Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
Add more filters










Publication year range
1.
Int J Oncol ; 34(5): 1403-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19360353

ABSTRACT

Enhanced motility of cancer cells by remodelling of the actin cytoskeleton is crucial in the process of cancer cell invasion and metastasis. Although several studies propose a tumor suppressor role for the actin bundling protein myopodin, it was also shown previously that overexpression of mouse myopodin promotes invasion in vitro. In the present study, the role of myopodin in human cancer cell motility and invasion was explored using RNA interference with siRNA duplexes designed to down-regulate all human myopodin isoforms currently identified. We show that down-regulation of myopodin expression in human cancer cells significantly reduces the invasive properties of these cells both in collagen type I and in Matrigel. Furthermore, the motile characteristics of cancer cells are also curbed by reduced myopodin expression whereas cell-cell contacts are reinforced. These results point to a role for myopodin as tumor activator. While these findings are at variance with the suggested tumor suppressor role for myopodin, we hypothesize that the subcellular localization of the protein is involved in its suppressor or activator function in tumorigenesis.


Subject(s)
Cell Movement/genetics , Microfilament Proteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Line, Tumor , Cells, Cultured , Down-Regulation/physiology , Gene Expression Regulation, Neoplastic/physiology , Gene Knockdown Techniques , Humans , Male , Microfilament Proteins/antagonists & inhibitors , Microfilament Proteins/metabolism , Microfilament Proteins/physiology , Models, Biological , Neoplasm Invasiveness , RNA, Small Interfering/pharmacology , Tissue Distribution
2.
FASEB J ; 23(3): 916-28, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19017743

ABSTRACT

We recently established the critical role of the PTEN/MAGI-1b signalosome in stabilization of cell-cell contacts and suppression of invasiveness. The PTEN tumor suppressor is recruited to E-cadherin junctional complexes through the binding to the second PDZ domain of the MAGI-1b scaffolding molecule, whereas beta-catenin interacts with the fifth PDZ domain. To identify additional effectors of this signalosome, we used yeast 2-hybrid screening. Among the clones identified, we focused on TRIP6, which belongs to the zyxin family of proteins. We demonstrated that TRIP6 interacted directly with MAGI-1b by binding to its fifth PDZ domain. Ectopic expression of TRIP6 induced invasiveness in the epithelial MDCK and MDCKts-src cells in a PI3-kinase- and a NF-kappaB-dependent manner and impaired cell-cell aggregation at least in part by uncoupling adherens junctional complexes from the cytoskeleton. The TRIP6Stop473 mutant, which lacks the PDZ binding motif, was still able to increase NF-kappaB and Akt activities but did not promote invasiveness or interfere with cell-cell aggregation. Intracellular delivery of competing peptides corresponding to TRIP6 or beta-catenin C terminus restored invasive properties in MDCKts-src TRIP6Stop473 cells, highlighting the requirement of PDZ scaffolds in junctional complexes activity. TRIP6 overexpression in colon tumors suggest its critical role in cancer progression.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Transcription Factors/metabolism , ATPases Associated with Diverse Cellular Activities , Actins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Caco-2 Cells , Cadherins/metabolism , Cell Adhesion , Cell Adhesion Molecules , Cell Adhesion Molecules, Neuronal/genetics , Cell Line , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cytoskeleton/metabolism , Dogs , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic/physiology , Guanylate Kinases , HeLa Cells , Humans , LIM Domain Proteins , NF-kappa B/metabolism , Proteasome Endopeptidase Complex , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factors/genetics , Transfection , Two-Hybrid System Techniques , rho GTP-Binding Proteins/metabolism
3.
Int J Cancer ; 122(3): 572-82, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-17955485

ABSTRACT

Dynamic crosstalk between cell adhesion molecules, extracellular matrix and soluble informative factors is essential for cancer cell migration and invasion. Here, we investigated the mechanisms by which the E-cadherin/catenin complex and alpha v integrin can modulate insulin-like growth factor-I (IGF-I)-induced cell migration. Human colon mucosa, human colon cancer cell lines, HT29-D4 and HCT-8 derivatives that differ in their expression of alpha-catenin, were used as models. Interactions between E-cadherin, alpha v integrin and IGF-I receptor (IGF-IR) were analyzed by coimmunoprecipitation and immunolocalization experiments. The impact of these interactions on cell mobility was determined by haptotaxis assays. We report that alpha v integrin, E-cadherin and IGF-IR form a ternary complex in both cultured cancer cells and human normal colonic mucosa. alpha-Catenin regulates the scaffolding of this complex. IGF-IR ligation by IGF-I induces the disruption of the complex and the relocalization of alpha v integrin from cell-cell contacts to focal contact sites. This perturbation is correlated with the observed increase in cell migration. These results suggest that regulation of the alpha v integrin/E-cadherin/IGF-IR scaffolding is essential for the modulation of cell mobility. Its alteration could be of major importance to sustain alterations in cell adhesion that occur during cancer cell invasion and metastasis.


Subject(s)
Cadherins/metabolism , Integrin alphaV/metabolism , Receptor, IGF Type 1/metabolism , alpha Catenin/pharmacology , Cell Adhesion , Cell Movement , Flow Cytometry , Fluorescent Antibody Technique , HT29 Cells/metabolism , Humans , Immunoprecipitation , Insulin Receptor Substrate Proteins , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA, Small Interfering/pharmacology
4.
Int J Oncol ; 31(6): 1501-8, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17982677

ABSTRACT

Increased src tyrosine kinase expression and activity has been associated with colon cancer cell invasion and survival. Several signaling pathways are involved in the oncogenic activation of src during the adenoma to carcinoma progression and cellular invasion. In the present study, the synthetic ether lipid analog ET-18-OMe was shown to promote invasion of HCT-8/S11 colon cancer cells into collagen type I through the concomitant activation of src by phosphorylation at Tyr416 (5-30 min) in alpha1-integrin immunoprecipitates containing the integrin binding proteins talin and paxillin, as well as the phoshorylated and activated forms of focal adhesion kinase (FAK) at Tyr397 (a FAK kinase activation signal), Tyr576 and Tyr861. This was associated with the lateral redistribution of alpha1-integrins in focal aggregates and persistent activation of the p130Cas/JNK pathways at 5-30 min, with the subsequent induction and activation of the matrix metalloproteinases MMP-2 and MMP-9 (2-12 h). These activated molecular scaffolds and signaling cascades were not observed in immunoprecipitates of alpha2- and beta1-integrins, and tetraspanin CD9, an invasion and metastasis suppressor linked to integrins and FAK signaling. Our data demonstrate that the lateral redistribution and clustering of alpha1-integrins results in the recruitment of the FAK/src motility-promoting signaling complex involved in cancer cell invasion. Disruption of this proinvasive pathway was accomplished by the dominant negative mutant of src (K295R, kinase dead), src pharmacological inhibitor (PP1) and alpha1-integrin function blocking antibodies. These findings support the notion that the alpha1-integrin- and src-dependent signalosome is a relevant therapeutic target against tumor progression in colon cancer patients.


Subject(s)
Colonic Neoplasms/pathology , Crk-Associated Substrate Protein/physiology , Focal Adhesion Kinase 1/physiology , Integrin alpha1/physiology , JNK Mitogen-Activated Protein Kinases/physiology , MAP Kinase Signaling System/physiology , src-Family Kinases/physiology , Cell Line, Tumor , Enzyme Activation , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness , Phosphorylation
5.
Cancer Lett ; 255(1): 57-70, 2007 Sep 18.
Article in English | MEDLINE | ID: mdl-17493746

ABSTRACT

Gelsolin and CapG are both actin binding proteins that modulate a variety of physiological processes by interacting differently with the actin cytoskeleton. Several studies suggest that overexpression of these proteins promotes invasion in vitro. In this study we explored the contribution of these proteins in human cancer cell invasion and motility. We show that down regulation of CapG or gelsolin in several types of cancer cells, including MDA-MB 231 and PC-3 cells, significantly reduces the invasive and motile properties of cells, as well as cell aggregation. These results point to a role for CapG and gelsolin as tumor activator.


Subject(s)
Gelsolin/biosynthesis , Gene Expression Regulation, Neoplastic , Neoplasms/metabolism , Actins/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Collagen/chemistry , Collagen Type I/metabolism , Drug Combinations , Humans , In Vitro Techniques , Laminin/chemistry , Microfilament Proteins/metabolism , Neoplasm Invasiveness , Proteoglycans/chemistry , RNA Interference , Signal Transduction , Wound Healing
6.
Cell Signal ; 19(8): 1722-32, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17478078

ABSTRACT

Recent data indicate that the Bone Morphogenetic Protein BMP-7 exhibits mucosal protection against experimental colitis in rats, suggesting that this cytokine exerts direct actions in intestinal epithelial cells during inflammatory bowel diseases and other precancerous lesions of the colon. In this study, we investigated the functional expression of BMP-7 and its receptors in normal human colon crypts, aberrant crypt foci (ACF) in sigmoiditis and colorectal tumors, and their derived cancer cell lines. Transcripts encoding BMP-7 receptors type II (BMPRII, ActRII, ActRIIB) and type I (ALK-2) were clearly detected by RT-PCR in several premalignant and carcinoma cell lines. The cytokine was identified by immunohistochemistry in surface epithelial cells and crypts in the normal colon mucosa, ACF in sigmoiditis, sporadic high grade dysplastic adenoma, and in 9 of 16 colon carcinomas (56.2%). In addition, the conditioned medium collected from the adenoma PC/AA/C1 and carcinoma HCT8/S11 and SW48 cell lines in culture contained significant levels of BMP-7 ranging from 0.17 to 0.38 ng/ml. We found that BMP-7 induced scattering and proinvasive responses (EC50=1 ng/ml) in kidney and colon cancer cell lines through SMAD4 and src -independent pathways and signaling cascades using FAK phosphorylation at Y925 and activation of ERK1/2, Rac1 and JNK. This phosphorylation of FAK on Y925 was also induced by the proinvasive agent EGF. Taken together, our findings suggest that BMP-7 exerts divergent effects in the colon mucosa, one counteracting transient inflammatory situations and the other linked to pejorative functions during chronic ulcerative diseases and the neoplastic progression.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Colonic Neoplasms/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Smad4 Protein/metabolism , Transforming Growth Factor beta/metabolism , rac GTP-Binding Proteins/metabolism , Bone Morphogenetic Protein 7 , Cell Line, Tumor , Colonic Neoplasms/enzymology , Colonic Neoplasms/metabolism , Enzyme-Linked Immunosorbent Assay , HT29 Cells , Humans , Immunohistochemistry , Retrospective Studies
7.
Exp Cell Res ; 313(9): 1790-804, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17451681

ABSTRACT

Syntenin is a tandem PDZ protein that has recently been shown to be overexpressed in several cancer cells and tissues, and that might play an active role in tumor cell invasion and metastasis. Here we show that overexpression of the tandem PDZ domains of syntenin in non-invasive cells is necessary and sufficient to stimulate these cells to invade a collagen I matrix, and this effect can be regulated by ligand binding to the PDZ domains. Furthermore, we show that syntenin-induced invasion requires signaling through ras, rho and PI3K/MAPK signaling pathways and involves changes in cell-cell adhesion. Inversely, when we used RNA interference to inhibit syntenin expression in different invasive cancer cell lines, we observed a drastically decreased ability of these cells to migrate and invade into collagen type I or Matrigel. RNAi-treated cells also show increased cell aggregation, indicating that syntenin is important for cell-cell adhesion in epithelial cells. Together, these results suggest that downregulation of syntenin by RNA interference could provide a means of inhibiting tumor invasion and possibly metastasis in different cancers, and point to syntenin as a potential cancer biomarker and drug target.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Movement/genetics , Neoplasm Invasiveness/genetics , Neoplasms/physiopathology , Syntenins/metabolism , Animals , Basement Membrane/metabolism , Biomarkers, Tumor/genetics , Cell Adhesion/physiology , Cell Aggregation/physiology , Cell Line, Tumor , Collagen/metabolism , Down-Regulation/physiology , Humans , MAP Kinase Signaling System/physiology , Neoplasms/genetics , Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Binding/physiology , Protein Structure, Tertiary/physiology , RNA Interference/physiology , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Rats , Subcellular Fractions/metabolism , Syntenins/chemistry , Syntenins/genetics , rho GTP-Binding Proteins/metabolism
8.
Cancer Res ; 66(20): 10007-15, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17047063

ABSTRACT

Down-regulation of the epithelial cell-cell adhesion molecule E-cadherin is frequently associated with tumor formation and progression. Besides its role in physical cell-cell adhesion, E-cadherin is also thought to be involved in intracellular signaling in normal epithelial cells. In these cells, the Armadillo catenin p120ctn binds to the cytoplasmic domain of E-cadherin and stabilizes the adhesion complexes. On loss of E-cadherin, cytoplasmic p120ctn might accumulate and contribute to tumor malignancy. We used suppression subtractive hybridization to search for genes regulated by E-cadherin expression. We isolated human Nanos1 as a transcript of which levels decrease on E-cadherin reexpression in a human breast cancer cell line. The hNanos1 protein bears a COOH-terminal (CCHC)(2) zinc finger domain and belongs to an evolutionarily conserved protein family sharing functions in germ cell development in both vertebrates and invertebrates. We found an inverse correlation between E-cadherin and hNanos1 expression in various cell lines and under diverse conditions. Conditional expression of hNanos1 in human colorectal DLD1 cancer cells functionally abolished cell-cell adhesion. It induced cytoplasmic translocation of p120ctn, as well as strong migratory and invasive properties. We also found that the NH(2)-terminal domain of hNanos1, which is conserved only among mammals, interacts with p120ctn. hNanos1 counteracted the stimulatory effect of p120ctn on cell protrusion formation. Together, these findings describe a new function for hNanos1 as a downstream effector of E-cadherin loss contributing to tumor progression. Targeting hNanos1 might be a promising strategy in the treatment of E-cadherin-negative tumors in particular.


Subject(s)
Cadherins/genetics , Cell Adhesion Molecules/metabolism , Cell Movement/physiology , Phosphoproteins/metabolism , RNA-Binding Proteins/genetics , Animals , Armadillos , Cadherins/biosynthesis , Cadherins/metabolism , Catenins , Cell Adhesion/physiology , Cell Adhesion Molecules/biosynthesis , Cell Line, Tumor , Cloning, Molecular , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Phosphoproteins/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/metabolism , Transcription, Genetic , Zinc Fingers , Delta Catenin
9.
Cancer Res ; 66(18): 9227-34, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16982767

ABSTRACT

A large number of hormones and local agonists activating guanine-binding protein-coupled receptors (GPCR) play a major role in cancer progression. Here, we characterize the new imidazo-pyrazine derivative BIM-46174, which acts as a selective inhibitor of heterotrimeric G-protein complex. BIM-46174 prevents the heterotrimeric G-protein signaling linked to several GPCRs mediating (a) cyclic AMP generation (Galphas), (b) calcium release (Galphaq), and (c) cancer cell invasion by Wnt-2 frizzled receptors and high-affinity neurotensin receptors (Galphao/i and Galphaq). BIM-46174 inhibits the growth of a large panel of human cancer cell lines, including anticancer drug-resistant cells. Exposure of cancer cells to BIM-46174 leads to caspase-3-dependent apoptosis and poly(ADP-ribose) polymerase cleavage. National Cancer Institute COMPARE analysis for BIM-46174 supports its novel pharmacologic profile compared with 12,000 anticancer agents. The growth rate of human tumor xenografts in athymic mice is significantly reduced after administration of BIM-46174 combined with either cisplatin, farnesyltransferase inhibitor, or topoisomerase inhibitors. Our data validate the feasibility of targeting heterotrimeric G-protein functions downstream the GPCRs to improve anticancer chemotherapy.


Subject(s)
Cysteine/analogs & derivatives , Heterotrimeric GTP-Binding Proteins/antagonists & inhibitors , Imidazoles/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Cysteine/pharmacology , Drug Screening Assays, Antitumor , Female , GTP-Binding Protein alpha Subunits/antagonists & inhibitors , GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein beta Subunits/antagonists & inhibitors , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/antagonists & inhibitors , GTP-Binding Protein gamma Subunits/metabolism , HL-60 Cells , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Mice , Neoplasm Invasiveness , Xenograft Model Antitumor Assays
10.
Mol Cancer Ther ; 5(8): 2070-7, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16928828

ABSTRACT

We recently showed by DNA microarray analysis that vascular endothelial growth factor (VEGF) receptor (VEGFR) is expressed in HCT8/S11 human colon cancer cells, suggesting that several angiogenic factors may target colon cancer cells themselves. In this study, transcripts encoding the VEGF-165 and semaphorin 3A (Sema3A) receptors and coreceptors Flt-1, KDR/Flk-1, plexin A1, and neuropilins NP-1 and NP-2 were identified by reverse transcription-PCR in the human colon cancer cell lines HCT8/S11, HT29, HCT116, and PCmsrc. Collagen invasion induced by VEGF-165 and Sema3A in HCT8/S11 cells (EC(50), 0.4-1 nmol/L) required p42/44 mitogen-activated protein kinase and signaling through RhoA/Rho-kinase-dependent and -independent pathways, respectively. As expected, the VEGFR signaling inhibitor ZD4190 selectively abrogated the proinvasive activity of VEGF in collagen gels (IC(50), 10 nmol/L) and chick heart fragments. We identify a novel function for VEGF-165 and Sema3A as proinvasive factors for human colorectal cancer cells. Interestingly, oral administration of the single drug ZD4190 to athymic mice (50 mg/kg/d, once daily) inhibited by 70% the growth of HCT8/S11 tumor cell xenografts. Combinations between the src tyrosine kinase inhibitor M475271 and ZD4190 or cisplatin resulted in additive therapeutic activity against LNM35 human lung tumor xenografts. Our data have significant implications for new therapeutic approaches and individualized treatment targeting VEGFR and src signaling pathways in combination with established clinical drugs at primary tumors and distant metastases in colon and lung cancer patients.


Subject(s)
Colonic Neoplasms/drug therapy , Quinazolines/pharmacology , Semaphorin-3A/metabolism , Triazoles/pharmacology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Cisplatin/pharmacology , Colonic Neoplasms/pathology , Enzyme Inhibitors/pharmacology , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Mice, Nude , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Invasiveness , Piperidines/pharmacology , Receptors, Vascular Endothelial Growth Factor/drug effects , Receptors, Vascular Endothelial Growth Factor/metabolism , Semaphorin-3A/drug effects , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
11.
Cancer Res ; 66(12): 6243-9, 2006 Jun 15.
Article in English | MEDLINE | ID: mdl-16778199

ABSTRACT

Emerging evidence supports neurotensin as a trophic and antiapoptotic factor, mediating its control via the high-affinity neurotensin receptor (NT1 receptor) in several human solid tumors. In a series of 51 patients with invasive ductal breast cancers, 34% of all tumors were positive for neurotensin and 91% positive for NT1 receptor. We found a coexpression of neurotensin and NT1 receptor in a large proportion (30%) of ductal breast tumors, suggesting a contribution of the neurotensinergic signaling cascade within breast cancer progression. Functionally expressed NT1 receptor, in the highly malignant MDA-MB-231 human breast cancer cell line, coordinated a series of transforming functions, including cellular migration, invasion, induction of the matrix metalloproteinase (MMP)-9 transcripts, and MMP-9 gelatinase activity. Disruption of NT1 receptor signaling by silencing RNA or use of a specific NT1 receptor antagonist, SR48692, caused the reversion of these transforming functions and tumor growth of MDA-MB-231 cells xenografted in nude mice. Our findings support the contribution of neurotensin in human breast cancer progression and point out the utility to develop therapeutic molecules targeting neurotensin or NT1 receptor signaling cascade. These strategies would increase the range of therapeutic approaches and be beneficial for specific patients.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Neurotensin/biosynthesis , Receptors, Neurotensin/biosynthesis , Animals , Carcinoma in Situ/metabolism , Carcinoma in Situ/pathology , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement/physiology , Disease Progression , Enzyme Activation , Female , Humans , Immunohistochemistry , Matrix Metalloproteinase 9/metabolism , Mice , Middle Aged , Neoplasm Invasiveness , Transplantation, Heterologous
12.
J Cell Sci ; 119(Pt 9): 1947-60, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16636079

ABSTRACT

L-plastin, a malignant transformation-associated protein, is a member of a large family of actin filament cross-linkers. Here, we analysed how phosphorylation of L-plastin on Ser5 of the headpiece domain regulates its intracellular distribution and its interaction with F-actin in transfected cells and in in vitro assays. Phosphorylated wild-type L-plastin localised to the actin cytoskeleton in transfected Vero cells. Ser5Ala substitution reduced the capacity of L-plastin to localise with peripheral actin-rich membrane protrusions. Conversely, a Ser5Glu variant mimicking a constitutively phosphorylated state, accumulated in actin-rich regions and promoted the formation of F-actin microspikes in two cell lines. Similar to phosphorylated wild-type L-plastin, this variant remained associated with cellular F-actin in detergent-treated cells, whereas the Ser5Ala variant was almost completely extracted. When compared with non-phosphorylated protein, phosphorylated L-plastin and the Ser5Glu variant bound F-actin more efficiently in an in vitro assay. Importantly, expression of L-plastin elicited collagen invasion in HEK293T cells, in a manner dependent on Ser5 phosphorylation. Based on our findings, we propose that conversely to other calponin homology (CH)-domain family members, phosphorylation of L-plastin switches the protein from a low-activity to a high-activity state. Phosphorylated L-plastin might act as an integrator of signals controlling the assembly of the actin cytoskeleton and cell motility in a 3D-space.


Subject(s)
Actins/metabolism , Microfilament Proteins/metabolism , Serine/metabolism , Actins/chemistry , Amino Acid Sequence , Animals , Cell Line , Chlorocebus aethiops , Cytoskeleton/metabolism , Humans , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Protein Binding
13.
Carcinogenesis ; 27(4): 708-16, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16299383

ABSTRACT

Alterations in the Wnt/APC (adenomatous polyposis coli) signalling pathway, resulting in beta-catenin/T cell factor (Tcf)-dependent transcriptional gene activation, are frequently detected in familial and sporadic colon cancers. The neuropeptide neurotensin (NT) is widely distributed in the gastrointestinal tract. Its proliferative and survival effects are mediated by a G-protein coupled receptor, the NT1 receptor. NT1 receptor is not expressed in normal colon epithelial cells, but is over expressed in a number of cancer cells and tissues suggesting a link to the outgrowth of human colon cancer. Our results demonstrate that the upregulation of NT1 receptor occurring in colon cancer is the result of Wnt/APC signalling pathway activation. We first established the functionality of the Tcf response element within the NT1 receptor promoter. Consequently, we observed the activation of NT1 receptor gene by agents causing beta-catenin cytosolic accumulation, as well as a strong decline of endogenous receptor when wt-APC was restored. At the cellular level, the re-establishment of wt-APC phenotype resulted in the impaired functionality of NT1 receptor, like the breakdown in NT-induced intracellular calcium mobilization and the loss of NT pro-invasive effect. We corroborated the Wnt/APC signalling pathway on the NT1 receptor promoter activation with human colon carcinogenesis, and showed that NT1 receptor gene activation was perfectly correlated with nuclear or cytoplasmic beta-catenin localization while NT1 receptor was absent when beta-catenin was localized at the cell-cell junction in early adenomas of patients with familial adenomatous polyposis, hereditary non-polyposis colorectal cancer and loss of heterozygosity tumours. In this report we establish a novel link in vitro between the Tcf/beta-catenin pathway and NT1 receptor promoter activation.


Subject(s)
Adenoma/genetics , Colonic Neoplasms/genetics , Receptors, Neurotensin/biosynthesis , TCF Transcription Factors/physiology , beta Catenin/physiology , Adenoma/physiopathology , Adenomatous Polyposis Coli Protein/physiology , Cell Proliferation , Cell Survival , Cell Transformation, Neoplastic , Colonic Neoplasms/physiopathology , Humans , Loss of Heterozygosity , Promoter Regions, Genetic , Receptors, Neurotensin/physiology , Signal Transduction , Up-Regulation , Wnt Proteins/physiology
14.
Nucleic Acids Res ; 33(20): 6566-78, 2005.
Article in English | MEDLINE | ID: mdl-16314317

ABSTRACT

SIP1/ZEB2 is a member of the deltaEF-1 family of two-handed zinc finger nuclear factors. The expression of these transcription factors is associated with epithelial mesenchymal transitions (EMT) during development. SIP1 is also expressed in some breast cancer cell lines and was detected in intestinal gastric carcinomas, where its expression is inversely correlated with that of E-cadherin. Here, we show that expression of SIP1 in human epithelial cells results in a clear morphological change from an epithelial to a mesenchymal phenotype. Induction of this epithelial dedifferentiation was accompanied by repression of several cell junctional proteins, with concomitant repression of their mRNA levels. Besides E-cadherin, other genes coding for crucial proteins of tight junctions, desmosomes and gap junctions were found to be transcriptionally regulated by the transcriptional repressor SIP1. Moreover, study of the promoter regions of selected genes by luciferase reporter assays and chromatin immunoprecipitation shows that repression is directly mediated by SIP1. These data indicate that, during epithelial dedifferentiation, SIP1 represses in a coordinated manner the transcription of genes coding for junctional proteins contributing to the dedifferentiated state; this repression occurs by a general mechanism mediated by Smad Interacting Protein 1 (SIP1)-binding sites.


Subject(s)
Epithelial Cells/metabolism , Homeodomain Proteins/physiology , Intercellular Junctions/metabolism , Membrane Proteins/genetics , Repressor Proteins/physiology , Binding Sites , Cadherins/genetics , Cadherins/metabolism , Cell Line , Cell Line, Tumor , Chromatin/metabolism , Connexins/genetics , Connexins/metabolism , Down-Regulation , Epithelial Cells/cytology , Humans , Membrane Proteins/metabolism , Mesoderm/cytology , Mutation , Promoter Regions, Genetic , RNA, Messenger/metabolism
15.
J Dairy Res ; 72 Spec No: 20-6, 2005.
Article in English | MEDLINE | ID: mdl-16180717

ABSTRACT

To define better the function of Nerve Growth Factor (NGF) in breast cancer progression, we investigated whether this polypeptide was able to induce breast cancer cell invasion. NGF inhibited aggregation of tumour cells through modulation of the E-cadherin/catenin complex function. In addition, NGF induced the breast cancer cells to invade into Matrigel. We focused our attention on how NGF prevents aggregation, in order to discover the signalling pathway that leads tumour cells to acquire the invasive phenotype. Moreover, studies on the identification of signalling pathways that are responsive for NGF-induced invasion will be basically described.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cadherins/metabolism , Nerve Growth Factor/pharmacology , Cadherins/analysis , Cadherins/genetics , Cell Line, Tumor , Collagen , Drug Combinations , Fluorescent Antibody Technique , Humans , Laminin , Microscopy, Fluorescence , Neoplasm Invasiveness , Proteoglycans , RNA/analysis , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
16.
Oncogene ; 24(56): 8240-51, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16091733

ABSTRACT

We recently reported that proteinase-activated receptors type I (PAR-1) are coupled to both negative and positive invasion pathways in colonic and kidney cancer cells cultured on collagen type I gels. Here, we found that treatments with the cell-permeant analog 8-Br-cGMP and the soluble guanylate cyclase activator BAY41-2272, and Rho kinase (ROK) inhibition by Y27632 or a dominant negative form of ROK lead to PAR-1-mediated invasion through differential Rac1 and Cdc42 signaling. Hypoxia or the counteradhesive matricellular protein SPARC/BM-40 (SPARC: secreted protein acidic rich in cysteine) overexpressed during cancer progression also commutated PAR-1 to cellular invasion through the cGMP/protein kinase G (PKG) cascade, RhoA inactivation, and Rac1-dependent or -independent signaling. Cultured primary cancer cells isolated from peritoneal and pleural effusions from patients with colon cancer or other malignant tumors harbored PAR-1, as shown by RT-PCR and FACS analyses. These malignant effusions also contained high levels of activated thrombin and fibrin, and induced a proinvasive response in HCT8/S11 human colorectal cancer cells. Our data underline the essential role of the tumor microenvironment and of several commutators targeting cGMP/PKG signaling and the RhoA-ROK axis in the control of PAR-1 proinvasive activity and metastatic potential of cancer cells in distant organs and peritoneal or pleural cavities. We also add new insights into the mechanisms linking the coagulation mediators thrombin and PAR-1 in the context of blood coagulation disorders and venous thrombosis often observed in cancer patients, as described in 1865 by Armand Trousseau.


Subject(s)
Neoplasm Invasiveness/pathology , Neoplasms/enzymology , Neoplasms/pathology , Protein Serine-Threonine Kinases/physiology , Receptor, PAR-1/physiology , Signal Transduction/physiology , rho GTP-Binding Proteins/metabolism , Aged , Aged, 80 and over , Animals , Cyclic GMP/physiology , Dogs , G-Protein-Coupled Receptor Kinase 1/antagonists & inhibitors , G-Protein-Coupled Receptor Kinase 1/physiology , Guanylate Cyclase , HCT116 Cells , HT29 Cells , Humans , Hypoxia/enzymology , Hypoxia/metabolism , Intracellular Signaling Peptides and Proteins , Middle Aged , Neoplasms/metabolism , Osteonectin/physiology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor, PAR-1/genetics , Receptors, Antigen/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/drug effects , Soluble Guanylyl Cyclase , Thrombin/metabolism , rho-Associated Kinases , rhoA GTP-Binding Protein/antagonists & inhibitors , rhoA GTP-Binding Protein/physiology
17.
Cancer Res ; 65(14): 6237-44, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-16024625

ABSTRACT

Abberant activation of the process of epithelial-mesenchymal transition in cancer cells is a late event in tumor progression. A key inducer of this transition is the transcription factor Snail, which represses E-cadherin. We report that conditional expression of the human transcriptional repressor Snail in colorectal cancer cells induces an epithelial dedifferentiation program that coincides with a drastic change in cell morphology. Snail target genes control the establishment of several junctional complexes, intermediate filament networks, and the actin cytoskeleton. Modulation of the expression of these genes is associated with loss of cell aggregation and induction of invasion. Chromatin immunoprecipitation experiments showed that repression of selected target genes is associated with increased binding of Snail to their promoters, which contain consensus Snail-binding sites. Thus, Snail constitutes a master switch that directly represses the epithelial phenotype, resulting in malignant carcinoma cells.


Subject(s)
Colorectal Neoplasms/pathology , Transcription Factors/physiology , Actins/genetics , Actins/metabolism , Cell Aggregation/physiology , Cell Differentiation/physiology , Cell Line, Tumor , Chromatin Immunoprecipitation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Cytoskeleton/genetics , Cytoskeleton/metabolism , Epithelial Cells/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness , Signal Transduction , Snail Family Transcription Factors , Transcription Factors/biosynthesis , Transcription Factors/genetics
18.
Traffic ; 6(4): 335-45, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15752138

ABSTRACT

T- and L-plastin are highly similar actin-bundling proteins implicated in the regulation of cell morphology, lamellipodium protrusion, bacterial invasion and tumor progression. We show that T-plastin localizes predominantly to the cytoplasm, whereas L-plastin distributes between nucleus and cytoplasm in HeLa or Cos cells. T-plastin shows nuclear accumulation upon incubation of cells with the CRM1 antagonist leptomycin B (LMB). We identified a Rev-like nuclear export sequence (NES) in T-plastin that is able to export an otherwise nuclear protein in an LMB-dependent manner. Deletion of the NES promotes nuclear accumulation of T-plastin. Mutation of residues L17, F21 or L26 in the T-plastin NES inhibits nuclear efflux. L-plastin harbors a less conserved NES and lacks the F21 T-plastin residue. Insertion of a Phe residue in the L-plastin NES specifically enhances its export activity. These findings explain why both isoforms exhibit specific distribution patterns in eukaryotic cells.


Subject(s)
Microfilament Proteins/metabolism , Phosphoproteins/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Chlorocebus aethiops , Fatty Acids, Unsaturated/pharmacology , Gene Products, rev/genetics , Gene Products, rev/metabolism , Humans , Leucine/genetics , Leucine/metabolism , Membrane Glycoproteins , Molecular Sequence Data , Phenotype , Phenylalanine/genetics , Phenylalanine/metabolism , Phosphoproteins/genetics , Protein Isoforms/metabolism , Protein Sorting Signals/drug effects , Sequence Alignment
19.
Oncogene ; 24(20): 3274-84, 2005 May 05.
Article in English | MEDLINE | ID: mdl-15735708

ABSTRACT

During a search to identify resveratrol (3,5,4'-trihydroxy-trans-stilbene, RV) target genes in the human erythroleukemic K562 cell line, we show here that the tensin gene and protein levels are remarkably induced by this dietary polyphenol. Tensin, a cell-matrix adhesion protein binding the integrins and cytoskeletal actin filaments also interacts with PI3-kinase and JNK signaling pathways. Tensin induction by RV is associated with increased K562 cell adhesion to fibronectin, cell spreading and actin polymerization. The same responses were observed in the tensin-deficient MCF7 human breast cancer cell line. In K562 and MCF7 cells treated by RV, tensin was found in punctate and intracytoplasmic areas. In MCF7 epithelial cells, induction of tensin is not exclusively associated with plasma membrane-bound vinculin, suggesting a dual localization of tensin in both focal and fibrillar adhesions. Pharmacological blockade of PI3-kinase and Rho GTPases/Rho-kinase resulted in selective depletion of focal adhesions, disorganization of tensin localization and disruption of stress fibers. RV increased cell motility and attachment to fibronectin in MCF7 cells submitted to mechanical laminar flow stress, and abrogated estrogen-induced MCF7 cancer cell invasion. Our data support the conclusion that induction of tensin by RV contributes to the chemopreventive and anti-invasive activity of this natural dietary compound in tensin-negative and -deficient leukemic cells or epithelioid cancers.


Subject(s)
Anticarcinogenic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Microfilament Proteins/biosynthesis , Neoplasms/prevention & control , Stilbenes/pharmacology , Actins/metabolism , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Movement , Cycloheximide/pharmacology , Cytoplasm/metabolism , Cytoskeleton/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Immunoblotting , K562 Cells , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/metabolism , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/metabolism , Resveratrol , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tensins , Time Factors
20.
Cancer Res ; 65(1): 195-202, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15665295

ABSTRACT

Signal transducer and activator of transcription (STAT) 3 is overexpressed or activated in most types of human tumors and has been classified as an oncogene. In the present study, we investigated the contribution of the STAT3s to the proinvasive activity of trefoil factors (TFF) and vascular endothelial growth factor (VEGF) in human colorectal cancer cells HCT8/S11 expressing VEGF receptors. Both intestinal trefoil peptide (TFF3) and VEGF, but not pS2 (TFF1), activate STAT3 signaling through Tyr(705) phosphorylation of both STAT3alpha and STAT3beta isoforms. Blockade of STAT3 signaling by STAT3beta, depletion of the STAT3alpha/beta isoforms by RNA interference, and pharmacologic inhibition of STAT3alpha/beta phosphorylation by cucurbitacin or STAT3 inhibitory peptide abrogates TFF- and VEGF-induced cellular invasion and reduces the growth of HCT8/S11 tumor xenografts in athymic mice. Differential gene expression analysis using DNA microarrays revealed that overexpression of STAT3beta down-regulates the VEGF receptors Flt-1, neuropilins 1 and 2, and the inhibitor of DNA binding/differentiation (Id-2) gene product involved in the neoplastic transformation. Taken together, our data suggest that TFF3 and the essential tumor angiogenesis regulator VEGF(165) exert potent proinvasive activity through STAT3 signaling in human colorectal cancer cells. We also validate new therapeutic strategies targeting STAT3 signaling by pharmacologic inhibitors and RNA interference for the treatment of colorectal cancer patients.


Subject(s)
Colonic Neoplasms/pathology , DNA-Binding Proteins/physiology , Mucins/physiology , Muscle Proteins/physiology , Trans-Activators/physiology , Vascular Endothelial Growth Factor A/physiology , Apoptosis , Base Sequence , Cell Division , Cell Line, Tumor , DNA Primers , Humans , Kinetics , Neoplasm Invasiveness , Peptides , Protein Isoforms/physiology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor , Signal Transduction/physiology , Trefoil Factor-3
SELECTION OF CITATIONS
SEARCH DETAIL
...