Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
EMBO Rep ; 24(4): e54895, 2023 04 05.
Article in English | MEDLINE | ID: mdl-36704936

ABSTRACT

Colon tumors of the mesenchymal subtype have the lowest overall survival. Snail1 is essential for the acquisition of this phenotype, characterized by increased tumor stemness and invasion, and high resistance to chemotherapy. Here, we find that Snail1 expression in colon tumor cells is dependent on an autocrine noncanonical Wnt pathway. Accordingly, depletion of Ror2, the co-receptor for noncanonical Wnts such as Wnt5a, potently decreases Snail1 expression. Wnt5a, Ror2, and Snail1 participate in a self-stimulatory feedback loop since Wnt5a increases its own synthesis in a Ror2- and Snail1-dependent fashion. This Wnt5a/Ror2/Snail1 axis controls tumor invasion, chemoresistance, and formation of tumor spheres. It also stimulates TGFß synthesis; consequently, tumor cells expressing Snail1 are more efficient in activating cancer-associated fibroblasts than the corresponding controls. Ror2 downmodulation or inhibition of the Wnt5a pathway decreases Snail1 expression in primary colon tumor cells and their ability to form tumors and liver metastases. Finally, the expression of SNAI1, ROR2, and WNT5A correlates in human colon and other tumors. These results identify inhibition of the noncanonical Wnt pathway as a putative colon tumor therapy.


Subject(s)
Colonic Neoplasms , Wnt Signaling Pathway , Humans , Drug Resistance, Neoplasm/genetics , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Fibroblasts
2.
Cancer Res ; 81(2): 438-451, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33229340

ABSTRACT

Tumors are complex tissues composed of transformed epithelial cells as well as cancer-activated fibroblasts (CAF) that facilitate epithelial tumor cell invasion. We show here that CAFs and other mesenchymal cells rely much more on glutamine than epithelial tumor cells; consequently, they are more sensitive to inhibition of glutaminase. Glutamine dependence drove CAF migration toward this amino acid when cultured in low glutamine conditions. CAFs also invaded a Matrigel matrix following a glutamine concentration gradient and enhanced the invasion of tumor cells when both cells were cocultured. Accordingly, glutamine directed invasion of xenografted tumors in immunocompromised mice. Stimulation of glutamine-driven epithelial tumor invasion by fibroblasts required previous CAF activation, which involved the TGFß/Snail1 signaling axis. CAFs moving toward Gln presented a polarized Akt2 distribution that was modulated by the Gln-dependent activity of TRAF6 and p62 in the migrating front, and depletion of these proteins prevented Akt2 polarization and Gln-driven CAF invasion. Our results demonstrate that glutamine deprivation promotes CAF migration and invasion, which in turn facilitates the movement of tumor epithelial cells toward nutrient-rich territories. These results provide a novel molecular mechanism for how metabolic stress enhances invasion and metastasis. SIGNIFICANCE: Cancer-associated fibroblasts migrate and invade toward free glutamine and facilitate invasion of tumor epithelial cells, accounting for their movement away from the hostile conditions of the tumor towards nutrient-rich adjacent tissues. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/2/438/F1.large.jpg.


Subject(s)
Breast Neoplasms/pathology , Cancer-Associated Fibroblasts/pathology , Cell Movement , Epithelial-Mesenchymal Transition , Glutamine/pharmacology , Neoplasms, Glandular and Epithelial/pathology , Animals , Apoptosis , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Cell Proliferation , Female , Humans , Mice , Mice, Nude , Neoplasms, Glandular and Epithelial/drug therapy , Neoplasms, Glandular and Epithelial/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Cell Mol Life Sci ; 77(5): 919-935, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31312879

ABSTRACT

Wnt ligands signal through canonical or non-canonical signaling pathways. Although both routes share common elements, such as the Fz2 receptor, they differ in the co-receptor and in many of the final responses; for instance, whereas canonical Wnts increase ß-catenin stability, non-canonical ligands downregulate it. However, both types of ligands stimulate tumor cell invasion. We show here that both the canonical Wnt3a and the non-canonical Wnt5a stimulate Fz2 tyrosine phosphorylation, Fyn binding to Fz2, Fyn activation and Fyn-dependent Stat3 phosphorylation. Wnt3a and Wnt5a require Src for Fz2 tyrosine phosphorylation; Src binds to canonical and non-canonical co-receptors (LRP5/6 and Ror2, respectively) and is activated by Wnt3a and Wnt5a. This Fz2/Fyn/Stat3 branch is incompatible with the classical Fz2/Dvl2 pathway as shown by experiments of over-expression or depletion. Fyn is necessary for transcription of genes associated with invasiveness, such as Snail1, and for activation of cell invasion by both Wnt ligands. Our results extend the knowledge about canonical Wnt pathways, demonstrating additional roles for Fyn in this pathway and describing how this protein kinase is activated by both canonical and non-canonical Wnts.


Subject(s)
Frizzled Receptors/metabolism , Proto-Oncogene Proteins c-fyn/metabolism , Wnt-5a Protein/metabolism , Wnt3A Protein/metabolism , src-Family Kinases/metabolism , Cell Line , Enzyme Activation/genetics , HEK293 Cells , Humans , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Neoplasm Invasiveness/genetics , Neoplasms/pathology , Phosphorylation/physiology , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , STAT3 Transcription Factor/metabolism , Transcription, Genetic/genetics , Wnt Signaling Pathway/physiology , beta Catenin/metabolism
4.
Cells ; 8(10)2019 09 25.
Article in English | MEDLINE | ID: mdl-31557964

ABSTRACT

In contrast to non-canonical ligands, canonical Wnts promote the stabilization of ß-catenin, which is a prerequisite for formation of the TCF4/ß-catenin transcriptional complex and activation of its target genes. This pathway is initiated by binding of Wnt ligands to the Frizzled/LRP5/6 receptor complex, and it increases the half-life of ß-catenin by precluding the phosphorylation of ß-catenin by GSK3 and its binding to the ßTrCP1 ubiquitin ligase. Other intercellular signals are also activated by Wnt ligands that do not inhibit GSK3 and increase ß-catenin protein but that either facilitate ß-catenin transcriptional activity or stimulate other transcriptional factors that cooperate with it. In this review, we describe the layers of complexity of these signals and discuss their crosstalk with ß-catenin in activation of transcriptional targets.


Subject(s)
Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Ligands , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/physiology , beta Catenin/metabolism , Animals , Glycogen Synthase Kinase 3/metabolism , Humans , Protein Stability , Receptor Cross-Talk/drug effects , Receptor Cross-Talk/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Transcriptional Activation/drug effects , Transcriptional Activation/physiology
5.
Mol Oncol ; 12(5): 611-629, 2018 05.
Article in English | MEDLINE | ID: mdl-29465811

ABSTRACT

Canonical and noncanonical Wnt pathways share some common elements but differ in the responses they evoke. Similar to Wnt ligands acting through the canonical pathway, Wnts that activate the noncanonical signaling, such as Wnt5a, promote Disheveled (Dvl) phosphorylation and its binding to the Frizzled (Fz) Wnt receptor complex. The protein kinase CK1ε is required for Dvl/Fz association in both canonical and noncanonical signaling. Here we show that differently to its binding to canonical Wnt receptor complex, CK1ε does not require p120-catenin for the association with the Wnt5a co-receptor Ror2. Wnt5a promotes the formation of the Ror2-Fz complex and enables the activation of Ror2-bound CK1ε by Fz-associated protein phosphatase 2A. Moreover, CK1ε also regulates Ror2 protein levels; CK1ε association stabilizes Ror2, which undergoes lysosomal-dependent degradation in the absence of this kinase. Although p120-catenin is not required for CK1ε association with Ror2, it also participates in this signaling pathway as p120-catenin binds and maintains Ror2 at the plasma membrane; in p120-depleted cells, Ror2 is rapidly internalized through a clathrin-dependent mechanism. Accordingly, downregulation of p120-catenin or CK1ε affects late responses to Wnt5a that are also sensitive to Ror2, such as SIAH2 transcription, cell invasion, or cortical actin polarization. Our results explain how CK1ε is activated by noncanonical Wnt and identify p120-catenin and CK1ε as two critical factors controlling Ror2 function.


Subject(s)
Casein Kinases/metabolism , Catenins/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Wnt Signaling Pathway , Animals , Endocytosis , HEK293 Cells , Humans , Ligands , Lysosomes/metabolism , Mice , Models, Biological , Phosphorylation , Protein Binding , Delta Catenin
6.
Mol Biol Cell ; 28(25): 3699-3708, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29021340

ABSTRACT

The ß4 isoform of the ß-subunits of voltage-gated calcium channel regulates cell proliferation and cell cycle progression. Herein we show that coexpression of the ß4-subunit with actors of the canonical Wnt/ß-catenin signaling pathway in a hepatoma cell line inhibits Wnt-responsive gene transcription and decreases cell division, in agreement with the role of the Wnt pathway in cell proliferation. ß4-subunit-mediated inhibition of Wnt signaling is observed in the presence of LiCl, an inhibitor of glycogen synthase kinase (GSK3) that promotes ß-catenin translocation to the nucleus. Expression of ß4-subunit mutants that lost the ability to translocate to the nucleus has no effect on Wnt signaling, suggesting that ß4-subunit inhibition of Wnt signaling occurs downstream from GSK3 and requires targeting of ß4-subunit to the nucleus. ß4-subunit coimmunoprecipitates with the TCF4 transcription factor and overexpression of TCF4 reverses the effect of ß4-subunit on the Wnt pathway. We thus propose that the interaction of nuclear ß4-subunit with TCF4 prevents ß-catenin binding to TCF4 and leads to the inhibition of the Wnt-responsive gene transcription. Thereby, our results show that ß4-subunit is a TCF4 repressor and therefore appears as an interesting candidate for the regulation of this pathway in neurons where ß4-subunit is specifically expressed.


Subject(s)
Calcium Channels/metabolism , Glycogen Synthase Kinase 3/metabolism , Wnt Signaling Pathway , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , CHO Cells , Calcium Channels/physiology , Cell Line , Cell Line, Tumor , Cell Proliferation/physiology , Cricetulus , Down-Regulation , Humans , Promoter Regions, Genetic , Protein Binding , Signal Transduction , Transcription Factor 4/metabolism , Transcription Factors/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , beta Catenin/physiology
7.
Crit Rev Biochem Mol Biol ; 52(3): 327-339, 2017 06.
Article in English | MEDLINE | ID: mdl-28276699

ABSTRACT

Canonical Wnt signaling controls ß-catenin protein stabilization, its translocation to the nucleus and the activation of ß-catenin/Tcf-4-dependent transcription. In this review, we revise and discuss the recent results describing actions of p120-catenin in different phases of this pathway. More specifically, we comment its involvement in four different steps: (i) the very early activation of CK1ɛ, essential for Dvl-2 binding to the Wnt receptor complex; (ii) the internalization of GSK3 and Axin into multivesicular bodies, necessary for a complete stabilization of ß-catenin; (iii) the activation of Rac1 small GTPase, required for ß-catenin translocation to the nucleus; and (iv) the release of the inhibitory action caused by Kaiso transcriptional repressor. We integrate these new results with the previously known action of other elements in this pathway, giving a particular relevance to the responses of the Wnt pathway not required for ß-catenin stabilization but for ß-catenin transcriptional activity. Moreover, we discuss the possible future implications, suggesting that the two cellular compartments where ß-catenin is localized, thus, the adherens junction complex and the Wnt signalosome, are more physically connected that previously thought.


Subject(s)
Catenins/metabolism , Receptors, Wnt/metabolism , Transcription, Genetic/physiology , Wnt Signaling Pathway/physiology , Animals , Dishevelled Proteins/metabolism , Drosophila Proteins/metabolism , Humans , Transcription Factors/metabolism , beta Catenin/metabolism , rac GTP-Binding Proteins/metabolism , Delta Catenin
13.
Mol Cell ; 53(3): 444-57, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24412065

ABSTRACT

The Wnt canonical ligands elicit the activation of ß-catenin transcriptional activity, a response dependent on, but not limited to, ß-catenin stabilization through the inhibition of GSK3 activity. Two mechanisms have been proposed for this inhibition, one dependent on the binding and subsequent block of GSK3 to LRP5/6 Wnt coreceptor and another one on its sequestration into multivesicular bodies (MVBs). Here we report that internalization of the GSK3-containing Wnt-signalosome complex into MVBs is dependent on the dissociation of p120-catenin/cadherin from this complex. Disruption of cadherin-LRP5/6 interaction is controlled by cadherin phosphorylation and requires the previous separation of p120-catenin; thus, p120-catenin and cadherin mutants unable to dissociate from the complex block GSK3 sequestration into MVBs. These mutants substantially inhibit, but do not completely prevent, the ß-catenin upregulation caused by Wnt3a. These results, besides elucidating how GSK3 is sequestered into MVBs, support this mechanism as cause of ß-catenin stabilization by Wnt.


Subject(s)
Cadherins/physiology , Catenins/physiology , Glycogen Synthase Kinase 3/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Multivesicular Bodies/metabolism , Wnt Signaling Pathway , Animals , Cadherins/metabolism , Catenins/metabolism , Caveolins/metabolism , HEK293 Cells , Humans , Low Density Lipoprotein Receptor-Related Protein-5/analysis , Low Density Lipoprotein Receptor-Related Protein-6/analysis , Mice , Phosphorylation , Wnt3A Protein/metabolism , Wnt3A Protein/physiology , Delta Catenin
14.
J Cell Sci ; 125(Pt 22): 5288-301, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22946057

ABSTRACT

A role for Rac1 GTPase in canonical Wnt signaling has recently been demonstrated, showing that it is required for ß-catenin translocation to the nucleus. In this study, we investigated the mechanism of Rac1 stimulation by Wnt. Upregulation of Rac1 activity by Wnt3a temporally correlated with enhanced p120-catenin binding to Rac1 and Vav2. Vav2 and Rac1 association with p120-catenin was modulated by phosphorylation of this protein, which was stimulated upon serine/threonine phosphorylation by CK1 and inhibited by tyrosine phosphorylation by Src or Fyn. Acting on these two post-translational modifications, Wnt3a induced the release of p120-catenin from E-cadherin, enabled the interaction of p120-catenin with Vav2 and Rac1, and facilitated Rac1 activation by Vav2. Given that p120-catenin depletion disrupts gastrulation in Xenopus, we analyzed p120-catenin mutants for their ability to rescue this phenotype. In contrast to the wild-type protein or other controls, p120-catenin point mutants that were deficient in the release from E-cadherin or in Vav2 or Rac1 binding failed to rescue p120-catenin depletion. Collectively, these results indicate that binding of p120-catenin to Vav2 and Rac1 is required for the activation of this GTPase upon Wnt signaling.


Subject(s)
Catenins/metabolism , Proto-Oncogene Proteins c-vav/metabolism , Wnt3A Protein/pharmacology , rac1 GTP-Binding Protein/metabolism , Animals , Cadherins/metabolism , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytosol/drug effects , Cytosol/metabolism , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Enzyme Activation/drug effects , Gastrulation/drug effects , Humans , Models, Biological , Mutant Proteins/metabolism , Phosphorylation/drug effects , Phosphoserine/metabolism , Phosphotyrosine/metabolism , Protein Binding/drug effects , Protein Transport/drug effects , Signal Transduction/drug effects , Xenopus/embryology , Xenopus/metabolism , beta Catenin/metabolism , Delta Catenin
15.
J Cell Sci ; 124(Pt 13): 2298-309, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21670201

ABSTRACT

p120-catenin is an E-cadherin-associated protein that modulates E-cadherin function and stability. In response to Wnt3a, p120-catenin is phosphorylated at Ser268 and Ser269, disrupting its interaction with E-cadherin. Here, we describe that Wnt-induced p120-catenin phosphorylation at Ser268 and Ser269 also enhances its binding to the transcriptional factor Kaiso, preventing Kaiso-mediated inhibition of the ß-catenin-Tcf-4 transcriptional complex. Kaiso-mediated repression of this complex is due to its association not only with Tcf-4 but also with ß-catenin. Disruption of Tcf-4-Kaiso and ß-catenin-Kaiso interactions by p120-catenin not only releases Tcf-4 and ß-catenin enabling its mutual association and the formation of the transcriptional complex but also permits Kaiso binding to methylated CpG islands, an interaction that is weakly inhibited by p120-catenin. Consequently, Wnt stimulates Kaiso association to the CDKN2A promoter, which contains CpG sequences, in cells where these sequences are extensively methylated, such as HT-29 M6, an effect accompanied by decreased expression of its gene product. These results indicate that, when released from E-cadherin by Wnt3a-stimulated phosphorylation, p120-catenin controls the activity of the Kaiso transcriptional factor, enhancing its binding to repressed promoters and relieving its inhibition of the ß-catenin-Tcf-4 transcriptional complex.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Catenins/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Wnt3A Protein/metabolism , Cadherins/metabolism , Catenins/genetics , CpG Islands , Genes, p16 , Humans , Methylation , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Signal Transduction/genetics , Transcription Factor 4 , beta Catenin/metabolism , Delta Catenin
16.
Mol Cell Biol ; 31(14): 2877-88, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21606194

ABSTRACT

Activation of the Wnt pathway promotes the progressive phosphorylation of coreceptor LRP5/6 (low-density lipoprotein receptor-related proteins 5 and 6), creating a phosphorylated motif that inhibits glycogen synthase kinase 3ß (GSK-3ß), which in turn stabilizes ß-catenin, increasing the transcription of ß-catenin target genes. Casein kinase 1 (CK1) kinase family members play a complex role in this pathway, either as inhibitors or as activators. In this report, we have dissected the roles of CK1 isoforms in the early steps of Wnt signaling. CK1ε is constitutively bound to LRP5/6 through its interaction with p120-catenin and E-cadherin or N-cadherin and is activated upon Wnt3a stimulation. CK1α also associates with the LRP5/6/p120-catenin complex but, differently from CK1ε, only after Wnt3a addition. Binding of CK1α is dependent on CK1ε and occurs in a complex with axin. The two protein kinases function sequentially: whereas CK1ε is required for early responses to Wnt3a stimulation, such as recruitment of Dishevelled 2 (Dvl-2), CK1α participates in the release of p120-catenin from the complex, which activates p120-catenin for further actions on this pathway. Another CK1, CK1γ, acts at an intermediate level, since it is not necessary for Dvl-2 recruitment but for LRP5/6 phosphorylation at Thr1479 and axin binding. Therefore, our results indicate that CK1 isoforms work coordinately to promote the full response to Wnt stimulus.


Subject(s)
Casein Kinase I/metabolism , Isoenzymes/metabolism , Signal Transduction/physiology , Wnt Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Axin Protein , Casein Kinase I/genetics , Catenins/genetics , Catenins/metabolism , Cell Line , Dishevelled Proteins , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Isoenzymes/genetics , LDL-Receptor Related Proteins/genetics , LDL-Receptor Related Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-5 , Low Density Lipoprotein Receptor-Related Protein-6 , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Wnt Proteins/genetics , Wnt3 Protein , Wnt3A Protein , Delta Catenin
17.
J Cell Sci ; 123(Pt 15): 2621-31, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20940130

ABSTRACT

p120-catenin is an E-cadherin-associated protein that modulates E-cadherin function and stability. We describe here that p120-catenin is required for Wnt pathway signaling. p120-catenin binds and is phosphorylated by CK1ε in response to Wnt3a. p120-catenin also associates to the Wnt co-receptor LRP5/6, an interaction mediated by E-cadherin, showing an unexpected physical link between adherens junctions and a Wnt receptor. Depletion of p120-catenin abolishes CK1ε binding to LRP5/6 and prevents CK1ε activation upon Wnt3a stimulation. Elimination of p120-catenin also inhibits early responses to Wnt, such as LRP5/6 and Dvl-2 phosphorylation and axin recruitment to the signalosome, as well as later effects, such as ß-catenin stabilization. Moreover, since CK1ε is also required for E-cadherin phosphorylation, a modification that decreases the affinity for ß-catenin, p120-catenin depletion prevents the increase in ß-catenin transcriptional activity even in the absence of ß-catenin degradation. Therefore, these results demonstrate a novel and crucial function of p120-catenin in Wnt signaling and unveil additional points of regulation by this factor of ß-catenin transcriptional activity different of ß-catenin stability.


Subject(s)
Casein Kinase 1 epsilon/metabolism , Catenins/metabolism , Wnt Proteins/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Cadherins/genetics , Cadherins/metabolism , Casein Kinase 1 epsilon/genetics , Catenins/genetics , Cell Line, Tumor , Dishevelled Proteins , Humans , Immunoprecipitation , LDL-Receptor Related Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-5 , Low Density Lipoprotein Receptor-Related Protein-6 , Mass Spectrometry , Phosphoproteins/metabolism , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Binding/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Delta Catenin
18.
Proc Natl Acad Sci U S A ; 106(15): 6315-20, 2009 Apr 14.
Article in English | MEDLINE | ID: mdl-19325125

ABSTRACT

Notch has been linked to beta-catenin-dependent tumorigenesis; however, the mechanisms leading to Notch activation and the contribution of the Notch pathway to colorectal cancer is not yet understood. By microarray analysis, we have identified a group of genes downstream of Wnt/beta-catenin (down-regulated when blocking Wnt/beta-catenin) that are directly regulated by Notch (repressed by gamma-secretase inhibitors and up-regulated by active Notch1 in the absence of beta-catenin signaling). We demonstrate that Notch is downstream of Wnt in colorectal cancer cells through beta-catenin-mediated transcriptional activation of the Notch-ligand Jagged1. Consistently, expression of activated Notch1 partially reverts the effects of blocking Wnt/beta-catenin pathway in tumors implanted s.c. in nude mice. Crossing APC(Min/+) with Jagged1(+/Delta) mice is sufficient to significantly reduce the size of the polyps arising in the APC mutant background indicating that Notch is an essential modulator of tumorigenesis induced by nuclear beta-catenin. We show that this mechanism is operating in human tumors from Familial Adenomatous Polyposis patients. We conclude that Notch activation, accomplished by beta-catenin-mediated up-regulation of Jagged1, is required for tumorigenesis in the intestine. The Notch-specific genetic signature is sufficient to block differentiation and promote vasculogenesis in tumors whereas proliferation depends on both pathways.


Subject(s)
Calcium-Binding Proteins/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Receptors, Notch/metabolism , Signal Transduction , Wnt Proteins/metabolism , Alleles , Animals , Calcium-Binding Proteins/genetics , Cell Line , Cell Nucleus/metabolism , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein , Membrane Proteins/genetics , Mice , Mice, Transgenic , Serrate-Jagged Proteins , TCF Transcription Factors/metabolism , Transcription, Genetic/genetics , beta Catenin/metabolism
19.
PLoS One ; 3(12): e4080, 2008.
Article in English | MEDLINE | ID: mdl-19114997

ABSTRACT

Presenilin1 (PS1) is a component of the gamma-secretase complex mutated in cases of Familial Alzheimer's disease (FAD). PS1 is synthesized as a 50 kDa peptide subsequently processed to two 29 and 20 kDa subunits that remain associated. Processing of PS1 is inhibited by several mutations detected in FAD patients. PS1 acts as negative modulator of beta-catenin.Tcf-4 transcriptional activity. In this article we show that in murine embryonic fibroblasts (MEFs) the mechanisms of action of the processed and non-processed forms of PS1 on beta-catenin.Tcf-4 transcription are different. Whereas non-processed PS1 inhibits beta-catenin.Tcf-4 activity through a mechanism independent of gamma-secretase and associated with the interaction of this protein with plakoglobin and Tcf-4, the effect of processed PS1 is prevented by gamma-secretase inhibitors, and requires its interaction with E- or N-cadherin and the generation of cytosolic terminal fragments of these two cadherins, which in turn destabilize the beta-catenin transcriptional cofactor CBP. Accordingly, the two forms of PS1 interact differently with E-cadherin or beta-catenin and plakoglobin: whereas processed PS1 binds E-cadherin with high affinity and beta-catenin or plakoglobin weakly, the non-processed form behaves inversely. Moreover, contrarily to processed PS1, that decreases the levels of c-fos RNA, non-processed PS1 inhibits the expression c-myc, a known target of beta-catenin.Tcf-4, and does not block the activity of other transcriptional factors requiring CBP. These results indicate that prevention of PS1 processing in FAD affects the mechanism of repression of the transcriptional activity dependent on beta-catenin.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Presenilin-1/metabolism , TCF Transcription Factors/genetics , Transcription, Genetic , beta Catenin/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Animals , Cell Proliferation , Cells, Cultured , Humans , Mice , TCF Transcription Factors/metabolism , Transfection , beta Catenin/genetics
20.
J Cell Biol ; 183(4): 697-710, 2008 Nov 17.
Article in English | MEDLINE | ID: mdl-19015318

ABSTRACT

The active vitamin D metabolite 1,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3)) inhibits proliferation and promotes differentiation of colon cancer cells through the activation of vitamin D receptor (VDR), a transcription factor of the nuclear receptor superfamily. Additionally, 1,25(OH)(2)D(3) has several nongenomic effects of uncertain relevance. We show that 1,25(OH)(2)D(3) induces a transcription-independent Ca(2+) influx and activation of RhoA-Rho-associated coiled kinase (ROCK). This requires VDR and is followed by activation of the p38 mitogen-activated protein kinase (p38MAPK) and mitogen- and stress-activated kinase 1 (MSK1). As shown by the use of chemical inhibitors, dominant-negative mutants and small interfering RNA, RhoA-ROCK, and p38MAPK-MSK1 activation is necessary for the induction of CDH1/E-cadherin, CYP24, and other genes and of an adhesive phenotype by 1,25(OH)(2)D(3). RhoA-ROCK and MSK1 are also required for the inhibition of Wnt-beta-catenin pathway and cell proliferation. Thus, the action of 1,25(OH)(2)D(3) on colon carcinoma cells depends on the dual action of VDR as a transcription factor and a nongenomic activator of RhoA-ROCK and p38MAPK-MSK1.


Subject(s)
Calcitriol/pharmacology , Colonic Neoplasms/enzymology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Vitamins/metabolism , Wnt Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Antigens, CD , Caco-2 Cells , Cadherins/genetics , Cadherins/metabolism , Calcitriol/metabolism , Calcium/metabolism , Cdh1 Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Colonic Neoplasms/genetics , Enzyme Activation/drug effects , Humans , Mice , NIH 3T3 Cells , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism , Transcription, Genetic/drug effects , Vitamin D3 24-Hydroxylase , Vitamins/pharmacology , Wnt Proteins/genetics , beta Catenin/genetics , beta Catenin/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , rho GTP-Binding Proteins/genetics , rho-Associated Kinases/genetics , rhoA GTP-Binding Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...