p>0.05). Interestingly, our data showed that expression of E-cadherin protein correlated with greater survival in SS patients. Overexpression of Snail, and TGF-β1 is associated with suppressed expression of E-cadherin in MFSSs, which supports the hypothesis that the MFSS subtype may have developed via neoplastic EMT.
Collapse
Affiliation(s)
- Yan Qi
- Department of Pathology, Shihezi University School of MedicineShihezi, Xinjiang, China
- Department of Oncology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and TechnologyWuhan, Hubei, China
| | - Cui-Cui Wang
- Department of Pathology, Shihezi University School of MedicineShihezi, Xinjiang, China
| | - Yong-Lai He
- Department of ICU Intensive Care, The First Affiliated Hospital Pathology, Shihezi University School of MedicineShihezi, Xinjiang, China
| | - Hong Zou
- Department of Pathology, Shihezi University School of MedicineShihezi, Xinjiang, China
| | - Chun-Xia Liu
- Department of Pathology, Shihezi University School of MedicineShihezi, Xinjiang, China
| | - Li-Juan Pang
- Department of Pathology, Shihezi University School of MedicineShihezi, Xinjiang, China
| | - Jian-Ming Hu
- Department of Pathology, Shihezi University School of MedicineShihezi, Xinjiang, China
| | - Jin-fang Jiang
- Department of Pathology, Shihezi University School of MedicineShihezi, Xinjiang, China
| | - Wen-Jie Zhang
- Department of Pathology, Shihezi University School of MedicineShihezi, Xinjiang, China
| | - Feng Li
- Department of Pathology, Shihezi University School of MedicineShihezi, Xinjiang, China
- Department of Oncology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and TechnologyWuhan, Hubei, China
| |
Collapse
|
172
|
Activation of Notch signaling is required for cholangiocarcinoma progression and is enhanced by inactivation of p53 in vivo. PLoS One 2013; 8:e77433. [PMID: 24204826 PMCID: PMC3813685 DOI: 10.1371/journal.pone.0077433] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 09/02/2013] [Indexed: 12/14/2022] Open
Abstract
Cholangiocacinoma (CC) is a cancer disease with rising incidence. Notch signaling has been shown to be deregulated in many cancers. However, the role of this signaling pathway in the carcinogenesis of CC is still not fully explored. In this study, we investigated the effects of Notch inhibition by γ-secretase inhibitor IX (GSI IX) in cultured human CC cell lines and we established a transgenic mouse model with liver specific expression of the intracellular domain of Notch (Notch-ICD) and inactivation of tumor suppressor p53. GSI IX treatment effectively impaired cell proliferation, migration, invasion, epithelial to mesenchymal transition and growth of softagar colonies. In vivo overexpression of Notch-ICD together with an inactivation of p53 significantly increased tumor burden and showed CC characteristics. Conclusion: Our study highlights the importance of Notch signaling in the tumorigenesis of CC and demonstrates that additional inactivation of p53 in vivo.
Collapse
|
173
|
Yamamoto N, Kinoshita T, Nohata N, Yoshino H, Itesako T, Fujimura L, Mitsuhashi A, Usui H, Enokida H, Nakagawa M, Shozu M, Seki N. Tumor-suppressive microRNA-29a inhibits cancer cell migration and invasion via targeting HSP47 in cervical squamous cell carcinoma. Int J Oncol 2013; 43:1855-63. [PMID: 24141696 PMCID: PMC3834344 DOI: 10.3892/ijo.2013.2145] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 09/23/2013] [Indexed: 01/04/2023] Open
Abstract
Our recent studies of microRNA (miRNA) expression signatures indicated that microRNA-29a (miR-29a) was significantly downregulated in several types of human cancers, suggesting that miR-29a may be a putative tumor-suppressive miRNA in human cancers. The aim of this study was to investigate the functional significance of miR-29a in cervical squamous cell carcinoma (SCC) and to identify novel miR-29a-regulated cancer pathways and target genes involved in cervical SCC oncogenesis and metastasis. Restoration of miR-29a in cervical cancer cell lines (CaSKi, HeLa, ME180 and Yumoto) revealed that this miRNA significantly inhibited cancer cell migration and invasion. Gene expression data and in silico analysis demonstrated that heat-shock protein 47 (HSP47), a member of the serpin superfamily of serine proteinase inhibitors and a molecular chaperone involved in the maturation of collagen molecules, was a potential target of miR-29a regulation. Luciferase reporter assays showed that miR-29a directly regulated HSP47. Moreover, silencing of the HSP47 gene significantly inhibited cell migration and invasion in cancer cells and the expression of HSP47 was upregulated in cancer tissues and cervical intraepithelial neoplasia (CIN), as demonstrated by immunostaining. Downregulation of miR-29a was a frequent event in cervical SCC and miR-29a acted as a tumor suppressor by directly targeting HSP47. Recognition of tumor-suppressive miRNA-regulated molecular targets provides new insights into the potential mechanisms of cervical SCC oncogenesis and metastasis and suggests novel therapeutic strategies for treatment of this disease.
Collapse
Affiliation(s)
- Noriko Yamamoto
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Chiba, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
174
|
Espinoza I, Pochampally R, Xing F, Watabe K, Miele L. Notch signaling: targeting cancer stem cells and epithelial-to-mesenchymal transition. Onco Targets Ther 2013; 6:1249-59. [PMID: 24043949 PMCID: PMC3772757 DOI: 10.2147/ott.s36162] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Notch signaling is an evolutionarily conserved pathway involved in cell fate control during development, stem cell self-renewal, and postnatal tissue differentiation. Roles for Notch in carcinogenesis, the biology of cancer stem cells, tumor angiogenesis, and epithelial-to-mesenchymal transition (EMT) have been reported. This review describes the role of Notch in the "stemness" program in cancer cells and in metastases, together with a brief update on the Notch inhibitors currently under investigation in oncology. These agents may be useful in targeting cancer stem cells and to reverse the EMT process.
Collapse
Affiliation(s)
- Ingrid Espinoza
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS, USA
| | - Radhika Pochampally
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Biochemistry, University of Mississippi Medical Center, Jackson, MS, USA
| | - Fei Xing
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - Kounosuke Watabe
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Microbiology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Lucio Miele
- Cancer Institute, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, USA
| |
Collapse
|
175
|
Xu K, Wang F, Pan X, Liu R, Ma J, Kong F, Tang B. High selectivity imaging of nitroreductase using a near-infrared fluorescence probe in hypoxic tumor. Chem Commun (Camb) 2013; 49:2554-6. [PMID: 23423494 DOI: 10.1039/c3cc38980d] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A highly selective and sensitive near-infrared (NIR) fluorescence probe (Cy-NO2) for imaging nitroreductase was developed and was successfully applied to investigating the relationship between epithelial-mesenchymal transitions (EMTs) in tumour progression and intracellular hypoxic level.
Collapse
Affiliation(s)
- Kehua Xu
- College of Chemistry, Chemical Engineering and Materials Science, Engineering Research Center of Pesticide and Medicine Intermediate Clean Production, Ministry of Education, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, P. R. China
| | | | | | | | | | | | | |
Collapse
|
176
|
Role of fucosyltransferase IV in epithelial-mesenchymal transition in breast cancer cells. Cell Death Dis 2013; 4:e735. [PMID: 23887626 PMCID: PMC3730415 DOI: 10.1038/cddis.2013.241] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 05/13/2013] [Accepted: 05/30/2013] [Indexed: 11/09/2022]
Abstract
Epithelial–mesenchymal transition (EMT) is a crucial step in tumor progression and has an important role during cancer invasion and metastasis. Although fucosyltransferase IV (FUT4) has been implicated in the modulation of cell migration, invasion and cancer metastasis, its role during EMT is unclear. This study explores the molecular mechanisms of the involvement of FUT4 in EMT in breast cancer cells. Breast cancer cell lines display increased expression of FUT4, which is accompanied by enhanced appearance of the mesenchymal phenotype and which can be reversed by knockdown of endogenous FUT4. Moreover, FUT4 induced activation of phosphatidylinositol 3-kinase (PI3K)/Akt, and inactivation of GSK3β and nuclear translocation of NF-κB, resulting in increased Snail and MMP-9 expression and greater cell motility. Taken together, these findings indicate that FUT4 has a role in EMT through activation of the PI3K/Akt and NF-κB signaling systems, which induce the key mediators Snail and MMP-9 and facilitate the acquisition of a mesenchymal phenotype. Our findings support the possibility that FUT4 is a novel regulator of EMT in breast cancer cells and a promising target for cancer therapy.
Collapse
|
177
|
Xu M, Cao Y, Jiang MM, Jie ZG, Li ZR. MiR495- and miR551a-mediated down-regulation of PRL-3 expression inhibits peritoneal metastasis in a mouse model of gastric cancer. Shijie Huaren Xiaohua Zazhi 2013; 21:1693-1700. [DOI: 10.11569/wcjd.v21.i18.1693] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [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 effect of miR495 and miR551 on PRL-3 expression in gastric cancer SGC7901 cells to explore gene interference therapies for gastric cancer.
METHODS: SGC7901 cells stably transfected with miR495 or miR551a and non-transfected SGC7901 cells were inoculated intraperitoneally in BALB/CA (nu/nu) nude mice. One month after inoculation, the mice were killed to observe the rate of tumor formation, tumor growth and migration. Real-time PCR was performed to quantify mRNA levels of miR495, miR551a and PRL-3.
RESULTS: Compared to nude mice inoculated with non-transfected cells, miR495 and miR551a levels were increased, PRL-3 level was decreased, and migration of gastric cancer was suppressed in mice inoculated with SGC7901 cells stably transfected with miR495 or miR551a.
CONCLUSION: MiR495 and miR551a can suppress the migration of gastric cancer by inhibiting PRL-3 expression in mice.
Collapse
|
178
|
Wei X, Yang X, Han ZP, Qu FF, Shao L, Shi YF. Mesenchymal stem cells: a new trend for cell therapy. Acta Pharmacol Sin 2013; 34:747-54. [PMID: 23736003 DOI: 10.1038/aps.2013.50] [Citation(s) in RCA: 683] [Impact Index Per Article: 56.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs), the major stem cells for cell therapy, have been used in the clinic for approximately 10 years. From animal models to clinical trials, MSCs have afforded promise in the treatment of numerous diseases, mainly tissue injury and immune disorders. In this review, we summarize the recent opinions on methods, timing and cell sources for MSC administration in clinical applications, and provide an overview of mechanisms that are significant in MSC-mediated therapies. Although MSCs for cell therapy have been shown to be safe and effective, there are still challenges that need to be tackled before their wide application in the clinic.
Collapse
|
179
|
Crosstalk between breast cancer stem cells and metastatic niche: emerging molecular metastasis pathway? Tumour Biol 2013; 34:2019-30. [PMID: 23686802 DOI: 10.1007/s13277-013-0831-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 05/01/2013] [Indexed: 12/21/2022] Open
Abstract
Metastatic colonization represents the final step of metastasis, and is the major cause of cancer mortality. Metastasis as an "inefficient" process requires the right population of tumor cells in a suitable microenvironment to form secondary tumors. Cancer stem cells are the only capable population of tumor cells to progress to overt metastasis. On the other hand, the occurrence of appropriate microenvironmental conditions within the target tissue would be critical for metastasis formation. Metastatic niche seems to be the specialized microenvironment to support tumor initiating cells at the distant organ. Master regulators not only determine cancer stem cell state, but also may have regulatory roles in metastatic niche elements. Meanwhile, both cancer stem cell and metastatic niche may function like two sides of the metastatic coin. Hypoxia inducible factors have multiple roles in regulation of both sides of this coin. TGF-β superfamily, also, have been considered as master regulators of epithelial to mesenchymal transition and metastasis and may play crucial roles in regulation of metastatic niche as well. In this regard, we hypothesize the presence of a possible emerging molecular pathway in the biological process of breast cancer metastasis. In this process, non-Smad TGF-β-induced metastasis connects cancer stem cell and metastatic niche formation through a central path, "Metastasis Pathway".
Collapse
|
180
|
Genome-wide association study identifies common variants in SLC39A6 associated with length of survival in esophageal squamous-cell carcinoma. Nat Genet 2013; 45:632-8. [PMID: 23644492 DOI: 10.1038/ng.2638] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 04/12/2013] [Indexed: 12/18/2022]
Abstract
We conducted a genome-wide scan of SNPs to identify variants associated with length of survival in 1,331 individuals with esophageal squamous-cell carcinoma (ESCC), with associations validated in 2 independent sets including 1,962 individuals with this cancer. We identified rs1050631 in SLC39A6 as associated with the survival times of affected individuals, with the hazard ratio for death from ESCC in the combined sample being 1.30 (95% confidence interval (CI) = 1.19-1.43; P = 3.77 × 10(-8)). rs7242481, located in the 5' UTR of SLC39A6, disturbs a transcriptional repressor binding site and results in upregulation of SLC39A6 expression. Immunohistochemical staining of ESCC tissues showed that higher expression of SLC39A6 protein was correlated with shorter length of survival in individuals with advanced ESCC (P = 0.013). Knockdown of SLC39A6 expression suppressed proliferation and invasion in ESCC cells. These results suggest that SLC39A6 has an important role in the prognosis of ESCC and may be a potential therapeutic target.
Collapse
|
181
|
Seo J, Park SJ, Kim J, Choi SJ, Moon SH, Chung HM. Effective method for the isolation and proliferation of primary lung cancer cells from patient lung tissues. Biotechnol Lett 2013; 35:1165-74. [PMID: 23592304 DOI: 10.1007/s10529-013-1189-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 03/20/2013] [Indexed: 02/02/2023]
Abstract
We have developed a technique for isolating and culturing primary lung cancer cells extracted from patient tissue to facilitate anti-cancer drug development. Patient-derived lung cancer tissues were mechanically dissociated to 40-100 μm. Dispase was then used to isolate cultured lung cancer cell populations, which were re-plated on Matrigel-coated dishes containing N2-supplemented medium and growth factors. This method allows pure populations of primary non-small cell lung cancer cells to be grown in vitro. The isolated cells exhibited hallmark cancerous properties such as abnormal chromosomes and in vivo tumor formation. The cell lines generated through this procedure may help to advance our knowledge of certain forms of lung cancer and may also be useful for developing patient-specific anti-cancer drug screening procedures.
Collapse
Affiliation(s)
- Joseph Seo
- Stem Cell Research Laboratory, CHA Stem Cell Institute, CHA University, 605-21, Yoeksam 1-dong, Gangnam-gu, Seoul, 135-907, South Korea
| | | | | | | | | | | |
Collapse
|
182
|
Capaccione KM, Pine SR. The Notch signaling pathway as a mediator of tumor survival. Carcinogenesis 2013; 34:1420-30. [PMID: 23585460 DOI: 10.1093/carcin/bgt127] [Citation(s) in RCA: 220] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The Notch signaling pathway is evolutionarily conserved and responsible for cell fate determination in the developing embryo and mature tissue. At the molecular level, ligand binding activates Notch signaling by liberating the Notch intracellular domain, which then translocates into the nucleus and activates gene transcription. Despite the elegant simplicity of this pathway, which lacks secondary messengers or a signaling cascade, Notch regulates gene expression in a highly context- and cell-type-dependent manner. Notch signaling is frequently dysregulated, most commonly by overactivation, across many cancers and confers a survival advantage on tumors, leading to poorer outcomes for patients. Recent studies demonstrate how Notch signaling increases tumor cell proliferation and provide evidence that active Notch signaling maintains the cancer stem-cell pool, induces epithelial-mesenchymal transition and promotes chemoresistance. These studies imply that pharmacological inhibition of Notch signaling may refine control of cancer therapy and improve patient survival. Gamma secretase inhibitors (GSIs) are drugs that inhibit Notch signaling and may be successful in controlling cancer cell growth in conjunction with standard chemotherapy, but substantial side effects have hampered their widespread use. Recent efforts have been aimed at the development of antibodies against specific Notch receptors and ligands with the hope of limiting side effects while providing the same therapeutic benefit as GSIs. Together, studies characterizing Notch signaling and modulation have offered hope that refined methods targeting Notch may become powerful tools in anticancer therapeutics.
Collapse
Affiliation(s)
- Kathleen M Capaccione
- Department of Medicine, The Cancer Institute of New Jersey, UMDNJ/Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | | |
Collapse
|
183
|
Okajima M, Kokura S, Ishikawa T, Mizushima K, Tsuchiya R, Matsuyama T, Adachi S, Okayama T, Sakamoto N, Kamada K, Katada K, Uchiyama K, Handa O, Takagi T, Yagi N, Naito Y, Yoshikawa T. Anoxia/reoxygenation induces epithelial-mesenchymal transition in human colon cancer cell lines. Oncol Rep 2013; 29:2311-7. [PMID: 23589103 DOI: 10.3892/or.2013.2401] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Accepted: 11/07/2012] [Indexed: 12/13/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is considered to be a crucial event in the development of cancer metastasis. Anoxia/reoxygenation (A/R) is known to occur in cancer tissues due to angiogenesis and changes in tissue pressure that occur during tumor growth. We investigated whether A/R induces EMT in the human colon cancer cell line HT-29. Colon cancer cells were exposed to anoxia (2 h) followed by reoxygenation (4-22 h) and evaluated for EMT changes using immunofluorescence and western blot analyses. We also investigated the expression of EMT-related transcription factors (Snail and ZEB1) using RT-PCR and evaluated the expression of NF-κB using ELISA. To determine whether NF-κB is involved in A/R-induced EMT, HT-29 cells were treated with proteasome inhibitors. Colon cancer cells exposed to A/R underwent EMT morphological changes; the cancer cells acquired a spindle-shaped phenotype. The expression of E-cadherin on the cell surface and the total amount of E-cadherin proteins were reduced after A/R. The expression of EMT-related transcription factors (Snail, ZEB1) was increased after A/R. Pretreatment with proteasome inhibitors significantly attenuated the downregulation of E-cadherin induced by A/R. These results indicate that A/R induces EMT in human colon cancer cells through an NF-κB-dependent transcriptional pathway.
Collapse
Affiliation(s)
- Manabu Okajima
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Chimal-Ramírez GK, Espinoza-Sánchez NA, Fuentes-Pananá EM. Protumor activities of the immune response: insights in the mechanisms of immunological shift, oncotraining, and oncopromotion. JOURNAL OF ONCOLOGY 2013; 2013:835956. [PMID: 23577028 PMCID: PMC3612474 DOI: 10.1155/2013/835956] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 01/25/2013] [Indexed: 12/15/2022]
Abstract
Experimental and clinical studies indicate that cells of the innate and adaptive immune system have both anti- and pro-tumor activities. This dual role of the immune system has led to a conceptual shift in the role of the immune system's regulation of cancer, in which immune-tumor cell interactions are understood as a dynamic process that comprises at least five phases: immunosurveillance, immunoselection, immunoescape, oncotraining, and oncopromotion. The tumor microenvironment shifts immune cells to perform functions more in tune with the tumor needs (oncotraining); these functions are related to chronic inflammation and tissue remodeling activities. Among them are increased proliferation and survival, increased angiogenesis and vessel permeability, protease secretion, acquisition of migratory mesenchymal characteristics, and self-renewal properties that altogether promote tumor growth and metastasis (oncopromotion). Important populations in all these pro-tumor processes are M2 macrophages, N2 neutrophils, regulatory T cells, and myeloid derived suppressor cells; the main effectors molecules are CSF-1, IL-6, metalloproteases, VEGF, PGE-2, TGF- β , and IL-10. Cancer prognosis correlates with densities and concentrations of protumoral populations and molecules, providing ideal targets for the intelligent design of directed preventive or anticancer therapies.
Collapse
Affiliation(s)
- G. K. Chimal-Ramírez
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias (UIMEIP), Hospital de Pediatría Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Avenida Cuauhtémoc 330, Colonia Doctores, 06725 Delegación Cuauhtémoc, DF, Mexico
- Programa de Doctorado en Ciencias Quimicobiológicas del Instituto Politécnico Nacional (IPN), Mexico
| | - N. A. Espinoza-Sánchez
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias (UIMEIP), Hospital de Pediatría Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Avenida Cuauhtémoc 330, Colonia Doctores, 06725 Delegación Cuauhtémoc, DF, Mexico
- Programa de Doctorado en Ciencias Biomédicas de la Universidad Autónoma de México (UNAM), Mexico
| | - E. M. Fuentes-Pananá
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias (UIMEIP), Hospital de Pediatría Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Avenida Cuauhtémoc 330, Colonia Doctores, 06725 Delegación Cuauhtémoc, DF, Mexico
| |
Collapse
|
185
|
Morrison CD, Parvani JG, Schiemann WP. The relevance of the TGF-β Paradox to EMT-MET programs. Cancer Lett 2013; 341:30-40. [PMID: 23474494 DOI: 10.1016/j.canlet.2013.02.048] [Citation(s) in RCA: 162] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 01/03/2013] [Accepted: 02/26/2013] [Indexed: 02/06/2023]
Abstract
The role of transforming growth factor-β (TGF-β) during tumorigenesis is complex and paradoxical, reflecting its ability to function as a tumor suppressor in normal and early-stage cancers, and as a tumor promoter in their late-stage counterparts. The switch in TGF-β function is known as the "TGF-β Paradox," whose manifestations are intimately linked to the initiation of epithelial-mesenchymal transition (EMT) programs in developing and progressing carcinomas. Indeed, as carcinoma cells emerge from EMT programs stimulated by TGF-β, they readily display a variety of acquired phenotypes that provide a selective advantage to growing carcinomas, including (i) enhanced cell migration and invasion; (ii) heightened resistance to cytotoxic agents, targeted chemotherapeutic, and radiation treatments; and (iv) boosted expansion of cancer-initiating and stem-like cell populations that underlie tumor metastasis and disease recurrence. At present, the molecular, cellular, and microenvironmental mechanisms that enable post-EMT and metastatic carcinoma cells to hijack the oncogenic activities of TGF-β remain incompletely understood. Additionally, the molecular mechanisms that counter EMT programs and limit the aggressiveness of late-stage carcinomas, events that transpire via mesenchymal-epithelial transition (MET) reactions, also need to be further elucidated. Here we review recent advances that provide new insights into how TGF-β promotes EMT programs in late-stage carcinoma cells, as well as how these events are balanced by MET programs during the development and metastatic progression of human carcinomas.
Collapse
Affiliation(s)
- Chevaun D Morrison
- Case Comprehensive Cancer Center, Division of General Medical Sciences-Oncology, Case Western Reserve University, Wolstein Research Building, 2103 Cornell Road Cleveland, OH 44106, United States
| | | | | |
Collapse
|
186
|
El Khatib M, Kalnytska A, Palagani V, Kossatz U, Manns MP, Malek NP, Wilkens L, Plentz RR. Inhibition of hedgehog signaling attenuates carcinogenesis in vitro and increases necrosis of cholangiocellular carcinoma. Hepatology 2013; 57:1035-45. [PMID: 23172661 DOI: 10.1002/hep.26147] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 10/01/2012] [Indexed: 12/22/2022]
Abstract
UNLABELLED The Hedgehog signaling pathway plays a pivotal role during embryonic development, stem cell maintenance, and wound healing. Hedgehog signaling also is deregulated in many cancers. However, the role of this signaling pathway in the carcinogenesis of cholangiocarcinoma (CCC) is still unknown. In this study, we investigated the effects of Hedgehog inhibition by cyclopamine and 5E1 in cultured human CCC cell lines and in vivo using a xenograft mouse model. We also investigated the involvement of Hedgehog in epithelial to mesenchymal transition (EMT), migration, and CCC tumor growth. Sonic hedgehog (Shh) ligand was highly expressed in 89% of human CCC tissues and in CCC cell lines. Cyclopamine and 5E1 treatments effectively inhibited cell proliferation, migration, and invasion by down-regulating the Hedgehog target genes glioblastoma 1 and glioblastoma 2. In vitro and in vivo, we detected an increase in epithelial marker, E-cadherin, after Hedgehog inhibition. In addition, we saw an increase in necrotic areas and a decrease in mitotic figures in cyclopamine and 5E1-treated CCC xenograft tumors. CONCLUSION This study supports the presence of autocrine Hedgehog signaling in human CCC, where CCC cells produce and respond to Shh ligand. Blocking the Hedgehog pathway inhibited EMT and decreased the viability of CCC cells. In addition, cyclopamine and 5E1 inhibited the growth of CCC xenograft tumors.
Collapse
Affiliation(s)
- Mona El Khatib
- Department of Internal Medicine I, Medical University Hospital, Tuebingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
187
|
Mooney BM, Raof NA, Li Y, Xie Y. Convergent mechanisms in pluripotent stem cells and cancer: Implications for stem cell engineering. Biotechnol J 2013; 8:408-19. [DOI: 10.1002/biot.201200202] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 12/03/2012] [Accepted: 01/02/2013] [Indexed: 12/24/2022]
|
188
|
Yang X, Hou J, Han Z, Wang Y, Hao C, Wei L, Shi Y. One cell, multiple roles: contribution of mesenchymal stem cells to tumor development in tumor microenvironment. Cell Biosci 2013; 3:5. [PMID: 23336752 PMCID: PMC3693909 DOI: 10.1186/2045-3701-3-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 11/20/2012] [Indexed: 12/13/2022] Open
Abstract
The discovery of tissue reparative and immunosuppressive abilities of mesenchymal stem cells (MSCs) has drawn more attention to tumor microenvironment and its role in providing the soil for the tumor cell growth. MSCs are recruited to tumor which is referred as the never healing wound and altered by the inflammation environment, thereby helping to construct the tumor microenvironment. The environment orchestrated by MSCs and other factors can be associated with angiogenesis, immunosuppression, inhibition of apoptosis, epithelial-mesenchymal transition (EMT), survival of cancer stem cells, which all contribute to tumor growth and progression. In this review, we will discuss how MSCs are recruited to the tumor microenvironment and what effects they have on tumor progression.
Collapse
Affiliation(s)
- Xue Yang
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, the Second Military Medicial University, 225 Changhai Road, Shanghai 200438, China.
| | | | | | | | | | | | | |
Collapse
|
189
|
Zhu QC, Gao RY, Wu W, Qin HL. Epithelial-mesenchymal transition and its role in the pathogenesis of colorectal cancer. Asian Pac J Cancer Prev 2013; 14:2689-98. [PMID: 23803016 DOI: 10.7314/apjcp.2013.14.5.2689] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a collection of events that allows the conversion of adherent epithelial cells, tightly bound to each other within an organized tissue, into independent fibroblastic cells possessing migratory properties and the ability to invade the extracellular matrix. EMT contributes to the complex architecture of the embryo by permitting the progression of embryogenesis from a simple single-cell layer epithelium to a complex three-dimensional organism composed of both epithelial and mesenchymal cells. However, in most tissues EMT is a developmentally restricted process and fully differentiated epithelia typically maintain their epithelial phenotype. Recently, elements of EMT, specially the loss of epithelial markers and the gain of mesenchymal markers, have been observed in pathological states, including epithelial cancers. Increasing evidence has confirmed its presence in human colon during colorectal carcinogenesis. In general, chronic inflammation is considered to be one of the causes of many human cancers including colorectal cancer(CRC). Accordingly, epidemiologic and clinical studies indicate that patients affected by ulcerative colitis and Crohn's disease, the two major forms of inflammatory bowel disease, have an increased risk of developing CRC. A large body of evidence supports roles for the SMAD/STAT3 signaling pathway, the NF-kB pathway, the Ras-mitogen- activated protein kinase/Snail/Slug and microRNAs in the development of colorectal cancers via epithelial-to- mesenchymal transition. Thus, EMT appears to be closely involved in the pathogenesis of colorectal cancer, and analysis refered to it can yield novel targets for therapy.
Collapse
Affiliation(s)
- Qing-Chao Zhu
- Department of Surgery, The Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | | | | | | |
Collapse
|
190
|
Wierstra I. FOXM1 (Forkhead box M1) in tumorigenesis: overexpression in human cancer, implication in tumorigenesis, oncogenic functions, tumor-suppressive properties, and target of anticancer therapy. Adv Cancer Res 2013; 119:191-419. [PMID: 23870513 DOI: 10.1016/b978-0-12-407190-2.00016-2] [Citation(s) in RCA: 138] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
FOXM1 (Forkhead box M1) is a typical proliferation-associated transcription factor and is also intimately involved in tumorigenesis. FOXM1 stimulates cell proliferation and cell cycle progression by promoting the entry into S-phase and M-phase. Additionally, FOXM1 is required for proper execution of mitosis. In accordance with its role in stimulation of cell proliferation, FOXM1 exhibits a proliferation-specific expression pattern and its expression is regulated by proliferation and anti-proliferation signals as well as by proto-oncoproteins and tumor suppressors. Since these factors are often mutated, overexpressed, or lost in human cancer, the normal control of the foxm1 expression by them provides the basis for deregulated FOXM1 expression in tumors. Accordingly, FOXM1 is overexpressed in many types of human cancer. FOXM1 is intimately involved in tumorigenesis, because it contributes to oncogenic transformation and participates in tumor initiation, growth, and progression, including positive effects on angiogenesis, migration, invasion, epithelial-mesenchymal transition, metastasis, recruitment of tumor-associated macrophages, tumor-associated lung inflammation, self-renewal capacity of cancer cells, prevention of premature cellular senescence, and chemotherapeutic drug resistance. However, in the context of urethane-induced lung tumorigenesis, FOXM1 has an unexpected tumor suppressor role in endothelial cells because it limits pulmonary inflammation and canonical Wnt signaling in epithelial lung cells, thereby restricting carcinogenesis. Accordingly, FOXM1 plays a role in homologous recombination repair of DNA double-strand breaks and maintenance of genomic stability, that is, prevention of polyploidy and aneuploidy. The implication of FOXM1 in tumorigenesis makes it an attractive target for anticancer therapy, and several antitumor drugs have been reported to decrease FOXM1 expression.
Collapse
|
191
|
Jia L, Zhang S, Ye Y, Li X, Mercado-Uribe I, Bast RC, Liu J. Paclitaxel inhibits ovarian tumor growth by inducing epithelial cancer cells to benign fibroblast-like cells. Cancer Lett 2012; 326:176-82. [PMID: 22902993 PMCID: PMC3495569 DOI: 10.1016/j.canlet.2012.08.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 08/03/2012] [Accepted: 08/07/2012] [Indexed: 11/25/2022]
Abstract
Paclitaxel is commonly used to treat multiple human malignancies, but its mechanism of action is still poorly defined. Human ovarian cancer SKOV3 cells (parental SKOV3) were treated with paclitaxel (1μM) for 2days, and the morphologic changes in the cells were monitored for more than 4months. Parental SKOV3 underwent a markedly morphologic transition from the epithelial to fibroblast-like phenotype following treatment with paclitaxel; the resulting cells were designated as SKOV3-P. The SKOV3-P cells' proliferative ability was assessed via a 3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide (MTT) assay. The molecular characteristics of these cells were assessed via immunocytochemical staining and Western blot analysis. Their invasiveness and tumor formation ability was evaluated via wound-scratch and colony formation assays. The tumorigenicity of SKOV3-P cells was assessed in vivo after subcutaneous injection of tumor cells between injections of parental and paclitaxel-treated cells in nude mice. SKOV3-P cells have decreased the proliferation and invasion ability, decreased colony-forming ability when cultured in Matrigel and lost their tumor formation as compared with parental SKOV3 cells when injected in nude mice. SKOV3-P cells have decreased expression of E-cadherin, cytokeratin, Snail, PI3K, and P-Akt-Ser473, and increased expression of fibronectin, vimentin, Slug, P27, and PTEN. These results demonstrated that paclitaxel can inhibit tumor growth by inducing ovarian cancer epithelial cells toward a benign fibroblast-like phenotype through dysregulation of previously known pathways involved in the regulation of epithelial to mesenchymal transition (EMT), which may represent a novel mechanism for paclitaxel-induced tumor suppression.
Collapse
Affiliation(s)
- Lizhou Jia
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Obstetrics and Gynecology, The Affiliated People’s Hospital of Inner Mongolia Medical College, Inner Mongolia Autonomous Region, China
| | - Shiwu Zhang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yanfen Ye
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Cancer Research Institute, South Medical University, Guangzhou, Guangdong, China
| | - Xin Li
- Cancer Research Institute, South Medical University, Guangzhou, Guangdong, China
| | - Imelda Mercado-Uribe
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert C. Bast
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jinsong Liu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
192
|
Saito D, Kyakumoto S, Chosa N, Ibi M, Takahashi N, Okubo N, Sawada S, Ishisaki A, Kamo M. Transforming growth factor-β1 induces epithelial–mesenchymal transition and integrin α3β1-mediated cell migration of HSC-4 human squamous cell carcinoma cells through Slug. ACTA ACUST UNITED AC 2012; 153:303-15. [DOI: 10.1093/jb/mvs144] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
193
|
Winer DA, Winer S, Rotstein L, Asa SL, Mete O. Villous papillary thyroid carcinoma: a variant associated with marfan syndrome. Endocr Pathol 2012; 23:254-9. [PMID: 22847364 DOI: 10.1007/s12022-012-9219-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Marfan syndrome (MFS) is an autosomal dominant hereditary disorder of connective tissue associated with perturbations in transforming growth factor β (TGF-β) biology, most often due to mutations in FBN1 gene that encodes fibrillin-1. To our knowledge, there is no known association of MFS with thyroid carcinoma. We report a 46-year-old man with known history of MFS who developed an unusual histological variant of papillary thyroid carcinoma. The tumor exhibited a widely invasive florid papillary growth pattern with prominent long villous fronds. Immunohistochemical and molecular analysis revealed a BRAF(V600E) mutation, evidence of aggressive biomarker expression (positivity for HBME-1, cytokeratin 19, galectin-3 and cyclin D1, and loss of p27), and changes associated with TGF-β-related epithelial-to-mesenchymal transition with active phospho-SMAD signaling. We introduce a unique histological pattern of papillary thyroid carcinoma that is associated with MFS. The combination of BRAF(V600E) mutation in the setting of altered TGF-β signaling and weak connective tissue integrity associated with MFS may cooperate and possibly be responsible to form this unique villous morphology with epithelial-to-mesenchymal transition and invasive growth.
Collapse
Affiliation(s)
- Daniel A Winer
- Department of Pathology, University Health Network, Toronto, ON, Canada
| | | | | | | | | |
Collapse
|
194
|
Zheng J, Xiong D, Sun X, Wang J, Hao M, Ding T, Xiao G, Wang X, Mao Y, Fu Y, Shen K, Wang J. Signification of Hypermethylated in Cancer 1 (HIC1) as Tumor Suppressor Gene in Tumor Progression. CANCER MICROENVIRONMENT : OFFICIAL JOURNAL OF THE INTERNATIONAL CANCER MICROENVIRONMENT SOCIETY 2012; 5:285-93. [PMID: 22528874 PMCID: PMC3460058 DOI: 10.1007/s12307-012-0103-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Accepted: 03/28/2012] [Indexed: 12/30/2022]
Abstract
Hypermethylated in cancer 1(HIC1) was identified as a strong suppressor gene in chromosome region 17p13.3 telomeric to TP53. This gene encodes a transcriptional repressor and is ubiquitously expressed in normal tissues but downexpressed in different tumor tissues where it is hypermethylated. The hypermethylation of this chromosomal region leads to epigenetic inactivation of HIC1, which would prompt cancer cells to alter survival and signaling pathways or specific transcription factors during the period of tumorigenesis. In vitro, HIC1 function is mainly a sequence-specific transcriptional repressor interacting with a still growing range of histone deacetylase(HDAC)-dependent and HDAC-independent corepressor complexes. Furthermore, a role for HIC1 in tumor development is firmly supported by Hic1 deficient mouse model and two double heterozygote models cooperate with p53 and Ptch1. Notably, our findings suggest that potential factors derived from tumor microenviroment may play a role in modulating HIC1 expression in tumor cells by epigenetic modification, which is responsible for tumor progression. In this review, we will describe genomic and proteinic structure of HIC1, and summary the potential role of HIC1 in human various solid tumors and leukemia, and explore the influence of tumor microenviroment on inducing HIC1 expression in tumor cells.
Collapse
Affiliation(s)
- Jianghua Zheng
- Department of Biochemistry and Molecular & Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Dan Xiong
- Department of Biochemistry and Molecular & Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Xueqing Sun
- Department of Biochemistry and Molecular & Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Jinglong Wang
- Department of Biochemistry and Molecular & Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Mingang Hao
- Department of Biochemistry and Molecular & Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Tao Ding
- Department of Urological Surgery, Shanghai the Tenth People’s Hospital of Tong Ji University, Shanghai, 200072 China
| | - Gang Xiao
- Department of Biochemistry and Molecular & Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Xiumin Wang
- Department of Biochemistry and Molecular & Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Yan Mao
- Shanghai Ruijin Hospital, Comprehensive Breast Health Center, Shanghai, 200025 China
| | - Yuejie Fu
- Department of Thoracic Surgery, RenJi Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Kunwei Shen
- Shanghai Ruijin Hospital, Comprehensive Breast Health Center, Shanghai, 200025 China
| | - Jianhua Wang
- Department of Biochemistry and Molecular & Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| |
Collapse
|
195
|
Bao B, Azmi AS, Ali S, Ahmad A, Li Y, Banerjee S, Kong D, Sarkar FH. The biological kinship of hypoxia with CSC and EMT and their relationship with deregulated expression of miRNAs and tumor aggressiveness. BIOCHIMICA ET BIOPHYSICA ACTA 2012; 1826:272-96. [PMID: 22579961 PMCID: PMC3788359 DOI: 10.1016/j.bbcan.2012.04.008] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 04/25/2012] [Accepted: 04/28/2012] [Indexed: 12/13/2022]
Abstract
Hypoxia is one of the fundamental biological phenomena that are intricately associated with the development and aggressiveness of a variety of solid tumors. Hypoxia-inducible factors (HIF) function as a master transcription factor, which regulates hypoxia responsive genes and has been recognized to play critical roles in tumor invasion, metastasis, and chemo-radiation resistance, and contributes to increased cell proliferation, survival, angiogenesis and metastasis. Therefore, tumor hypoxia with deregulated expression of HIF and its biological consequence lead to poor prognosis of patients diagnosed with solid tumors, resulting in higher mortality, suggesting that understanding of the molecular relationship of hypoxia with other cellular features of tumor aggressiveness would be invaluable for developing newer targeted therapy for solid tumors. It has been well recognized that cancer stem cells (CSCs) and epithelial-to-mesenchymal transition (EMT) phenotypic cells are associated with therapeutic resistance and contribute to aggressive tumor growth, invasion, metastasis and believed to be the cause of tumor recurrence. Interestingly, hypoxia and HIF signaling pathway are known to play an important role in the regulation and sustenance of CSCs and EMT phenotype. However, the molecular relationship between HIF signaling pathway with the biology of CSCs and EMT remains unclear although NF-κB, PI3K/Akt/mTOR, Notch, Wnt/β-catenin, and Hedgehog signaling pathways have been recognized as important regulators of CSCs and EMT. In this article, we will discuss the state of our knowledge on the role of HIF-hypoxia signaling pathway and its kinship with CSCs and EMT within the tumor microenvironment. We will also discuss the potential role of hypoxia-induced microRNAs (miRNAs) in tumor development and aggressiveness, and finally discuss the potential effects of nutraceuticals on the biology of CSCs and EMT in the context of tumor hypoxia.
Collapse
Affiliation(s)
- Bin Bao
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Asfar S. Azmi
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Shadan Ali
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Aamir Ahmad
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Yiwei Li
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Sanjeev Banerjee
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Dejuan Kong
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Fazlul H. Sarkar
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| |
Collapse
|
196
|
IL-8 Regulates The Epithelial-mesenchymal Transition (EMT) of Renal Cancer Through PKC/ERK Signaling Pathway*. PROG BIOCHEM BIOPHYS 2012. [DOI: 10.3724/sp.j.1206.2012.00298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
197
|
Palagani V, El Khatib M, Kossatz U, Bozko P, Müller MR, Manns MP, Krech T, Malek NP, Plentz RR. Epithelial mesenchymal transition and pancreatic tumor initiating CD44+/EpCAM+ cells are inhibited by γ-secretase inhibitor IX. PLoS One 2012; 7:e46514. [PMID: 23094026 PMCID: PMC3477166 DOI: 10.1371/journal.pone.0046514] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 09/04/2012] [Indexed: 12/14/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with a high rate of metastasis. Recent studies have indicated that the Notch signalling pathway is important in PDAC initiation and maintenance, although the specific cell biological roles of the pathway remain to be established. Here we sought to examine this question in established pancreatic cancer cell lines using the γ-secretase inhibitor IX (GSI IX) to inactivate Notch. Based on the known roles of Notch in development and stem cell biology, we focused on effects on epithelial mesenchymal transition (EMT) and on pancreatic tumor initiating CD44+/EpCAM+ cells. We analyzed the effect of the GSI IX on growth and epithelial plasticity of human pancreatic cancer cell lines, and on the tumorigenicity of pancreatic tumor initiating CD44+/EpCAM+ cells. Notably, apoptosis was induced after GSI IX treatment and EMT markers were selectively targeted. Furthermore, under GSI IX treatment, decline in the growth of pancreatic tumor initiating CD44+/EpCAM+ cells was observed in vitro and in a xenograft mouse model. This study demonstrates a central role of Notch signalling pathway in pancreatic cancer pathogenesis and identifies an effective approach to inhibit selectively EMT and suppress tumorigenesis by eliminating pancreatic tumor initiating CD44+/EpCAM+ cells.
Collapse
Affiliation(s)
- Vindhya Palagani
- Department of Internal Medicine I, Medical University Hospital, Tuebingen, Germany
| | - Mona El Khatib
- Department of Internal Medicine I, Medical University Hospital, Tuebingen, Germany
| | - Uta Kossatz
- Department of Internal Medicine I, Medical University Hospital, Tuebingen, Germany
| | - Przemyslaw Bozko
- Department of Internal Medicine I, Medical University Hospital, Tuebingen, Germany
| | - Martin R. Müller
- Department of Internal Medicine II, Medical University Hospital, Tuebingen, Germany
| | - Michael P. Manns
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Till Krech
- Institute for Pathology, Hannover Medical School, Hannover, Germany
| | - Nisar P. Malek
- Department of Internal Medicine I, Medical University Hospital, Tuebingen, Germany
| | - Ruben R. Plentz
- Department of Internal Medicine I, Medical University Hospital, Tuebingen, Germany
| |
Collapse
|
198
|
Luo W, Fang W, Li S, Yao K. Aberrant expression of nuclear vimentin and related epithelial-mesenchymal transition markers in nasopharyngeal carcinoma. Int J Cancer 2012; 131:1863-1873. [PMID: 22307379 DOI: 10.1002/ijc.27467] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2011] [Accepted: 01/17/2012] [Indexed: 02/05/2023]
Abstract
Expression of vimentin and the epithelial to mesenchymal transition (EMT) markers E-cadherin, β-catenin is essential for the progression of various human cancers. Our study aimed to investigate the aberrant localization E-cadherin, β-catenin and vimentin, and their prognostic significance in 122 nasopharyngeal carcinoma (NPC) patients by immunohistochemistry and immunofluorescence. Our results showed that both membranous and cytoplasmic localization of E-cadherin staining were associated with lymph node metastasis (p = 0.000 and 0.005, respectively) and clinical stage (p = 0.000 and 0.007, respectively). High cytoplasmic β-catenin correlated significantly with larger tumor size (p = 0.020), lymph node metastasis (p = 0.000) and advanced clinical stage (p = 0.036). However, no significant difference was observed between membranous β-catenin and clinicopathologic features (p ≥ 0.05). High nuclear vimentin expression correlated significantly with positive lymph node metastasis (p = 0.000) and advanced clinical stage (p = 0.000). Multivariate analysis showed that nuclear vimentin and cytoplasmic E-cadherin were independent prognostic factors (p = 0.016 and 0.001, respectively), as well as M classification (p = 0.001). More importantly, patients with high coexpression of nuclear vimentin and cytoplasmic E-cadherin had shorter survival time (p = 0.000). Furthermore, high coexpression of these two proteins was closely associated with lymph node metastasis (p = 0.000) and advanced clinical stage (p = 0.000). Our studies provide convincing evidence that EMT may play an important role in the biological progression of NPC, and nuclear vimentin and cytoplasmic E-cadherin might have independent prognostic value in NPC patients and serve as novel targets for prognostic therapeutics.
Collapse
Affiliation(s)
- Weiren Luo
- Cancer Research Institute, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | | | | | | |
Collapse
|
199
|
Ruan JS, Liu YP, Zhang L, Yan LG, Fan FT, Shen CS, Wang AY, Zheng SZ, Wang SM, Lu Y. Luteolin reduces the invasive potential of malignant melanoma cells by targeting β3 integrin and the epithelial-mesenchymal transition. Acta Pharmacol Sin 2012; 33:1325-31. [PMID: 22983392 DOI: 10.1038/aps.2012.93] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
AIM To investigate whether luteolin, a highly prevalent flavonoid, reverses the effects of epithelial-mesenchymal transition (EMT) in vitro and in vivo and to determine the mechanisms underlying this reversal. METHODS Murine malignant melanoma B16F10 cells were exposed to 1% O(2) for 24 h. Cellular mobility and adhesion were assessed using Boyden chamber transwell assay and cell adhesion assay, respectively. EMT-related proteins, such as E-cadherin and N-cadherin, were examined using Western blotting. Female C57BL/6 mice (6 to 8 weeks old) were injected with B16F10 cells (1×10(6) cells in 0.2 mL per mouse) via the lateral tail vein. The mice were treated with luteolin (10 or 20 mg/kg, ip) daily for 23 d. On the 23rd day after tumor injection, the mice were sacrificed, and the lungs were collected, and metastatic foci in the lung surfaces were photographed. Tissue sections were analyzed with immunohistochemistry and HE staining. RESULTS Hypoxia changed the morphology of B16F10 cells in vitro from the cobblestone-like to mesenchymal-like strips, which was accompanied by increased cellular adhesion and invasion. Luteolin (5-50 μmol/L) suppressed the hypoxia-induced changes in the cells in a dose-dependent manner. Hypoxia significantly decreased the expression of E-cadherin while increased the expression of N-cadherin in the cells (indicating the occurrence of EMT-like transformation), which was reversed by luteolin (5 μmol/L). In B16F10 cells, luteolin up-regulated E-cadherin at least partly via inhibiting the β3 integrin/FAK signal pathway. In experimental metastasis model mice, treatment with luteolin (10 or 20 mg/kg) reduced metastatic colonization in the lungs by 50%. Furthermore, the treatment increased the expression of E-cadherin while reduced the expression of vimentin and β3 integrin in the tumor tissues. CONCLUSION Luteolin inhibits the hypoxia-induced EMT in malignant melanoma cells both in vitro and in vivo via the regulation of β3 integrin, suggesting that luteolin may be applied as a potential anticancer chemopreventative and chemotherapeutic agent.
Collapse
|
200
|
Benson MD, Serrano MJ. Ephrin regulation of palate development. Front Physiol 2012; 3:376. [PMID: 23055980 PMCID: PMC3458271 DOI: 10.3389/fphys.2012.00376] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 09/02/2012] [Indexed: 11/17/2022] Open
Abstract
Studies of palate development are motivated by the all too common incidence of cleft palate, a birth defect that imposes a tremendous health burden and can leave lasting disfigurement. Although, mechanistic studies of palate growth and fusion have focused on growth factors such as Transforming Growth Factor ß-3 (Tgfß3), recent studies have revealed that the ephrin family of membrane bound ligands and their receptors, the Ephs, play central roles in palatal morphogenesis, growth, and fusion. In this mini-review, we will discuss the recent findings by our group and others on the functions of ephrins in palatal development.
Collapse
Affiliation(s)
- M Douglas Benson
- Department of Biomedical Sciences, Texas A&M Health Science Center Baylor College of Dentistry Dallas, TX, USA
| | | |
Collapse
|