Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Theranostics ; 11(16): 7671-7684, 2021.
Article in English | MEDLINE | ID: mdl-34335957

ABSTRACT

Snail1 is a transcriptional factor required for epithelial to mesenchymal transition and activation of cancer-associated fibroblasts (CAF). Apart from that, tumor endothelial cells also express Snail1. Here, we have unraveled the role of Snail1 in this tissue in a tumorigenic context. Methods: We generated transgenic mice with an endothelial-specific and inducible Snail1 depletion. This murine line was crossed with MMTV-PyMT mice that develop mammary gland tumors and the consequence of Snail1 depletion in the endothelium were investigated. We also interfere Snail1 expression in cultured endothelial cells. Results: Specific Snail1 depletion in the endothelium of adult mice does not promote an overt phenotype; however, it delays the formation of mammary gland tumors in MMTV-PyMT mice. These effects are associated to the inability of Snail1-deficient endothelial cells to undergo angiogenesis and to enhance CAF activation in a paracrine manner. Moreover, tumors generated in mice with endothelium-specific Snail1 depletion are less advanced and show a papillary phenotype. Similar changes on onset and tumor morphology are observed by pretreatment of MMTV-PyMT mice with the angiogenic inhibitor Bevacizumab. Human breast papillary carcinomas exhibit a lower angiogenesis and present lower staining of Snail1, both in endothelial and stromal cells, compared with other breast neoplasms. Furthermore, human breast tumors datasets show a strong correlation between Snail1 expression and high angiogenesis. Conclusion: These findings show a novel role for Snail1 in endothelial cell activation and demonstrate that these cells impact not only on angiogenesis, but also on tumor onset and phenotype.


Subject(s)
Breast Neoplasms/genetics , Snail Family Transcription Factors/metabolism , Animals , Breast Neoplasms/metabolism , Cancer-Associated Fibroblasts/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Endothelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/physiology , Female , Fibroblasts/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neovascularization, Pathologic/pathology , Snail Family Transcription Factors/genetics , Transcription Factors/metabolism
2.
Nat Commun ; 12(1): 3932, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34168145

ABSTRACT

Chemical descriptors encode the physicochemical and structural properties of small molecules, and they are at the core of chemoinformatics. The broad release of bioactivity data has prompted enriched representations of compounds, reaching beyond chemical structures and capturing their known biological properties. Unfortunately, bioactivity descriptors are not available for most small molecules, which limits their applicability to a few thousand well characterized compounds. Here we present a collection of deep neural networks able to infer bioactivity signatures for any compound of interest, even when little or no experimental information is available for them. Our signaturizers relate to bioactivities of 25 different types (including target profiles, cellular response and clinical outcomes) and can be used as drop-in replacements for chemical descriptors in day-to-day chemoinformatics tasks. Indeed, we illustrate how inferred bioactivity signatures are useful to navigate the chemical space in a biologically relevant manner, unveiling higher-order organization in natural product collections, and to enrich mostly uncharacterized chemical libraries for activity against the drug-orphan target Snail1. Moreover, we implement a battery of signature-activity relationship (SigAR) models and show a substantial improvement in performance, with respect to chemistry-based classifiers, across a series of biophysics and physiology activity prediction benchmarks.


Subject(s)
Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Cell Line, Tumor , Databases, Pharmaceutical , Drug Evaluation, Preclinical/methods , Humans , Snail Family Transcription Factors/antagonists & inhibitors , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism
3.
Nat Commun ; 9(1): 3420, 2018 08 24.
Article in English | MEDLINE | ID: mdl-30143639

ABSTRACT

Lamins (A/C and B) are major constituents of the nuclear lamina (NL). Structurally conserved lamina-associated domains (LADs) are formed by genomic regions that contact the NL. Lamins are also found in the nucleoplasm, with a yet unknown function. Here we map the genome-wide localization of lamin B1 in an euchromatin-enriched fraction of the mouse genome and follow its dynamics during the epithelial-to-mesenchymal transition (EMT). Lamin B1 associates with actively expressed and open euchromatin regions, forming dynamic euchromatin lamin B1-associated domains (eLADs) of about 0.3 Mb. Hi-C data link eLADs to the 3D organization of the mouse genome during EMT and correlate lamin B1 enrichment at topologically associating domain (TAD) borders with increased border strength. Having reduced levels of lamin B1 alters the EMT transcriptional signature and compromises the acquisition of mesenchymal traits. Thus, during EMT, the process of genome reorganization in mouse involves dynamic changes in eLADs.


Subject(s)
Lamin Type B/metabolism , Animals , Cell Nucleus/metabolism , Chromatin Immunoprecipitation , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/physiology , Euchromatin/chemistry , Euchromatin/genetics , Euchromatin/metabolism , Fluorescence Recovery After Photobleaching , Humans , Lamin Type B/chemistry , Lamin Type B/genetics , Mice
4.
Sci Rep ; 8(1): 11467, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30065348

ABSTRACT

Syndecan 1 (SDC-1) is a cell surface proteoglycan with a significant role in cell adhesion, maintaining epithelial integrity. SDC1 expression is inversely related to aggressiveness in prostate cancer (PCa). During epithelial to mesenchymal transition (EMT), loss of epithelial markers is mediated by transcriptional repressors such as SNAIL, SLUG, or ZEB1/2 that bind to E-box promoter sequences of specific genes. The effect of these repressors on SDC-1 expression remains unknown. Here, we demonstrated that SNAIL, SLUG and ZEB1 expressions are increased in advanced PCa, contrarily to SDC-1. SNAIL, SLUG and ZEB1 also showed an inversion to SDC-1 in prostate cell lines. ZEB1, but not SNAIL or SLUG, represses SDC-1 as demonstrated by experiments of ectopic expression in epithelial prostate cell lines. Inversely, expression of ZEB1 shRNA in PCa cell line increased SDC-1 expression. The effect of ZEB1 is transcriptional since ectopic expression of this gene represses SDC-1 promoter activity and ZEB1 binds to the SDC-1 promoter as detected by ChIP assays. An epigenetic mark associated to transcription repression H3K27me3 was bound to the same sites that ZEB1. In conclusion, this study identifies ZEB1 as a key repressor of SDC-1 during PCa progression and point to ZEB1 as a potentially diagnostic marker for PCa.


Subject(s)
Prostatic Neoplasms/genetics , Syndecan-1/genetics , Transcription Factors/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics , Cell Adhesion/genetics , Cell Line, Tumor , Epigenesis, Genetic/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , PC-3 Cells , Promoter Regions, Genetic/genetics , Snail Family Transcription Factors/genetics , Transcription, Genetic/genetics
6.
Am J Pathol ; 187(1): 55-69, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27863213

ABSTRACT

The aim of the present study was to analyze in vivo the role of zinc finger protein SNAI1 (SNAI1) on renal fibrosis. Unilateral ureteral obstruction injury was induced in Snai1 knockout mice. Snai1 gene deletion was, however, only partial and could therefore not be correlated to reduced fibrosis. Expression of SNAI1 protein and epithelial-mesenchymal transformation markers was then assessed in human chronic allograft nephropathy biopsy specimens. Significant up-regulation of SNAI1 protein was detected within cytoplasm of proximal tubules localized, for some of them, near foci of fibrosis and tubular atrophy. No concomitant epithelial-mesenchymal transformation could, however, be demonstrated analyzing the expression of the fibroblast markers vimentin, α-smooth muscle actin, and S100A4. SNAI1 cytoplasmic up-regulation was particularly evident in biopsy specimens obtained from calcineurin inhibitor-treated patients, which might be because of, as suggested by in vitro experiments, a decrease of the proteasome chimotrypsin activity. Deeper analysis on chronic allograft nephropathy biopsy specimens suggested that SNAI1 cytoplasmic up-regulation was preceded by a transient increase of phosphorylated heat shock protein 27, p38 mitogen-activated protein kinase, and glycogen synthase kinase 3ß. Concomitant down-regulation of the polyubuquitinylated conjugates was detected in SNAI1+ tubules. Altogether, these results might suggest that calcineurin inhibitor-induced tubular SNAI1 protein cytoplasmic accumulation, possibly because of impaired SNAI1 proteasomal degradation and nuclear translocation, might be a sign of a diseased profibrotic epithelial phenotype.


Subject(s)
Cytoplasm/metabolism , Epithelial Cells/metabolism , Kidney Transplantation , Kidney Tubules/metabolism , Kidney Tubules/pathology , Snail Family Transcription Factors/metabolism , Zinc Fingers , Allografts/drug effects , Animals , Biopsy , Calcineurin Inhibitors/pharmacology , Chronic Disease , Dogs , Epithelial-Mesenchymal Transition/drug effects , Female , Fibrosis , Glycogen Synthase Kinase 3 beta/metabolism , HSP27 Heat-Shock Proteins/metabolism , Kidney Transplantation/adverse effects , Kidney Tubules/drug effects , Madin Darby Canine Kidney Cells , Male , Mice, Knockout , Phenotype , Phosphorylation/drug effects , Proteasome Endopeptidase Complex/metabolism , Up-Regulation/drug effects , Ureteral Obstruction/pathology , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Semin Cancer Biol ; 36: 71-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26506454

ABSTRACT

F-box proteins are the key recognition subunit of multimeric E3 ubiquitin ligase complexes that participate in the proteasome degradation of specific substrates. In the last years, a discrete number of F-box proteins have been shown to regulate the epithelial-to-mesenchymal transition (EMT), a process defined by a rapid change of cell phenotype, the loss of epithelial characteristics and the acquisition of a more invasive phenotype. Specific EMT transcription factors (EMT-TFs), such as Snail, Slug, Twist and Zeb, control EMT induction both during development and in cancer. These EMT-TFs are short-lived proteins that are targeted to the proteasome system by specific F-box proteins, keeping them at low levels. F-box proteins also indirectly regulate the EMT process by controlling EMT inducers, such as Notch, c-Myc or mTOR. Here we summarize the role that these F-box proteins (Fbxw1, Fbxw7, Fbxl14, Fbxl5, Fbxo11 and Fbxo45) play in controlling EMT during development and cancer progression, a process dependent on post-translational modifications that govern their interaction with target proteins.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , F-Box Proteins/genetics , F-Box Proteins/metabolism , Animals , Cullin Proteins/chemistry , Cullin Proteins/genetics , Cullin Proteins/metabolism , Gene Expression Regulation , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Multiprotein Complexes , Neoplasms/etiology , Neoplasms/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Snail Family Transcription Factors/metabolism , Twist-Related Protein 1/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism
10.
Nucleic Acids Res ; 43(12): 5785-97, 2015 Jul 13.
Article in English | MEDLINE | ID: mdl-25990740

ABSTRACT

In this report we have analyzed the role of antisense transcription in the control of LEF1 transcription factor expression. A natural antisense transcript (NAT) is transcribed from a promoter present in the first intron of LEF1 gene and undergoes splicing in mesenchymal cells. Although this locus is silent in epithelial cells, and neither NAT transcript nor LEF1 mRNA are expressed, in cell lines with an intermediate epithelial-mesenchymal phenotype presenting low LEF1 expression, the NAT is synthesized and remains unprocessed. Contrarily to the spliced NAT, this unspliced NAT down-regulates the main LEF1 promoter activity and attenuates LEF1 mRNA transcription. Unspliced LEF1 NAT interacts with LEF1 promoter and facilitates PRC2 binding to the LEF1 promoter and trimethylation of lysine 27 in histone 3. Expression of the spliced form of LEF1 NAT in trans prevents the action of unspliced NAT by competing for interaction with the promoter. Thus, these results indicate that LEF1 gene expression is attenuated by an antisense non-coding RNA and that this NAT function is regulated by the balance between its spliced and unspliced forms.


Subject(s)
Gene Expression Regulation , Lymphoid Enhancer-Binding Factor 1/genetics , RNA Splicing , RNA, Antisense/metabolism , Cell Line , Epithelial Cells/metabolism , Humans , Lymphoid Enhancer-Binding Factor 1/biosynthesis , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic , Transcription, Genetic
11.
Oncoscience ; 2(2): 131-41, 2015.
Article in English | MEDLINE | ID: mdl-25859556

ABSTRACT

Many studies have demonstrated that the endocannabinoid system (ECS) is altered in different tumor types, including colon cancer. However, little is known about the role of the ECS in tumor progression. Here we report the correlation between CB 2 expression and pathological data in a series of 175 colorectal cancer patients, as well as the response of the HT29 colon cancer-derived cell line upon CB 2 activation. CB 2 mRNA was detected in 28.6% of samples tested. It was more frequent in N+ patients and predicts disease free survival and overall survival in colon cancer. In positive samples, CB 2 was expressed with great intensity in tumor epithelial cells and correlated with tumor growth. Treatment of HT29 with CB 2 agonist revealed membrane loss of E-cadherin and SNAIL1 overexpression. A direct correlation between CB 2 and SNAIL1 expression was also found in human tumors. CB 2 receptor expression is a poor prognostic marker for colon cancer and the activation of this receptor, with non-apoptotic doses of agonists, could be collaborating with disease progression. These results raise the question whether the activation of CB 2 should be considered as anti-tumoral therapy.

12.
PLoS One ; 9(7): e102041, 2014.
Article in English | MEDLINE | ID: mdl-25025131

ABSTRACT

The Wilms' tumor transcription factor (WT1) was originally classified as a tumor suppressor, but it is now known to also be associated with cancer progression and poor prognosis in several malignancies. WT1 plays an essential role in orchestrating a developmental process known as mesenchymal-to-epithelial transition (MET) during kidney development, but also induces the reverse process, epithelial-to-mesenchymal transition (EMT) during heart development. WT1 is not expressed in the adult kidney, but shows elevated expression in clear cell renal cell carcinoma (ccRCC). However, the role of WT1 in this disease has not been characterized. In this study, we demonstrate that WT1 is upregulated in ccRCC cells that are deficient in the expression of the von Hippel-Lindau tumor suppressor protein (VHL). We found that WT1 transcriptionally activated Snail, a master transcriptional repressor that is known to induce EMT. Although Snail represses E-cadherin and induces mesenchymal characteristics, we found partial maintenance of E-cadherin and associated epithelial characteristics in kidney cells and ccRCC cells that express WT1, since WT1 upregulates E-cadherin expression and competes with Snail repression. These findings support a novel paradigm in which WT1 induces an epithelial-mesenchymal hybrid transition (EMHT), characterized by Snail up-regulation with E-cadherin maintenance, a tumor cell differentiation state in which cancer cells keep both EMT and MET characteristics which may promote tumor cell plasticity and tumor progression.


Subject(s)
Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Epithelial-Mesenchymal Transition/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Wnt Proteins/genetics , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Snail Family Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , WT1 Proteins/genetics
13.
Int J Cancer ; 134(12): 2984-90, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24242829

ABSTRACT

Snail1 is a transcriptional factor that plays an important role in epithelial-mesenchymal transition and in the acquisition of invasive properties by epithelial cells. In colon tumors, Snail1 expression in the stroma correlates with lower specific survival of cancer patients. However, the role(s) of Snail1 expression in stroma and its association with patients' survival have not been determined. We used human primary carcinoma-associated fibroblasts (CAFs) or normal fibroblasts (NFs) and fibroblast cell lines to analyze the effects of Snail1 expression on the protumorigenic capabilities in colon cancer cells. Snail1 expression was higher in CAFs than in NFs and, as well as α-SMA, a classic marker of activated CAFs. Moreover, in tumor samples from 50 colon cancer patients, SNAI1 expression was associated with expression of other CAF markers, such as α-SMA and fibroblast activation protein. Interestingly, coculture of CAFs with colon cells induced a significant increase in epithelial cell migration and proliferation, which was associated with endogenous SNAI1 expression levels. Ectopic manipulation of Snail1 in fibroblasts demonstrated that Snail1 expression controlled migration as well as proliferation of cocultured colon cancer cells in a paracrine manner. Furthermore, expression of Snail1 in fibroblasts was required for the coadjuvant effect of these cells on colon cancer cell growth and invasion when coxenografted in nude mice. Finally, cytokine profile changes, particularly MCP-3 expression, in fibroblasts are put forward as mediators of Snail1-derived effects on colon tumor cell migration. In summary, these studies demonstrate that Snail1 is necessary for the protumorigenic effects of fibroblasts on colon cancer cells.


Subject(s)
Carcinogenesis , Colonic Neoplasms/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Actins/genetics , Actins/metabolism , Animals , Cell Cycle , Cell Movement , Cell Proliferation , Coculture Techniques , Colonic Neoplasms/genetics , Cytokines/metabolism , Endopeptidases , Female , Fibroblasts/pathology , Gelatinases/genetics , Gelatinases/metabolism , Gene Expression , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , RNA, Messenger/biosynthesis , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Snail Family Transcription Factors , Tumor Cells, Cultured
14.
Mol Cell Oncol ; 1(2): e960770, 2014.
Article in English | MEDLINE | ID: mdl-27308335

ABSTRACT

Constitutive heterochromatin, an essential structure that has been conserved throughout evolution, is required to maintain genome stability. Although heterochromatin is enriched for repressive traits, it can be actively transcribed to generate thousands of noncoding RNAs that are required for correct chromatin assembly. Despite the importance of this structure, how and why heterochromatin transcription is regulated, and the proteins responsible for this regulation, remain poorly understood. Here, we summarize recent findings in heterochromatin transcription regulation during different cellular processes with a focus on the epithelial-mesenchymal transition (EMT), which elicits important changes in cell behavior, has a key role in early development, and is involved in cancer progression.

15.
Clin Cancer Res ; 19(21): 6006-19, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24025712

ABSTRACT

PURPOSE: Cancer-associated fibroblasts (CAF) are essential components of the stroma that play a critical role in cancer progression. This study aimed to identify novel CAFs markers that might contribute to the invasion and the prognosis of colorectal cancer. EXPERIMENTAL DESIGN: The azoxymethane/dextran sodium sulfate mouse model of sporadic colon cancer represents an adequate source for the isolation of CAFs and normal fibroblasts. By using the explants technique, we purified CAFs and normal fibroblasts from colon tissues. Whole-cell extracts and supernatants were subjected to in-depth quantitative proteomic analysis by tandem mass spectrometry. Further validations of upregulated proteins in CAFs were carried out by chemokine microarray and immunohistochemical analyses of mouse and human tissues. RESULTS: Using a fold-change of 1.4 or more, we found 132 and 125 differentially expressed proteins in whole-cell extracts and supernatants, respectively. We found CAFs-associated proinflammatory and desmoplastic signatures. The proinflammatory signature was composed of several cytokines. Among them, CCL2 and CCL8 caused an increase in migration and invasion of colorectal cancer KM12 cells. The desmoplastic signature was composed of 30 secreted proteins. In mouse and human samples, expression of LTBP2, CDH11, OLFML3, and, particularly, FSTL1 was significantly increased in the tumoral stroma, without significant expression in the cancer epithelial cells. The combination of CALU and CDH11 stromal expression showed a significant association with disease-free survival and poor prognosis. CONCLUSION: We have identified LTBP2, CDH11, OLFML3, and FSTL1 as selective biomarkers of cancer stroma, and CALU and CDH11 as candidate stromal biomarkers of prognostic significance in colon cancer.


Subject(s)
Colorectal Neoplasms/metabolism , Fibroblasts/metabolism , Inflammation Mediators/metabolism , Proteome , Animals , Biomarkers , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Cytokines/metabolism , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Mice , Myofibroblasts/metabolism , Prognosis , Protein Interaction Maps , Reproducibility of Results , Stromal Cells/metabolism
16.
Clin Cancer Res ; 19(21): 5914-26, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24052018

ABSTRACT

PURPOSE: Cancer-associated fibroblasts (CAF) actively participate in reciprocal communication with tumor cells and with other cell types in the microenvironment, contributing to a tumor-permissive neighborhood and promoting tumor progression. The aim of this study is the characterization of how CAFs from primary human colon tumors promote migration of colon cancer cells. EXPERIMENTAL DESIGN: Primary CAF cultures from 15 primary human colon tumors were established. Their enrichment in CAFs was evaluated by the expression of various epithelial and myofibroblast specific markers. Coculture assays of primary CAFs with different colon tumor cells were performed to evaluate promigratory CAF-derived effects on cancer cells. Gene expression profiles were developed to further investigate CAF characteristics. RESULTS: Coculture assays showed significant differences in fibroblast-derived paracrine promigratory effects on cancer cells. Moreover, the association between CAFs' promigratory effects on cancer cells and classic fibroblast activation or stemness markers was observed. CAF gene expression profiles were analyzed by microarray to identify deregulated genes in different promigratory CAFs. The gene expression signature, derived from the most protumorogenic CAFs, was identified. Interestingly, this "CAF signature" showed a remarkable prognostic value for the clinical outcome of patients with colon cancer. Moreover, this prognostic value was validated in an independent series of 142 patients with colon cancer, by quantitative real-time PCR (qRT-PCR), with a set of four genes included in the "CAF signature." CONCLUSIONS: In summary, these studies show for the first time the heterogeneity of primary CAFs' effect on colon cancer cell migration. A CAF gene expression signature able to classify patients with colon cancer into high- and low-risk groups was identified.


Subject(s)
Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Transcriptome , Biomarkers/metabolism , Cell Line, Tumor , Cell Movement , Cluster Analysis , Coculture Techniques , Colonic Neoplasms/mortality , Colonic Neoplasms/pathology , Fibroblasts/pathology , Gene Expression Profiling , Golgi Apparatus/metabolism , Humans , Paracrine Communication , Phenotype , Prognosis
17.
J Dermatol Sci ; 72(2): 93-102, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23928229

ABSTRACT

BACKGROUND: Cutaneous squamous cell carcinoma (cSCC) is the second most common malignancy in humans and approximately 5% metastasize, usually to regional lymph nodes. Epithelial to mesenchymal transition (EMT) is a process involving loss of intercellular adhesion, acquisition of a mesenchymal phenotype and enhanced migratory potential; epithelial markers, such as E-cadherin, are down-regulated and mesenchymal proteins (Vimentin), increased. OBJECTIVE: To investigate the expression of EMT markers in metastatic SCC (MSCC) and their corresponding metastases, and to correlate them with clinico-pathological factors associated with an increased risk of metastasis. METHODS: We performed a retrospective study that included 146 cSCC samples (51 primary non-metastatic, 56 primary metastatic, 39 lymphatic metastases). Immunohistochemistry for E-cadherin, Vimentin, Snail, beta-catenin, Twist, Zeb1 and Podoplanin was performed. RESULTS: Loss of membranous E-cadherin was observed in 77% cSCCs, with no differences between MSCC and non-MSCC. Among the transcriptional factors controlling EMT, no significant Snail1 expression was detected. Twist, Zeb1, Vimentin, beta-catenin and Podoplanin were significantly overexpressed in MSCCs. Twist ectopic expression in SCC13 cells induced Zeb1, Vimentin and Podoplanin expression and E-cadherin delocalization. These changes resulted in a scattered migration pattern in vitro. Expression of EMT markers was decreased in the metastases when compared with the corresponding primary tumors. CONCLUSION: These results suggest that a partial EMT, characterized by the expression of Twist but without a total E-cadherin depletion, is involved in the acquisition of invasive traits by cSCC, but the process is downregulated in lymph node metastases.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/metabolism , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Lymphatic Metastasis , Skin Neoplasms/metabolism , Antigens, CD , Cadherins/metabolism , Down-Regulation , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Membrane Glycoproteins/metabolism , Nuclear Proteins/metabolism , Phenotype , Retrospective Studies , Risk , Snail Family Transcription Factors , Transcription Factors/metabolism , Twist-Related Protein 1/metabolism , Vimentin/metabolism , Zinc Finger E-box-Binding Homeobox 1 , beta Catenin/metabolism
18.
PLoS Genet ; 9(6): e1003531, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23785295

ABSTRACT

PARP inhibition can induce anti-neoplastic effects when used as monotherapy or in combination with chemo- or radiotherapy in various tumor settings; however, the basis for the anti-metastasic activities resulting from PARP inhibition remains unknown. PARP inhibitors may also act as modulators of tumor angiogenesis. Proteomic analysis of endothelial cells revealed that vimentin, an intermediary filament involved in angiogenesis and a specific hallmark of EndoMT (endothelial to mesenchymal transition) transformation, was down-regulated following loss of PARP-1 function in endothelial cells. VE-cadherin, an endothelial marker of vascular normalization, was up-regulated in HUVEC treated with PARP inhibitors or following PARP-1 silencing; vimentin over-expression was sufficient to drive to an EndoMT phenotype. In melanoma cells, PARP inhibition reduced pro-metastatic markers, including vasculogenic mimicry. We also demonstrated that vimentin expression was sufficient to induce increased mesenchymal/pro-metastasic phenotypic changes in melanoma cells, including ILK/GSK3-ß-dependent E-cadherin down-regulation, Snail1 activation and increased cell motility and migration. In a murine model of metastatic melanoma, PARP inhibition counteracted the ability of melanoma cells to metastasize to the lung. These results suggest that inhibition of PARP interferes with key metastasis-promoting processes, leading to suppression of invasion and colonization of distal organs by aggressive metastatic cells.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Melanoma, Experimental/genetics , Poly(ADP-ribose) Polymerases/genetics , Vimentin , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Breast Neoplasms/pathology , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Dogs , Endothelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells , Humans , MCF-7 Cells , Melanoma, Experimental/pathology , Mice , Neoplasm Invasiveness/genetics , Neoplasm Metastasis , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors , Vimentin/genetics , Vimentin/metabolism
19.
PLoS One ; 8(5): e63695, 2013.
Article in English | MEDLINE | ID: mdl-23696849

ABSTRACT

Fibroblast growth factor receptor 4 (FGFR4) is vital in early development and tissue repair. FGFR4 expression levels are very restricted in adult tissues, except in several solid tumors including colorectal cancer, which showed overexpression of FGFR4. Here, FGFR4 mutation analysis discarded the presence of activating mutations, other than Arg(388), in different colorectal cancer cell lines and tumoral samples. Stable shRNA FGFR4-silencing in SW480 and SW48 cell lines resulted in a significant decrease in cell proliferation, adhesion, cell migration and invasion. This decrease in the tumorigenic and invasive capabilities of colorectal cancer cells was accompanied by a decrease of Snail, Twist and TGFß gene expression levels and an increase of E-cadherin, causing a reversion to a more epithelial phenotype, in three different cell lines. In addition, FGFR4-signaling activated the oncogenic SRC, ERK1/2 and AKT pathways in colon cancer cells and promoted an increase in cell survival. The relevance of FGFR4 in tumor growth was supported by two different strategies. Kinase inhibitors abrogated FGFR4-related cell growth and signaling pathways at the same extent than FGFR4-silenced cells. Specific FGFR4-targeting using antibodies provoked a similar reduction in cell growth. Moreover, FGFR4 knock-down cells displayed a reduced capacity for in vivo tumor formation and angiogenesis in nude mice. Collectively, our data support a crucial role for FGFR4 in tumorigenesis, invasion and survival in colorectal cancer. In addition, FGFR4 targeting demonstrated its applicability for colorectal cancer therapy.


Subject(s)
Colorectal Neoplasms/metabolism , Epithelial-Mesenchymal Transition/physiology , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Blotting, Western , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Humans , Mice , Mice, Nude , Polymorphism, Single Nucleotide/genetics , Real-Time Polymerase Chain Reaction , Receptor, Fibroblast Growth Factor, Type 4/genetics
20.
J Immunol ; 190(8): 4408-19, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23509364

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a morphogenetic process characterized by the acquisition of mesenchymal properties linked with an invasive phenotype and metastasis of tumor cells. NK group 2, member D (NKG2D) is an NK cell-activating receptor crucially involved in cancer immunosurveillance. In this study, we show that induction of EMT by TGF-ß stimulation of human keratinocytes, by glycogen synthase kinase-3ß inhibition in several epithelial tumor cell lines, and by Snail1 overexpression in colorectal cancer cells strongly upregulated the expression of NKG2D ligands (NKG2DLs), MHC class I chain-related molecules A and B (MICA/B) and ULBP1-3. Overexpression of Snail1 and inhibition of glycogen synthase kinase-3ß in colorectal tumor cells markedly induced the activity of Sp1 transcription factor, which plays a key role in the upregulation of NKG2DL expression during EMT. The stimulation of MICA/B expression by TGF-ß treatment was independent of Sp1, but it involved posttranslational mechanisms mediated by mammalian target of rapamycin pathway. Accordingly, with the increased expression of NKG2DLs, triggering of EMT rendered cancer cells more susceptible to NKG2D-mediated killing by NK cells. In agreement, MICA/B were expressed in vivo in well-differentiated colorectal tumors with retained epithelial characteristics, whereas no expression of MICA/B was detected in poorly differentiated and invasive colorectal tumors that have lost epithelial characteristics. This decrease of MICA/B expression was associated with a dramatic increase of NKG2D(+)-tumor infiltrating lymphocytes. Overall, our findings indicate that EMT is a relevant checkpoint in the control of tumor progression through NKG2D-mediated immune responses.


Subject(s)
Colorectal Neoplasms/immunology , Epithelium/immunology , Mesoderm/immunology , NK Cell Lectin-Like Receptor Subfamily K/physiology , Animals , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Epithelium/metabolism , Epithelium/pathology , HT29 Cells , HeLa Cells , Hep G2 Cells , Humans , Immunophenotyping , Mesoderm/metabolism , Mesoderm/pathology , Mice , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Neoplasm Invasiveness/immunology , Neoplasm Invasiveness/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...