46 results on '"Javelaud D"'
Search Results
2. Stable Overexpression of Smad7 in Human Melanoma Cells Inhibits Bone Metastasis In Vivo
- Author
-
Javelaud, D, Mohammad, K S, McKenna, C R, Niewolna, M, André, J, Delmas, V, Larue, L, Guise, T A, and Mauviel, A
- Published
- 2006
3. Stable overexpression of Smad7 in human melanoma cells inhibits their tumorigenicity in vitro and in vivo: 874
- Author
-
Javelaud, D, Moller, M, Sextius, P, Delmas, V, Menashi, S, Larue, L, and Mauviel, A
- Published
- 2005
4. STABLE OVEREXPRESSION OF SMAD7 IN HUMAN MELANOMA CELLS SPECIFICALLY ALTERS THEIR INVASIVE PHENOTYPE IN VITRO
- Author
-
Javelaud, D., Möller, M., Dennler, S., Menashi, S., Larue, L., and Mauviel, Alain
- Published
- 2004
5. 487 Expression of metastasis and invasion related molecules in malignant melanoma
- Author
-
Kim, J., primary, Gilbert, C., additional, Javelaud, D., additional, Mauviel, A., additional, and Chung, K., additional
- Published
- 2016
- Full Text
- View/download PDF
6. GLI2-mediated melanoma invasion and metastasis.
- Author
-
Alexaki, V.I., Javelaud, D., Kempen, L.C.L.T. van, Mohammad, K.S., Dennler, S., Luciani, F., Hoek, K.S., Juarez, P., Goydos, J.S., Fournier, P.J., Sibon, C., Bertolotto, C., Verrecchia, F., Saule, S., Delmas, V., Ballotti, R., Larue, L., Saiag, P., Guise, T.A., Mauviel, A., Alexaki, V.I., Javelaud, D., Kempen, L.C.L.T. van, Mohammad, K.S., Dennler, S., Luciani, F., Hoek, K.S., Juarez, P., Goydos, J.S., Fournier, P.J., Sibon, C., Bertolotto, C., Verrecchia, F., Saule, S., Delmas, V., Ballotti, R., Larue, L., Saiag, P., Guise, T.A., and Mauviel, A.
- Abstract
Contains fulltext : 88566.pdf (publisher's version ) (Closed access), BACKGROUND: The transforming growth factor-beta (TGF-beta) pathway, which has both tumor suppressor and pro-oncogenic activities, is often constitutively active in melanoma and is a marker of poor prognosis. Recently, we identified GLI2, a mediator of the hedgehog pathway, as a transcriptional target of TGF-beta signaling. METHODS: We used real-time reverse transcription-polymerase chain reaction (RT-PCR) and western blotting to determine GLI2 expression in human melanoma cell lines and subsequently classified them as GLI2high or as GLI2low according to their relative GLI2 mRNA and protein expression levels. GLI2 expression was reduced in a GLI2high cell line with lentiviral expression of short hairpin RNA targeting GLI2. We assessed the role of GLI2 in melanoma cell invasiveness in Matrigel assays. We measured secretion of matrix metalloproteinase (MMP)-2 and MMP-9 by gelatin zymography and expression of E-cadherin by western blotting and RT-PCR. The role of GLI2 in development of bone metastases was determined following intracardiac injection of melanoma cells in immunocompromised mice (n = 5-13). Human melanoma samples (n = 79) at various stages of disease progression were analyzed for GLI2 and E-cadherin expression by immunohistochemistry, in situ hybridization, or RT-PCR. All statistical tests were two-sided. RESULTS: Among melanoma cell lines, increased GLI2 expression was associated with loss of E-cadherin expression and with increased capacity to invade Matrigel and to form bone metastases in mice (mean osteolytic tumor area: GLI2high vs GLI2low, 2.81 vs 0.93 mm(2), difference = 1.88 mm(2), 95% confidence interval [CI] = 1.16 to 2.60, P < .001). Reduction of GLI2 expression in melanoma cells that had expressed high levels of GLI2 substantially inhibited both basal and TGF-beta-induced cell migration, invasion (mean number of Matrigel invading cells: shGLI2 vs shCtrl (control), 52.6 vs 100, difference = 47.4, 95% CI = 37.0 to 57.8, P = .024; for shGLI2 + TGF
- Published
- 2010
7. GLI2-mediated melanoma invasion and metastasis
- Author
-
Alexaki, V I, Javelaud, D, Van Kempen, L C L, Mohammad, K S, Dennler, S, Luciani, F, Hoek, K S, Juàrez, P, Goydos, J S, Fournier, P J, Sibon, C, Bertolotto, C, Verrecchia, F, Saule, S, Delmas, V, Ballotti, R, Larue, L, Saiag, P, Guise, T A, Mauviel, A, Alexaki, V I, Javelaud, D, Van Kempen, L C L, Mohammad, K S, Dennler, S, Luciani, F, Hoek, K S, Juàrez, P, Goydos, J S, Fournier, P J, Sibon, C, Bertolotto, C, Verrecchia, F, Saule, S, Delmas, V, Ballotti, R, Larue, L, Saiag, P, Guise, T A, and Mauviel, A
- Abstract
BACKGROUND: The transforming growth factor-beta (TGF-beta) pathway, which has both tumor suppressor and pro-oncogenic activities, is often constitutively active in melanoma and is a marker of poor prognosis. Recently, we identified GLI2, a mediator of the hedgehog pathway, as a transcriptional target of TGF-beta signaling. METHODS: We used real-time reverse transcription-polymerase chain reaction (RT-PCR) and western blotting to determine GLI2 expression in human melanoma cell lines and subsequently classified them as GLI2high or as GLI2low according to their relative GLI2 mRNA and protein expression levels. GLI2 expression was reduced in a GLI2high cell line with lentiviral expression of short hairpin RNA targeting GLI2. We assessed the role of GLI2 in melanoma cell invasiveness in Matrigel assays. We measured secretion of matrix metalloproteinase (MMP)-2 and MMP-9 by gelatin zymography and expression of E-cadherin by western blotting and RT-PCR. The role of GLI2 in development of bone metastases was determined following intracardiac injection of melanoma cells in immunocompromised mice (n = 5-13). Human melanoma samples (n = 79) at various stages of disease progression were analyzed for GLI2 and E-cadherin expression by immunohistochemistry, in situ hybridization, or RT-PCR. All statistical tests were two-sided. RESULTS: Among melanoma cell lines, increased GLI2 expression was associated with loss of E-cadherin expression and with increased capacity to invade Matrigel and to form bone metastases in mice (mean osteolytic tumor area: GLI2high vs GLI2low, 2.81 vs 0.93 mm(2), difference = 1.88 mm(2), 95% confidence interval [CI] = 1.16 to 2.60, P < .001). Reduction of GLI2 expression in melanoma cells that had expressed high levels of GLI2 substantially inhibited both basal and TGF-beta-induced cell migration, invasion (mean number of Matrigel invading cells: shGLI2 vs shCtrl (control), 52.6 vs 100, difference = 47.4, 95% CI = 37.0 to 57.8, P = .024; for shGLI2 + TGF
- Published
- 2010
8. CDKN1A (cyclin-dependent kinase inhibitor 1A)
- Author
-
Javelaud, D, primary and Besançon, F, additional
- Published
- 2011
- Full Text
- View/download PDF
9. Stable overexpression of Smad7 in human melanoma cells inhibits bone metastasis
- Author
-
Javelaud, D., primary, Mohammad, K., additional, McKenna, C., additional, Fournier, P., additional, Luciani, F., additional, Andr??, J., additional, Delmas, V., additional, Larue, L., additional, Guise, T., additional, and Mauviel, A., additional
- Published
- 2006
- Full Text
- View/download PDF
10. Transforming growth factor-βs : signalisation et rôles physiopathologiques
- Author
-
Javelaud, D, primary and Mauviel, A, additional
- Published
- 2004
- Full Text
- View/download PDF
11. p21( WAF1/CIP1) : un inhibiteur de l'apoptose.
- Author
-
Javelaud, D, primary and Besançon, F, additional
- Published
- 2000
- Full Text
- View/download PDF
12. Transforming growth factor-betas: smad signaling and roles in physiopathology.
- Author
-
Javelaud, D. and Mauviel, A.
- Subjects
- *
TRANSFORMING growth factors , *PEPTIDES , *EXTRACELLULAR matrix , *CELL migration - Abstract
Transforming growth factor-β (TGF-β) family members are multifunctional peptide growth factors that regulate cell growth, differentiation, extracellular matrix production and cell migration and embryonic development. Knock-out experiments for the three mammalian isoforms of TGF-β in mice have demonstrated their importance in regulating inflammation and tissue repair. Also, TGF-β has been implicated in the pathogenesis of human diseases, including tissue fibrosis and carcinogenesis. In the latter case, it may exert both tumor suppressor and pro-oncogenic activities depending on the stage of the tumor. Smads proteins constitute the core components of the intracellular signaling cascade initiated by TGF-β receptors, as they carry signals from the cell surface directly to the nucleus; where they act as transcription factors. [Copyright &y& Elsevier]
- Published
- 2004
- Full Text
- View/download PDF
13. Efficient TGF-β/SMAD signaling in human melanoma cells associated with high c-SKI/SnoN expression
- Author
-
Alexaki Vasileia I, van Kempen Leon, Javelaud Delphine, Le Scolan Erwan, Luo Kunxin, and Mauviel Alain
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background SKI and SnoN proteins have been shown to inhibit TGF-β signaling, acting both as transcriptional co-repressors in the cell nucleus, and as sequestrators of SMAD proteins in the cytoplasm. TGF-β, on the other hand, induces rapid, proteasome-mediated, degradation of both proteins. How elevated SKI and SnoN protein levels co-exist with active autocrine TGF-β signaling in cancer cells is yet to be understood. Results In this study, we found elevated SKI and SnoN protein levels in a panel of melanoma cell lines, as compared to normal melanocytes. There was no correlation between SKI protein content and the capacity of melanoma cells to invade Matrigel™, to form subcutaneous tumors, or to metastasize to bone after intracardiac inoculation into nude mice. Nor did we find a correlation between SKI expression and histopathological staging of human melanoma. TGF-β induced a rapid and dose-dependent degradation of SKI protein, associated with SMAD3/4 specific transcriptional response and induction of pro-metastatic target genes, partially prevented by pharmacologic blockade of proteasome activity. SKI knockdown in 1205Lu melanoma cells did not alter their invasive capacity or transcriptional responses to TGF-β, and did not allow p21 expression in response to TGF-β or reveal any growth inhibitory activity of TGF-β. Conclusions Despite high expression in melanoma cells, the role of SKI in melanoma remains elusive: SKI does not efficiently interfere with the pro-oncogenic activities of TGF-β, unless stabilized by proteasome blockade. Its highly labile nature makes it an unlikely target for therapeutic intervention.
- Published
- 2011
- Full Text
- View/download PDF
14. IpaA reveals distinct modes of vinculin activation during Shigella invasion and cell-matrix adhesion.
- Author
-
Cocom-Chan B, Khakzad H, Konate M, Aguilar DI, Bello C, Valencia-Gallardo C, Zarrouk Y, Fattaccioli J, Mauviel A, Javelaud D, and Tran Van Nhieu G
- Subjects
- Humans, Bacterial Proteins metabolism, Bacterial Proteins genetics, Mutation, Host-Pathogen Interactions, HeLa Cells, Protein Binding, Shigella metabolism, Shigella genetics, Antigens, Bacterial metabolism, Antigens, Bacterial genetics, Dysentery, Bacillary microbiology, Dysentery, Bacillary metabolism, Vinculin metabolism, Vinculin genetics, Focal Adhesions metabolism, Cell Adhesion
- Abstract
Vinculin is a cytoskeletal linker strengthening cell adhesion. The Shigella IpaA invasion effector binds to vinculin to promote vinculin supra-activation associated with head-domain-mediated oligomerization. Our study investigates the impact of mutations of vinculin D1D2 subdomains' residues predicted to interact with IpaA VBS3. These mutations affected the rate of D1D2 trimer formation with distinct effects on monomer disappearance, consistent with structural modeling of a closed and open D1D2 conformer induced by IpaA. Notably, mutations targeting the closed D1D2 conformer significantly reduced Shigella invasion of host cells as opposed to mutations targeting the open D1D2 conformer and later stages of vinculin head-domain oligomerization. In contrast, all mutations affected the formation of focal adhesions (FAs), supporting the involvement of vinculin supra-activation in this process. Our findings suggest that IpaA-induced vinculin supra-activation primarily reinforces matrix adhesion in infected cells, rather than promoting bacterial invasion. Consistently, shear stress studies pointed to a key role for IpaA-induced vinculin supra-activation in accelerating and strengthening cell-matrix adhesion., (© 2024 Cocom-Chan et al.)
- Published
- 2024
- Full Text
- View/download PDF
15. GLI1/GLI2 functional interplay is required to control Hedgehog/GLI targets gene expression.
- Author
-
Tolosa EJ, Fernandez-Barrena MG, Iguchi E, McCleary-Wheeler AL, Carr RM, Almada LL, Flores LF, Vera RE, Alfonse GW, Marks DL, Hogenson TL, Vrabel AM, Horn IP, Koenig AN, Safgren SL, Sigafoos AN, Erkan M, Romecin-Duran PA, Sarabia Gonzalez A, Zhou B, Javelaud D, Marsaud V, Graham RP, Mauviel A, Elsawa SF, and Fernandez-Zapico ME
- Subjects
- Cell Line, Tumor, Hedgehog Proteins genetics, Humans, Nuclear Proteins genetics, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Protein Multimerization, Rhabdomyosarcoma genetics, Rhabdomyosarcoma pathology, Zinc Finger Protein GLI1 genetics, Zinc Finger Protein Gli2 genetics, Gene Expression Regulation, Neoplastic, Hedgehog Proteins metabolism, Nuclear Proteins metabolism, Pancreatic Neoplasms metabolism, Rhabdomyosarcoma metabolism, Zinc Finger Protein GLI1 metabolism, Zinc Finger Protein Gli2 metabolism
- Abstract
The Hedgehog-regulated transcription factors GLI1 and GLI2 play overlapping roles in development and disease; however, the mechanisms underlying their interplay remain elusive. We report for the first time that GLI1 and GLI2 physically and functionally interact in cancer cells. GLI1 and GLI2 were shown to co-immunoprecipitate in PANC1 pancreatic cancer cells and RMS13 rhabdomyosarcoma cells. Mapping analysis demonstrated that the zinc finger domains of both proteins are required for their heteromerization. RNAi knockdown of either GLI1 or GLI2 inhibited expression of many well-characterized GLI target genes (BCL2, MYCN, PTCH2, IL7 and CCND1) in PANC1 cells, whereas PTCH1 expression was only inhibited by GLI1 depletion. qPCR screening of a large set of putative canonical and non-canonical Hedgehog/GLI targets identified further genes (e.g. E2F1, BMP1, CDK2) strongly down-regulated by GLI1 and/or GLI2 depletion in PANC1 cells, and demonstrated that ANO1, AQP1 and SOCS1 are up-regulated by knockdown of either GLI1 or GLI2. Chromatin immunoprecipitation showed that GLI1 and GLI2 occupied the same regions at the BCL2, MYCN and CCND1 promoters. Furthermore, depletion of GLI1 inhibited GLI2 occupancy at these promoters, suggesting that GLI1/GLI2 interaction is required for the recruitment of GLI2 to these sites. Together, these findings indicate that GLI1 and GLI2 co-ordinately regulate the transcription of some genes, and provide mechanistic insight into the roles of GLI proteins in carcinogenesis., (© 2020 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society.)
- Published
- 2020
- Full Text
- View/download PDF
16. Large-scale pan-cancer analysis reveals broad prognostic association between TGF-β ligands, not Hedgehog, and GLI1/2 expression in tumors.
- Author
-
de Reyniès A, Javelaud D, Elarouci N, Marsaud V, Gilbert C, and Mauviel A
- Subjects
- Computational Biology, Gene Expression Profiling, Humans, Ligands, Multivariate Analysis, Neoplasms genetics, Oligonucleotide Array Sequence Analysis, Prognosis, Proportional Hazards Models, Risk Factors, Signal Transduction genetics, Transcriptome, Gene Expression Regulation, Neoplastic, Hedgehog Proteins genetics, Neoplasms diagnosis, Nuclear Proteins genetics, Transforming Growth Factor beta metabolism, Zinc Finger Protein GLI1 genetics, Zinc Finger Protein Gli2 genetics
- Abstract
GLI1 expression is broadly accepted as a marker of Hedgehog pathway activation in tumors. Efficacy of Hedgehog inhibitors is essentially limited to tumors bearing activating mutations of the pathway. GLI2, a critical Hedgehog effector, is necessary for GLI1 expression and is a direct transcriptional target of TGF-β/SMAD signaling. We examined the expression correlations of GLI1/2 with TGFB and HH genes in 152 distinct transcriptome datasets totaling over 23,500 patients and representing 37 types of neoplasms. Their prognostic value was measured in over 15,000 clinically annotated tumor samples from 26 tumor types. In most tumor types, GLI1 and GLI2 follow a similar pattern of expression and are equally correlated with HH and TGFB genes. However, GLI1/2 broadly share prognostic value with TGFB genes and a mesenchymal/EMT signature, not with HH genes. Our results provide a likely explanation for the frequent failure of anti-Hedgehog therapies in tumors, as they suggest a key role for TGF-β, not Hedgehog, ligands, in tumors with elevated GLI1/2-expression.
- Published
- 2020
- Full Text
- View/download PDF
17. Transcriptional repression of the tyrosinase-related protein 2 gene by transforming growth factor-β and the Kruppel-like transcription factor GLI2.
- Author
-
Pierrat MJ, Marsaud V, Mauviel A, and Javelaud D
- Subjects
- Cell Line, Tumor, Computational Biology, Humans, Melanoma pathology, Mutagenesis, Site-Directed, Promoter Regions, Genetic genetics, Skin Neoplasms pathology, Transcription, Genetic, Gene Expression Regulation, Neoplastic, Intramolecular Oxidoreductases genetics, Melanoma genetics, Nuclear Proteins metabolism, Skin Neoplasms genetics, Transforming Growth Factor beta metabolism, Zinc Finger Protein Gli2 metabolism
- Abstract
Background: Tyrosinase-Related Protein 2 (TRP2) is an enzyme involved in melanogenesis, that also exerts proliferative, anti-apoptotic and immunogenic functions in melanoma cells. TRP2 transcription is regulated by the melanocytic master transcription factor MITF. GLI2, a transcription factor that acts downstream of Hedgehog signaling, is also a direct transcriptional target of the TGF-β/SMAD pathway that contributes to melanoma progression and exerts transcriptional antagonistic activities against MITF., Objectives: To characterize the molecular events responsible for TGF-β and GLI2 repression of TRP2 expression., Methods: In silico promoter analysis, transient cell transfection experiments with 5'-end TRP2 promoter deletion constructs, chromatin immuno-precipitation, and site-directed promoter mutagenesis were used to dissect the molecular mechanisms of TRP2 gene regulation by TGF-β and GLI2., Results: We demonstrate that TGF-β and GLI2-specific TRP2 repression involves direct mechanisms that occur in addition to MITF downregulation by TGF-β and GLI2. We identify two functional GLI2 binding sites within the TRP2 promoter that are critical for TGF-β and GLI2 responsiveness, one of them overlapping a CREB binding site. GLI2 and CREB competing for the same cis-element is associated with opposite transcriptional outcome., Conclusion: Our results further refine the understanding of how TGF-β and GLI2 control the phenotypic plasticity of melanoma cells. In particular, we identify critical GLI2-binding cis-elements within the TRP2 promoter region that allow for its transcriptional repression independently from MITF concomitant downregulation., (Copyright © 2019 Japanese Society for Investigative Dermatology. Published by Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
18. How Bad Is the Hedgehog? GLI-Dependent, Hedgehog-Independent Cancers on the Importance of Biomarkers for Proper Patients Selection.
- Author
-
Javelaud D, De Reyniès A, and Mauviel A
- Abstract
Hedgehog (HH) signaling plays an important role both during embryonic development and adult life. It is involved in the regulation of cell differentiation, cell proliferation and tissue polarity, as well as in the maintenance of stem cells, tissue repair, and regeneration (Briscoe and Therond, 2013; Jiang and Hui, 2008). Three ligands, Indian, Sonic, and Desert HH, can activate this pathway. Binding of HH ligands to their receptor, PTCH1 (Figure 1) lift its inhibition on SMO, resulting in activation and nuclear translocation of GLI transcription factors (Javelaud et al., 2012). The vertebrate GLI gene family is composed of three distinct genes GLI1, GLI2, and GLI3, encoding Krüppel-like transcription factors. GLI proteins exhibit distinct regulations, biochemical properties, and target genes. GLI3 acts as the main repressor of the pathway in the absence of HH ligands, whereas, in their presence, GLI2 is the main HH effector that drives the expression of GLI1 (Briscoe and Therond, 2013)., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
19. Cell density sensing alters TGF-β signaling in a cell-type-specific manner, independent from Hippo pathway activation.
- Author
-
Nallet-Staub F, Yin X, Gilbert C, Marsaud V, Ben Mimoun S, Javelaud D, Leof EB, and Mauviel A
- Subjects
- Acyltransferases, Blotting, Western, Cell Count, Cell Cycle Proteins, Cell Proliferation, Cells, Cultured, Fluorescent Antibody Technique, Hippo Signaling Pathway, Humans, Immunoenzyme Techniques, Immunoprecipitation, Nuclear Proteins genetics, Phosphorylation, Protein Serine-Threonine Kinases genetics, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Transcription Factors genetics, Transforming Growth Factor beta genetics, Cytoplasm metabolism, Nuclear Proteins metabolism, Protein Serine-Threonine Kinases metabolism, Receptors, Transforming Growth Factor beta metabolism, Signal Transduction, Transcription Factors metabolism, Transforming Growth Factor beta metabolism
- Abstract
Cell-cell contacts inhibit cell growth and proliferation in part by activating the Hippo pathway that drives the phosphorylation and nuclear exclusion of the transcriptional coactivators YAP and TAZ. Cell density and Hippo signaling have also been reported to block transforming growth factor β (TGF-β) responses, based on the ability of phospho-YAP/TAZ to sequester TGF-β-activated SMAD complexes in the cytoplasm. Herein, we provide evidence that epithelial cell polarization interferes with TGF-β signaling well upstream and independent of cytoplasmic YAP/TAZ. Rather, polarized basolateral presentation of TGF-β receptors I and II deprives apically delivered TGF-β of access to its receptors. Basolateral ligand delivery nonetheless remains entirely effective to induce TGF-β responses. These data demonstrate that cell-type-specific inhibition of TGF-β signaling by cell density is restricted to polarized epithelial cells and reflects the polarized distribution of TGF-β receptors, which thus affects SMAD activation irrespective of Hippo pathway activation., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
20. GLI2 cooperates with ZEB1 for transcriptional repression of CDH1 expression in human melanoma cells.
- Author
-
Perrot CY, Gilbert C, Marsaud V, Postigo A, Javelaud D, and Mauviel A
- Subjects
- Antigens, CD, Base Sequence, Binding Sites, Cell Line, Tumor, Cell Nucleus drug effects, Cell Nucleus pathology, DNA Methylation genetics, Gene Knockdown Techniques, Gene Silencing drug effects, Humans, Melanoma pathology, Models, Biological, Molecular Sequence Data, Promoter Regions, Genetic, Protein Binding drug effects, Repressor Proteins metabolism, Skin Neoplasms pathology, Snail Family Transcription Factors, Transforming Growth Factor beta pharmacology, Zinc Finger E-box-Binding Homeobox 1, Zinc Finger Protein Gli2, Cadherins genetics, Gene Expression Regulation, Neoplastic drug effects, Homeodomain Proteins metabolism, Kruppel-Like Transcription Factors metabolism, Melanoma genetics, Nuclear Proteins metabolism, Skin Neoplasms genetics, Transcription Factors metabolism, Transcription, Genetic drug effects
- Abstract
In melanoma cells, high expression of the transcription factor GLI2 is associated with increased invasive potential and loss of E-cadherin expression, an event reminiscent of the epithelial-to-mesenchymal transition (EMT). Herein, we provide evidence that GLI2 represses E-cadherin gene (CDH1) expression in melanoma cells via distinct mechanisms, enhancing transcription of the EMT-activator ZEB1 and cooperative repression of CDH1 gene transcription via direct binding of both GLI2 and ZEB1 to two closely positioned Kruppel-like factor-binding sites within the CDH1 promoter. GLI2 silencing rescued CDH1 expression except in melanoma cell lines in which the CDH1 promoter was hypermethylated. Proximity ligation assays identified GLI2-ZEB1 complexes in melanoma cell nuclei, proportional to endogenous GLI2 and ZEB1 expression, and whose accumulation was enhanced by the classical EMT inducer TGF-β. These data identify GLI2 as a critical modulator of the cadherin switch in melanoma, a molecular process that is critical for metastatic spread of the disease., (© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2013
- Full Text
- View/download PDF
21. Insights into the Transforming Growth Factor-β Signaling Pathway in Cutaneous Melanoma.
- Author
-
Perrot CY, Javelaud D, and Mauviel A
- Abstract
Transforming growth factor-β (TGF-β) is a pleiotropic growth factor with broad tissue distribution that plays critical roles during embryonic development, normal tissue homeostasis, and cancer. While its cytostatic activity on normal epithelial cells initially defined TGF-β signaling as a tumor suppressor pathway, there is ample evidence indicating that TGF-β is a potent pro-tumorigenic agent, acting via autocrine and paracrine mechanisms to promote peri-tumoral angiogenesis, together with tumor cell migration, immune escape, and dissemination to metastatic sites. This review summarizes the current knowledge on the implication of TGF-β signaling in melanoma.
- Published
- 2013
- Full Text
- View/download PDF
22. Overlapping activities of TGF-β and Hedgehog signaling in cancer: therapeutic targets for cancer treatment.
- Author
-
Perrot CY, Javelaud D, and Mauviel A
- Subjects
- Animals, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents administration & dosage, Antineoplastic Agents pharmacology, Clinical Trials as Topic, Hedgehog Proteins genetics, Humans, Neoplasms metabolism, Oligonucleotides, Antisense administration & dosage, Oligonucleotides, Antisense pharmacology, Oligonucleotides, Antisense therapeutic use, Small Molecule Libraries administration & dosage, Small Molecule Libraries pharmacology, Small Molecule Libraries therapeutic use, Transforming Growth Factor alpha genetics, Antineoplastic Agents therapeutic use, Hedgehog Proteins antagonists & inhibitors, Neoplasms drug therapy, Signal Transduction drug effects, Transforming Growth Factor alpha antagonists & inhibitors
- Abstract
Recent advances in the field of cancer therapeutics come from the development of drugs that specifically recognize validated oncogenic or pro-metastatic targets. The latter may be mutated proteins with altered function, such as kinases that become constitutively active, or critical components of growth factor signaling pathways, whose deregulation leads to aberrant malignant cell proliferation and dissemination to metastatic sites. We herein focus on the description of the overlapping activities of two important developmental pathways often exacerbated in cancer, namely Transforming Growth Factor-β (TGF-β) and Hedgehog (HH) signaling, with a special emphasis on the unifying oncogenic role played by GLI1/2 transcription factors. The latter are the main effectors of the canonical HH pathway, yet are direct target genes of TGF-β/SMAD signal transduction. While tumor-suppressor in healthy and pre-malignant tissues, TGF-β is often expressed at high levels in tumors and contributes to tumor growth, escape from immune surveillance, invasion and metastasis. HH signaling regulates cell proliferation, differentiation and apoptosis, and aberrant HH signaling is found in a variety of cancers. We discuss the current knowledge on HH and TGF-β implication in cancer including cancer stem cell biology, as well as the current state, both successes and failures, of targeted therapeutics aimed at blocking either of these pathways in the pre-clinical and clinical settings., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
23. Halofuginone inhibits the establishment and progression of melanoma bone metastases.
- Author
-
Juárez P, Mohammad KS, Yin JJ, Fournier PG, McKenna RC, Davis HW, Peng XH, Niewolna M, Javelaud D, Chirgwin JM, Mauviel A, and Guise TA
- Subjects
- Animals, Apoptosis drug effects, Bone Neoplasms metabolism, Cell Growth Processes drug effects, Cell Line, Tumor, Disease Progression, Female, Gene Expression, Humans, Melanoma metabolism, Melanoma pathology, Melanoma secondary, Mice, Mice, Nude, Signal Transduction, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Bone Neoplasms prevention & control, Bone Neoplasms secondary, Melanoma drug therapy, Piperidines pharmacology, Quinazolinones pharmacology
- Abstract
TGF-β derived from bone fuels melanoma bone metastases by inducing tumor secretion of prometastatic factors that act on bone cells to change the skeletal microenvironment. Halofuginone is a plant alkaloid derivative that blocks TGF-β signaling with antiangiogenic and antiproliferative properties. Here, we show for the first time that halofuginone therapy decreases development and progression of bone metastasis caused by melanoma cells through the inhibition of TGF-β signaling. Halofuginone treatment of human melanoma cells inhibited cell proliferation, phosphorylation of SMAD proteins in response to TGF-β, and TGF-β-induced SMAD-driven transcription. In addition, halofuginone reduced expression of TGF-β target genes that enhance bone metastases, including PTHrP, CTGF, CXCR4, and IL11. Also, cell apoptosis was increased in response to halofuginone. In nude mice inoculated with 1205 Lu melanoma cells, a preventive protocol with halofuginone inhibited bone metastasis. The beneficial effects of halofuginone treatment were comparable with those observed with other anti-TGF-β strategies, including systemic administration of SD208, a small-molecule inhibitor of TGF-β receptor I kinase, or forced overexpression of Smad7, a negative regulator of TGF-β signaling. Furthermore, mice with established bone metastases treated with halofuginone had significantly less osteolysis than mice receiving placebo assessed by radiography. Thus, halofuginone is also effective in reducing the progression of melanoma bone metastases. Moreover, halofuginone treatment reduced melanoma metastasis to the brain, showing the potential of this novel treatment against cancer metastasis.
- Published
- 2012
- Full Text
- View/download PDF
24. Crosstalk between TGF-β and hedgehog signaling in cancer.
- Author
-
Javelaud D, Pierrat MJ, and Mauviel A
- Subjects
- Animals, Epithelial-Mesenchymal Transition, Fibrosis metabolism, Humans, Kruppel-Like Transcription Factors metabolism, Ligands, Mice, Models, Biological, Nuclear Proteins metabolism, Oncogene Proteins metabolism, RNA Processing, Post-Transcriptional, Rats, Signal Transduction, Trans-Activators metabolism, Zinc Finger Protein GLI1, Zinc Finger Protein Gli2, Hedgehog Proteins metabolism, Neoplasms metabolism, Transforming Growth Factor beta metabolism
- Abstract
Hedgehog (HH) and TGF-β signals control various aspects of embryonic development and cancer progression. While their canonical signal transduction cascades have been well characterized, there is increasing evidence that these pathways are able to exert overlapping activities that challenge efficient therapeutic targeting. We herein review the current knowledge on HH signaling and summarize the recent findings on the crosstalks between the HH and TGF-β pathways in cancer., (Copyright © 2012 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
25. Expression of microphthalmia-associated transcription factor (MITF), which is critical for melanoma progression, is inhibited by both transcription factor GLI2 and transforming growth factor-β.
- Author
-
Pierrat MJ, Marsaud V, Mauviel A, and Javelaud D
- Subjects
- Base Sequence, Cell Line, Tumor, Chromatin Immunoprecipitation, DNA Primers, DNA, Neoplasm genetics, Disease Progression, Electrophoretic Mobility Shift Assay, Gene Expression Regulation, Humans, Microphthalmia-Associated Transcription Factor physiology, Molecular Sequence Data, Promoter Regions, Genetic, Regulatory Sequences, Nucleic Acid, Transcription, Genetic, Zinc Finger Protein Gli2, Kruppel-Like Transcription Factors physiology, Melanoma pathology, Microphthalmia-Associated Transcription Factor genetics, Nuclear Proteins physiology, Transforming Growth Factor beta physiology
- Abstract
The melanocyte-specific transcription factor M-MITF is involved in numerous aspects of melanoblast lineage biology including pigmentation, survival, and migration. It plays complex roles at all stages of melanoma progression and metastasis. We established previously that GLI2, a Kruppel-like transcription factor that acts downstream of Hedgehog signaling, is a direct transcriptional target of the TGF-β/SMAD pathway and contributes to melanoma progression, exerting antagonistic activities against M-MITF to control melanoma cell invasiveness. Herein, we dissected the molecular mechanisms underlying both TGF-β and GLI2-driven M-MITF gene repression. Using transient cell transfection experiments with M-MITF promoter constructs, chromatin immunoprecipitation, site-directed mutagenesis, and electrophoretic mobility shift assays, we identified a GLI2 binding site within the -334/-296 region of the M-MITF promoter, critical for GLI2-driven transcriptional repression. This region is, however, not needed for inhibition of M-MITF promoter activity by TGF-β. We determined that TGF-β rapidly repressed protein kinase A activity, thus reducing both phospho-cAMP-response element-binding protein (CREB) levels and CREB-dependent transcription of the M-MITF promoter. Increased GLI2 binding to its cognate cis-element, associated with reduced CREB-dependent transcription, allowed maximal inhibition of the M-MITF promoter via two distinct mechanisms.
- Published
- 2012
- Full Text
- View/download PDF
26. Systematic classification of melanoma cells by phenotype-specific gene expression mapping.
- Author
-
Widmer DS, Cheng PF, Eichhoff OM, Belloni BC, Zipser MC, Schlegel NC, Javelaud D, Mauviel A, Dummer R, and Hoek KS
- Subjects
- Cell Proliferation, Gene Expression Regulation, Neoplastic, Genes, Neoplasm genetics, Genetic Association Studies, Humans, Melanoma genetics, Melanoma pathology, Microarray Analysis, Neoplasm Invasiveness, Phenotype, Reproducibility of Results, Skin Neoplasms genetics, Skin Neoplasms pathology, Tissue Distribution, Tumor Cells, Cultured classification, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Gene Expression Profiling methods, Melanoma classification, Skin Neoplasms classification
- Abstract
There is growing evidence that the metastatic spread of melanoma is driven not by a linear increase in tumorigenic aggressiveness, but rather by switching back and forth between two different phenotypes of metastatic potential. In vitro these phenotypes are respectively defined by the characteristics of strong proliferation/weak invasiveness and weak proliferation/strong invasiveness. Melanoma cell phenotype is tightly linked to gene expression. Taking advantage of this, we have developed a gene expression-based tool for predicting phenotype called Heuristic Online Phenotype Prediction. We demonstrate the predictive utility of this tool by comparing phenotype-specific signatures with measurements of characteristics of melanoma phenotype-specific biology in different melanoma cell lines and short-term cultures. We further show that 86% of 536 tested melanoma lines and short-term cultures are significantly associated with the phenotypes we describe. These findings reinforce the concept that a two-state system, as described by the phenotype switching model, underlies melanoma progression., (© 2012 John Wiley & Sons A/S.)
- Published
- 2012
- Full Text
- View/download PDF
27. GLI2 and M-MITF transcription factors control exclusive gene expression programs and inversely regulate invasion in human melanoma cells.
- Author
-
Javelaud D, Alexaki VI, Pierrat MJ, Hoek KS, Dennler S, Van Kempen L, Bertolotto C, Ballotti R, Saule S, Delmas V, and Mauviel A
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation drug effects, Cyclic AMP metabolism, Cyclic AMP-Dependent Protein Kinases metabolism, Humans, Kruppel-Like Transcription Factors genetics, Melanoma physiopathology, Mice, Mice, Nude, Microphthalmia-Associated Transcription Factor genetics, Neoplasm Invasiveness, Nuclear Proteins genetics, Pigmentation drug effects, Signal Transduction drug effects, Signal Transduction genetics, Skin Neoplasms physiopathology, Smad Proteins metabolism, Subcutaneous Tissue drug effects, Subcutaneous Tissue pathology, Transforming Growth Factor beta metabolism, Transforming Growth Factor beta pharmacology, Xenograft Model Antitumor Assays, Zinc Finger Protein Gli2, Gene Expression Regulation, Neoplastic drug effects, Kruppel-Like Transcription Factors metabolism, Melanoma genetics, Melanoma pathology, Microphthalmia-Associated Transcription Factor metabolism, Nuclear Proteins metabolism, Skin Neoplasms genetics, Skin Neoplasms pathology
- Abstract
We recently identified GLI2, the most active of GLI transcription factors, as a direct TGF-β/SMAD target, whose expression in melanoma cells is associated with increased invasiveness and metastatic capacity. In this work, we provide evidence that high GLI2 expression is inversely correlated with that of the melanocyte-specific transcription factor M-microphthalmia transcription factor (M-MITF) and associated transcriptional program. GLI2-expressing cell lines were characterized by the loss of M-MITF-dependent melanocytic differentiation markers and reduced pigmentation. The balance between M-MITF and GLI2 expression did not correlate with the presence or absence of BRAF-activating mutations, but rather was controlled by two distinct pathways: the TGF-β pathway, which favors GLI2 expression, and the protein kinase A (PKA)/cAMP pathway, which pushes the balance toward high M-MITF expression. Furthermore, overexpression and knockdown experiments demonstrated that GLI2 and M-MITF reciprocally repress each other's expression and control melanoma cell invasion in an opposite manner. These findings thus identify GLI2 as a critical transcription factor antagonizing M-MITF function to promote melanoma cell phenotypic plasticity and invasive behavior., (© 2011 John Wiley & Sons A/S.)
- Published
- 2011
- Full Text
- View/download PDF
28. TGF-β/SMAD/GLI2 signaling axis in cancer progression and metastasis.
- Author
-
Javelaud D, Alexaki VI, Dennler S, Mohammad KS, Guise TA, and Mauviel A
- Subjects
- Bone Neoplasms genetics, Bone Neoplasms metabolism, Bone Neoplasms secondary, Breast Neoplasms genetics, Breast Neoplasms metabolism, Breast Neoplasms pathology, Disease Progression, Female, Humans, Melanoma genetics, Melanoma metabolism, Melanoma secondary, Neoplasm Metastasis, Neoplasms genetics, Neoplasms metabolism, Signal Transduction, Skin Neoplasms genetics, Skin Neoplasms metabolism, Skin Neoplasms pathology, Zinc Finger Protein Gli2, Hedgehog Proteins metabolism, Kruppel-Like Transcription Factors metabolism, Neoplasms pathology, Nuclear Proteins metabolism, Smad Proteins metabolism, Transforming Growth Factor beta metabolism
- Abstract
The Hedgehog (HH) and TGF-β signaling pathways represent essential regulators of cell proliferation and differentiation during embryogenesis. Pathway deregulation is a characteristic of various cancers. Recently, evidence for a convergence of these pathways at the level of the GLI2 transcription factor in the context of tumor initiation and progression to metastasis has emerged. This short review summarizes recent knowledge about GLI2 function and mechanisms of action downstream of TGF-β in cancer., (©2011 AACR.)
- Published
- 2011
- Full Text
- View/download PDF
29. TGF-beta-RI kinase inhibitor SD-208 reduces the development and progression of melanoma bone metastases.
- Author
-
Mohammad KS, Javelaud D, Fournier PG, Niewolna M, McKenna CR, Peng XH, Duong V, Dunn LK, Mauviel A, and Guise TA
- Subjects
- Animals, Base Sequence, Bone Neoplasms secondary, Cell Line, Tumor, DNA Primers, Disease Models, Animal, Disease Progression, Gene Expression Profiling, Humans, Mice, Mice, Nude, Neoplasm Invasiveness, Phosphorylation, Smad Proteins metabolism, Bone Neoplasms prevention & control, Melanoma pathology, Protein Kinase Inhibitors pharmacology, Pteridines pharmacology, Receptors, Transforming Growth Factor beta antagonists & inhibitors
- Abstract
Melanoma often metastasizes to bone where it is exposed to high concentrations of TGF-β. Constitutive Smad signaling occurs in human melanoma. Because TGF-β promotes metastases to bone by several types of solid tumors including breast cancer, we hypothesized that pharmacologic blockade of the TGF-β signaling pathway may interfere with the capacity of melanoma cells to metastasize to bone. In this study, we tested the effect of a small molecule inhibitor of TGF-β receptor I kinase (TβRI), SD-208, on various parameters affecting the development and progression of melanoma, both in vitro and in a mouse model of human melanoma bone metastasis. In melanoma cell lines, SD-208 blocked TGF-β induction of Smad3 phosphorylation, Smad3/4-specific transcription, Matrigel invasion and expression of the TGF-β target genes PTHrP, IL-11, CTGF, and RUNX2. To assess effects of SD-208 on melanoma development and metastasis, nude mice were inoculated with 1205Lu melanoma cells into the left cardiac ventricle and drug was administered by oral gavage on prevention or treatment protocols. SD-208 (60 mg/kg/d), started 2 days before tumor inoculation prevented the development of osteolytic bone metastases compared with vehicle. In mice with established bone metastases, the size of osteolytic lesions was significantly reduced after 4 weeks treatment with SD-208 compared with vehicle-treated mice. Our results demonstrate that therapeutic targeting of TGF-β may prevent the development of melanoma bone metastases and decrease the progression of established osteolytic lesions., (© 2010 AACR.)
- Published
- 2011
- Full Text
- View/download PDF
30. Smad7 restricts melanoma invasion by restoring N-cadherin expression and establishing heterotypic cell-cell interactions in vivo.
- Author
-
DiVito KA, Trabosh VA, Chen YS, Chen Y, Albanese C, Javelaud D, Mauviel A, Simbulan-Rosenthal CM, and Rosenthal DS
- Subjects
- Cell Line, Tumor, Dermis metabolism, Dermis pathology, Disease Progression, Green Fluorescent Proteins metabolism, Humans, Keratinocytes metabolism, Male, Models, Biological, Neoplasm Invasiveness, Skin Transplantation, beta Catenin metabolism, Antigens, CD metabolism, Cadherins metabolism, Cell Communication, Melanoma metabolism, Melanoma pathology, Skin Neoplasms metabolism, Skin Neoplasms pathology, Smad7 Protein metabolism
- Abstract
The list of transforming growth factor-beta (TGF-β)-related proteins in non-canonical TGF-β signaling is growing. Examples include receptor-Smads directing micro-RNA processing and inhibitory-Smads, e.g. Smad7, directing cell adhesion. Human skin grafts with fluorescently tagged melanoma cells revealed Smad7-expressing cells positioned themselves proximal to the dermal-epidermal junction and failed to form tumors, while control cells readily invaded and formed tumors within the dermis. Smad7 significantly inhibited β-catenin T41/S45 phosphorylation associated with degradation and induced a 4.5-fold increase in full-length N-cadherin. Cell adhesion assays confirmed a strong interaction between Smad7-expressing cells and primary dermal fibroblasts mediated via N-cadherin, while control cells were incapable of such interaction. Immunofluorescent analysis of skin grafts indicated N-cadherin homotypic interaction at the surface of both Smad7 cells and primary dermal fibroblasts, in contrast to control melanoma cells. We propose that Smad7 suppresses β-catenin degradation and promotes interaction with N-cadherin, stabilizing association with neighboring dermal fibroblasts, thus mitigating invasion.
- Published
- 2010
- Full Text
- View/download PDF
31. GLI2-mediated melanoma invasion and metastasis.
- Author
-
Alexaki VI, Javelaud D, Van Kempen LC, Mohammad KS, Dennler S, Luciani F, Hoek KS, Juàrez P, Goydos JS, Fournier PJ, Sibon C, Bertolotto C, Verrecchia F, Saule S, Delmas V, Ballotti R, Larue L, Saiag P, Guise TA, and Mauviel A
- Subjects
- Animals, Blotting, Western, Cadherins metabolism, Cell Line, Tumor, Collagen, Drug Combinations, Gene Expression Regulation, Neoplastic, Hedgehog Proteins metabolism, Humans, Immunocompromised Host, Immunohistochemistry, In Situ Hybridization, Kruppel-Like Transcription Factors genetics, Laminin, Matrix Metalloproteinase 2 metabolism, Matrix Metalloproteinase 9 metabolism, Melanoma secondary, Mice, Neoplasm Invasiveness, Nuclear Proteins genetics, Polymerase Chain Reaction, Proteoglycans, RNA, Messenger metabolism, Signal Transduction, Up-Regulation, Zinc Finger Protein Gli2, Bone Neoplasms metabolism, Bone Neoplasms secondary, Kruppel-Like Transcription Factors metabolism, Melanoma metabolism, Melanoma pathology, Nuclear Proteins metabolism, Skin Neoplasms metabolism, Skin Neoplasms pathology
- Abstract
Background: The transforming growth factor-beta (TGF-beta) pathway, which has both tumor suppressor and pro-oncogenic activities, is often constitutively active in melanoma and is a marker of poor prognosis. Recently, we identified GLI2, a mediator of the hedgehog pathway, as a transcriptional target of TGF-beta signaling., Methods: We used real-time reverse transcription-polymerase chain reaction (RT-PCR) and western blotting to determine GLI2 expression in human melanoma cell lines and subsequently classified them as GLI2high or as GLI2low according to their relative GLI2 mRNA and protein expression levels. GLI2 expression was reduced in a GLI2high cell line with lentiviral expression of short hairpin RNA targeting GLI2. We assessed the role of GLI2 in melanoma cell invasiveness in Matrigel assays. We measured secretion of matrix metalloproteinase (MMP)-2 and MMP-9 by gelatin zymography and expression of E-cadherin by western blotting and RT-PCR. The role of GLI2 in development of bone metastases was determined following intracardiac injection of melanoma cells in immunocompromised mice (n = 5-13). Human melanoma samples (n = 79) at various stages of disease progression were analyzed for GLI2 and E-cadherin expression by immunohistochemistry, in situ hybridization, or RT-PCR. All statistical tests were two-sided., Results: Among melanoma cell lines, increased GLI2 expression was associated with loss of E-cadherin expression and with increased capacity to invade Matrigel and to form bone metastases in mice (mean osteolytic tumor area: GLI2high vs GLI2low, 2.81 vs 0.93 mm(2), difference = 1.88 mm(2), 95% confidence interval [CI] = 1.16 to 2.60, P < .001). Reduction of GLI2 expression in melanoma cells that had expressed high levels of GLI2 substantially inhibited both basal and TGF-beta-induced cell migration, invasion (mean number of Matrigel invading cells: shGLI2 vs shCtrl (control), 52.6 vs 100, difference = 47.4, 95% CI = 37.0 to 57.8, P = .024; for shGLI2 + TGF-beta vs shCtrl + TGF-beta, 31.0 vs 161.9, difference = -130.9, 95% CI = -96.2 to -165.5, P = .002), and MMP secretion in vitro and the development of experimental bone metastases in mice. Within human melanoma lesions, GLI2 expression was heterogeneous, associated with tumor regions in which E-cadherin was lost and increased in the most aggressive tumors., Conclusion: GLI2 was directly involved in driving melanoma invasion and metastasis in this preclinical study.
- Published
- 2010
- Full Text
- View/download PDF
32. JNK supports survival in melanoma cells by controlling cell cycle arrest and apoptosis.
- Author
-
Alexaki VI, Javelaud D, and Mauviel A
- Subjects
- Animals, Anthracenes pharmacology, Apoptosis drug effects, Cell Cycle drug effects, Cell Proliferation drug effects, Cell Survival drug effects, Cell Survival physiology, Humans, JNK Mitogen-Activated Protein Kinases antagonists & inhibitors, Mice, Mice, Nude, Mitogen-Activated Protein Kinase 8 antagonists & inhibitors, Mitogen-Activated Protein Kinase 8 metabolism, Mitogen-Activated Protein Kinase 8 physiology, Mitogen-Activated Protein Kinase 9 antagonists & inhibitors, Mitogen-Activated Protein Kinase 9 metabolism, Mitogen-Activated Protein Kinase 9 physiology, RNA, Small Interfering pharmacology, Tumor Cells, Cultured, Apoptosis physiology, Cell Cycle physiology, JNK Mitogen-Activated Protein Kinases physiology, Melanoma pathology
- Abstract
JNK1/2 proteins belong to the family of stress-activated protein kinases. They play a complex role in growth regulation, inducing either cell death or growth support. In this report, we provide evidence that, in human melanoma cells, JNK inhibition with the small molecule inhibitor SP600125 induces either predominantly a G2/M arrest or apoptosis depending on the cell line. In 1205Lu cells, JNK inhibition induced cell cycle arrest through p53-dependent induction of p21 Cip1/Waf1 expression, while in WM983B cells, induction of apoptosis by JNK inhibition was accompanied by p53, Bad and Bax induction, not p21 Cip1/Waf1. JNK inhibition with the small molecule inhibitor SP600125 slowed growth of all cell lines, although the effect was markedly greater in cells exhibiting high phospho- (P-)JNK1 levels. Specific gene knockdown of JNK1 by means of siRNA oligonucleotides inhibited cell growth only in melanoma cell lines exhibiting high P-JNK1 levels. siRNAs directed against JNK2 did not reduce cell growth in any of the cell lines tested. Together, our findings demonstrate that JNK, and in particular the JNK1 isoform, support the growth of melanoma cells, by controlling either cell cycle progression or apoptosis depending on the cellular context.
- Published
- 2008
- Full Text
- View/download PDF
33. Transforming growth factor-beta suppresses the ability of Ski to inhibit tumor metastasis by inducing its degradation.
- Author
-
Le Scolan E, Zhu Q, Wang L, Bandyopadhyay A, Javelaud D, Mauviel A, Sun L, and Luo K
- Subjects
- Animals, DNA-Binding Proteins physiology, Female, Genes, Tumor Suppressor drug effects, Humans, Mice, Mice, Inbred BALB C, Mice, Nude, Nuclear Proteins physiology, Proto-Oncogene Proteins physiology, Smad Proteins physiology, Transplantation, Heterologous, Tumor Cells, Cultured, Ubiquitin-Protein Ligases, DNA-Binding Proteins antagonists & inhibitors, DNA-Binding Proteins metabolism, Neoplasm Metastasis pathology, Protein Processing, Post-Translational drug effects, Proto-Oncogene Proteins antagonists & inhibitors, Proto-Oncogene Proteins metabolism, Transforming Growth Factor beta pharmacology
- Abstract
c-Ski is an important corepressor of transforming growth factor-beta (TGF-beta) signaling through its ability to bind to and repress the activity of the Smad proteins. It was initially identified as an oncogene that promotes anchorage-independent growth of chicken and quail embryo fibroblasts when overexpressed. Although increased Ski expression is detected in many human cancer cells, the roles of Ski in mammalian carcinogenesis have yet to be defined. Here, we report that reducing Ski expression in breast and lung cancer cells does not affect tumor growth but enhances tumor metastasis in vivo. Thus, in these cells, Ski plays an antitumorigenic role. We also showed that TGF-beta, a cytokine that is often highly expressed in metastatic tumors, induces Ski degradation through the ubiquitin-dependent proteasome in malignant human cancer cells. On TGF-beta treatment, the E3 ubiquitin ligase Arkadia mediates degradation of Ski in a Smad-dependent manner. Although Arkadia interacts with Ski in the absence of TGF-beta, binding of phosphorylated Smad2 or Smad3 to Ski is required to induce efficient degradation of Ski by Arkadia. Our results suggest that the ability of TGF-beta to induce degradation of Ski could be an additional mechanism contributing to its protumorigenic activity.
- Published
- 2008
- Full Text
- View/download PDF
34. Transforming growth factor-beta in cutaneous melanoma.
- Author
-
Javelaud D, Alexaki VI, and Mauviel A
- Subjects
- Extracellular Signal-Regulated MAP Kinases metabolism, Humans, Melanoma drug therapy, Signal Transduction drug effects, Skin Neoplasms drug therapy, Smad Proteins metabolism, Transforming Growth Factor beta drug effects, Antineoplastic Agents pharmacology, Melanoma physiopathology, Skin Neoplasms physiopathology, Transforming Growth Factor beta physiology
- Abstract
Transforming growth factor-beta (TGF-beta) plays a complex role during carcinogenesis. It may either act as a tumor suppressor through its broad antiproliferative potential or as a tumor promoter either via direct effects on tumor cell aggressiveness or indirectly by modulating stromal responses, angiogenesis and immune surveillance. Increased production of TGF-beta by cancer cells is often associated with tumor grade. Melanoma cells largely escape cell cycle arrest normally induced by TGF-beta in normal melanocytes, yet produce active TGF-beta and are capable of efficient transcriptional responses to the growth factor. In this review, we summarize the current knowledge about the role played by TGF-beta in melanoma progression and hypothesize about the appropriateness of targeting TGF-beta signaling for therapeutic intervention.
- Published
- 2008
- Full Text
- View/download PDF
35. Stable overexpression of Smad7 in human melanoma cells impairs bone metastasis.
- Author
-
Javelaud D, Mohammad KS, McKenna CR, Fournier P, Luciani F, Niewolna M, André J, Delmas V, Larue L, Guise TA, and Mauviel A
- Subjects
- Animals, Bone Neoplasms genetics, Female, Humans, Melanoma metabolism, Mice, Mice, Nude, Neoplasm Metastasis genetics, Smad7 Protein metabolism, Survival Analysis, Transfection, Transforming Growth Factor beta antagonists & inhibitors, Transforming Growth Factor beta genetics, Transforming Growth Factor beta metabolism, Tumor Cells, Cultured, Bone Neoplasms secondary, Melanoma genetics, Melanoma pathology, Smad7 Protein genetics
- Abstract
Melanoma has a propensity to metastasize to bone, where it is exposed to high concentrations of transforming growth factor-beta (TGF-beta). Because TGF-beta promotes bone metastases from other solid tumors, such as breast cancer, we tested the role of TGF-beta in melanoma metastases to bone. 1205Lu melanoma cells, stably transfected to overexpress the natural TGF-beta/Smad signaling inhibitor Smad7, were studied in an experimental model of bone metastasis whereby tumor cells are inoculated into the left cardiac ventricle of nude mice. All mice bearing parental and mock-transfected 1205Lu cells developed osteolytic bone metastases 5 weeks post-tumor inoculation. Mice bearing 1205Lu-Smad7 tumors had significantly less osteolysis on radiographs and longer survival compared with parental and mock-transfected 1205Lu mice. To determine if the reduced bone metastases observed in mice bearing 1205Lu-Smad7 clones was due to reduced expression of TGF-beta target genes known to enhance metastases to bone from breast cancer cells, we analyzed gene expression of osteolytic factors, parathyroid hormone-related protein (PTHrP) and interleukin-11 (IL-11), the chemotactic receptor CXCR4, and osteopontin in 1205Lu cells. Quantitative reverse transcription-PCR analysis indicated that PTHrP, IL-11, CXCR4, and osteopontin mRNA steady-state levels were robustly increased in response to TGF-beta and that Smad7 and the TbetaRI small-molecule inhibitor, SB431542, prevented such induction. In addition, 1205Lu-Smad7 bone metastases expressed significantly lower levels of IL-11, connective tissue growth factor, and PTHrP. These data suggest that TGF-beta promotes osteolytic bone metastases due to melanoma by stimulating the expression of prometastatic factors via the Smad pathway. Blockade of TGF-beta signaling may be an effective treatment for melanoma metastasis to bone.
- Published
- 2007
- Full Text
- View/download PDF
36. Stable overexpression of Smad7 in human melanoma cells inhibits their tumorigenicity in vitro and in vivo.
- Author
-
Javelaud D, Delmas V, Möller M, Sextius P, André J, Menashi S, Larue L, and Mauviel A
- Subjects
- Animals, Cell Adhesion, Cell Movement, Gene Expression Profiling, Humans, Matrix Metalloproteinase 9 metabolism, Melanoma metabolism, Mice, Mice, Nude, Neoplasm Invasiveness, Neoplasm Metastasis, Signal Transduction, Skin Neoplasms metabolism, Smad7 Protein physiology, Transforming Growth Factor beta, Tumor Cells, Cultured, Melanoma pathology, Skin Neoplasms pathology, Smad7 Protein biosynthesis
- Abstract
We previously identified constitutive Smad signaling in human melanoma cells despite resistance to transforming growth factor-beta (TGF-beta) control of cell proliferation. This led us to investigate the effect of inhibitory Smad7 overexpression on melanoma cell behavior. Using the highly metastatic cell line, 1205-Lu, we thus generated melanoma cell clones constitutively expressing Smad7, and their mock-transfected counterparts. Stable expression of Smad7 resulted in an inhibition of constitutive Smad2/3 phosphorylation, and in a reduced TGF-beta response of Smad3/Smad4-driven gene transactivation, as measured using transfected Smad3/4-specific reporter gene constructs. Smad7 overexpression, however, did not alter their proliferative capacity and resistance to TGF-beta-driven growth inhibition. On the other hand, expression of Smad7 efficiently reduced the capacity of human melanoma cells to invade Matrigel in Boyden migration chambers, while not affecting their motility and adhesion to collagen and laminin. Gelatin zymography identified reduced MMP-2 and MMP-9 secretion by Smad7-expressing melanoma cells as compared with their control counterparts. Smad7-expressing melanoma cells exhibited a dramatically reduced capacity to form colonies under anchorage-independent culture conditions, and, when injected subcutaneously into nude mice, were largely delayed in their ability to form tumors. These results suggest that TGF-beta production by melanoma cells not only affects the tumor environment but also directly contributes to tumor cell aggressiveness through autocrine activation of Smad signaling.
- Published
- 2005
- Full Text
- View/download PDF
37. Crosstalk mechanisms between the mitogen-activated protein kinase pathways and Smad signaling downstream of TGF-beta: implications for carcinogenesis.
- Author
-
Javelaud D and Mauviel A
- Subjects
- Enzyme Activation, Humans, Smad Proteins, Transcription Factors physiology, DNA-Binding Proteins physiology, MAP Kinase Signaling System physiology, Neoplasms etiology, Signal Transduction physiology, Trans-Activators physiology, Transforming Growth Factor beta physiology
- Abstract
Transforming growth factor-beta (TGF-beta) superfamily members signal via membrane-bound heteromeric serine-threonine kinase receptor complexes. Upon ligand-binding, receptor activation leads to phosphorylation of cytoplasmic protein substrates of the Smad family. Following phosphorylation and oligomerization, the latter move into the nucleus to act as transcription factors to regulate target gene expression. TGF-beta responses are not solely the result of the activation Smad cascade, but are highly cell-type specific and dependent upon interactions of Smad signaling with a variety of other intracellular signaling mechanisms, initiated or not by TGF-beta itself, that may either potentiate, synergize, or antagonize, the rather linear TGF-beta/Smad pathway. These include, (a), regulation of Smad activity by mitogen-activated protein kinases (MAPKs), (b), nuclear interaction of activated Smads with transcriptional cofactors, whether coactivators or corepressors, that may be themselves be regulated by diverse signaling mechanisms, and (c), negative feedback loops exerted by inhibitory Smads, transcriptional targets of the Smad cascade. This review focuses on how MAPKs modulate the outcome of Smad activation by TGF-beta, and how cross-signaling mechanisms between the Smad and MAPK pathways may take place and affect cell fate in the context of carcinogenesis.
- Published
- 2005
- Full Text
- View/download PDF
38. NF-kappaB activation prevents apoptotic oxidative stress via an increase of both thioredoxin and MnSOD levels in TNFalpha-treated Ewing sarcoma cells.
- Author
-
Djavaheri-Mergny M, Javelaud D, Wietzerbin J, and Besançon F
- Subjects
- Adolescent, Antioxidants metabolism, Caspase 3, Caspases metabolism, Child, Enzyme Activation, Humans, JNK Mitogen-Activated Protein Kinases metabolism, Lipid Peroxidation, Reactive Oxygen Species metabolism, Tumor Cells, Cultured, Apoptosis physiology, NF-kappa B metabolism, Oxidative Stress, Sarcoma, Ewing metabolism, Superoxide Dismutase metabolism, Thioredoxins metabolism, Tumor Necrosis Factor-alpha metabolism
- Abstract
Repression of activation of c-Jun N-terminal kinase (JNK) participates in the anti-apoptotic effect of nuclear factor-kappaB (NF-kappaB) in TNFalpha-treated Ewing sarcoma cells. As oxidative stress is one of the most prominent activators of JNK, we investigated the relationship between TNFalpha-induced NF-kappaB activation and the control of oxidative stress. Inhibition of NF-kappaB activation resulted in an increase in TNFalpha-induced ROS production, lipid peroxidation and protein oxidation. Those ROS and lipid peroxides were both involved in TNFalpha-induced apoptosis, whereas only ROS elevation triggered sustained JNK activation. TNFalpha increased the level of two antioxidant enzymes, thioredoxin and manganese superoxide dismutase by an NF-kappaB-dependent mechanism. Inhibition of expression or activity of these enzymes sensitized cells to TNFalpha-induced apoptosis, indicating their functional role in protection from cell death. Thus, agents that inhibit activities of these enzymes may prove helpful in the treatment of Ewing tumors.
- Published
- 2004
- Full Text
- View/download PDF
39. TGF-beta-induced SMAD signaling and gene regulation: consequences for extracellular matrix remodeling and wound healing.
- Author
-
Schiller M, Javelaud D, and Mauviel A
- Subjects
- Animals, Humans, Smad Proteins, DNA-Binding Proteins metabolism, Extracellular Matrix physiology, Gene Expression Regulation, Signal Transduction physiology, Trans-Activators metabolism, Transforming Growth Factor beta physiology, Wound Healing physiology
- Abstract
Members of the transforming growth factor-beta (TGF-beta) superfamily are pleiotropic cytokines that have the ability to regulate numerous cell functions, including proliferation, differentiation, apoptosis, epithelial-mesenchymal transition, and production of extracellular matrix, allowing them to play an important role during embryonic development and for maintenance of tissue homeostasis. Three TGF-beta isoforms have been identified in mammals. They propagate their signal via a signal transduction network involving receptor serine/threonine kinases at the cell surface and their substrates, the SMAD proteins. Upon phosphorylation and oligomerization, the latter move into the nucleus to regulate transcription of target genes. This review will summarize recent advances in the understanding of the mechanisms underlying SMAD modulation of extracellular matrix gene expression in the context of wound healing and tissue fibrosis.
- Published
- 2004
- Full Text
- View/download PDF
40. Mammalian transforming growth factor-betas: Smad signaling and physio-pathological roles.
- Author
-
Javelaud D and Mauviel A
- Subjects
- Animals, DNA-Binding Proteins physiology, Disease etiology, Humans, Mice, Protein Isoforms metabolism, Protein Isoforms physiology, Protein Isoforms therapeutic use, Receptors, Transforming Growth Factor beta physiology, Smad Proteins, Trans-Activators physiology, Transforming Growth Factor beta metabolism, Transforming Growth Factor beta therapeutic use, DNA-Binding Proteins metabolism, Signal Transduction, Trans-Activators metabolism, Transforming Growth Factor beta physiology
- Abstract
Since its discovery in the early 1980s, transforming growth factor-beta (TGF-beta) has emerged as a family of growth factors involved in essential physiological processes, including embryonic development, differentiation, tissue repair and cell growth control. Knockout experiments for the three mammalian isoforms of TGF-betas in mice have demonstrated their importance in regulating inflammation and tissue repair. Also, TGF-beta has been implicated in the pathogenesis of human diseases, including tissue fibrosis and carcinogenesis where, in the latter case, it may exert both tumor suppressor and pro-oncogenic activities depending on the stage of the tumor. Cellular signaling by TGF-beta family members is initiated by the assembly of specific cell surface serine/threonine kinase type receptors that activate transcription factors of the Smad family.
- Published
- 2004
- Full Text
- View/download PDF
41. Amelioration of radiation-induced fibrosis: inhibition of transforming growth factor-beta signaling by halofuginone.
- Author
-
Xavier S, Piek E, Fujii M, Javelaud D, Mauviel A, Flanders KC, Samuni AM, Felici A, Reiss M, Yarkoni S, Sowers A, Mitchell JB, Roberts AB, and Russo A
- Subjects
- Animals, Blotting, Northern, Blotting, Western, COS Cells, Carcinoma, Squamous Cell drug therapy, Cell Line, Cell Line, Tumor, Cells, Cultured, DNA-Binding Proteins metabolism, Dose-Response Relationship, Drug, Down-Regulation, Fibrosis metabolism, Gene Deletion, Genes, Reporter, Humans, Immunoblotting, MAP Kinase Signaling System, Mice, Mice, Inbred C3H, Microscopy, Confocal, Microscopy, Fluorescence, Piperidines, Plasmids metabolism, Quinazolinones, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction, Smad3 Protein, Time Factors, Trans-Activators metabolism, Transforming Growth Factor beta1, Protein Synthesis Inhibitors therapeutic use, Quinazolines therapeutic use, Radiation Pneumonitis drug therapy, Transforming Growth Factor beta metabolism
- Abstract
Radiation-induced fibrosis is an untoward effect of high dose therapeutic and inadvertent exposure to ionizing radiation. Transforming growth factor-beta (TGF-beta) has been proposed to be critical in tissue repair mechanisms resulting from radiation injury. Previously, we showed that interruption of TGF-beta signaling by deletion of Smad3 results in resistance to radiation-induced injury. In the current study, a small molecular weight molecule, halofuginone (100 nm), is demonstrated by reporter assays to inhibit the TGF-beta signaling pathway, by Northern blotting to elevate inhibitory Smad7 expression within 15 min, and by Western blotting to inhibit formation of phospho-Smad2 and phospho-Smad3 and to decrease cytosolic and membrane TGF-beta type II receptor (TbetaRII). Attenuation of TbetaRII levels was noted as early as 1 h and down-regulation persisted for 24 h. Halofuginone blocked TGF-beta-induced delocalization of tight junction ZO-1, a marker of epidermal mesenchymal transition, in NMuMg mammary epithelial cells and suggest halofuginone may have in vivo anti-fibrogenesis characteristics. After documenting the in vitro cellular effects, halofuginone (intraperitoneum injection of 1, 2.5, or 5 microg/mouse/day) efficacy was assessed using ionizing radiation-induced (single dose, 35 or 45 Gy) hind leg contraction in C3H/Hen mice. Halofuginone treatment alone exerted no toxicity but significantly lessened radiation-induced fibrosis. The effectiveness of radiation treatment (2 gray/day for 5 days) of squamous cell carcinoma (SCC) tumors grown in C3H/Hen was not affected by halofuginone. The results detail the molecular effects of halofuginone on the TGF-beta signal pathway and show that halofuginone may lessen radiation-induced fibrosis in humans.
- Published
- 2004
- Full Text
- View/download PDF
42. Disruption of basal JNK activity differentially affects key fibroblast functions important for wound healing.
- Author
-
Javelaud D, Laboureau J, Gabison E, Verrecchia F, and Mauviel A
- Subjects
- Cell Movement genetics, Collagen physiology, Fibroblasts enzymology, Humans, MAP Kinase Kinase 4, Mitogen-Activated Protein Kinase Kinases genetics, Signal Transduction drug effects, Signal Transduction genetics, Transforming Growth Factor beta pharmacology, Fibroblasts physiology, JNK Mitogen-Activated Protein Kinases, Mitogen-Activated Protein Kinase Kinases physiology, Wound Healing genetics
- Abstract
We used both a gene knockout approach and pharmacologic modulation to study the implication of the JNK pathway in regulating fibroblast motility, capacity to contract mechanically unloaded collagen gels, and type I collagen gene expression in vitro. These parameters, which are important for tissue repair, are positively regulated by transforming growth factor (TGF)-beta, a cytokine viewed as playing a master role during wound healing. We demonstrate that basal JNK activity is critical for fibroblast motility because (a) mouse embryo jnk-/- fibroblasts exhibit significantly lower ability to close mechanically induced cell layer wounds than their wild-type (wt) counterparts, and (b) wound closure by human dermal fibroblasts is dramatically impaired by the specific JNK inhibitor SP600125. junAA fibroblasts, in which amino acids Ser63 and Ser73 of c-Jun are replaced by two Ala residues so that c-Jun cannot be phosphorylated by JNK, also exhibited impaired motility, suggesting that c-Jun phosphorylation by JNK is critical for fibroblast migration. In sharp contrast to their lesser motility on plastic, jnk-/- and junAA fibroblasts contracted free-floating, mechanically unloaded, collagen lattices markedly faster than wt fibroblasts. Furthermore, basal mRNA steady-state levels for types I and III collagen genes were similar in jnk-/- and wt fibroblasts. Likewise, overexpression of a dominant-negative mutant form of MKK4 in dermal fibroblasts did not affect collagen expression. We also demonstrate that basal JNK activity does not affect either TGF-beta-induced collagen gene expression or lattice contraction, whereas on the other hand, the blockage of motility initiated by JNK inhibition cannot be overcome by TGF-beta. Together these results demonstrate discrete, yet significant and highly specific, regulation of fibroblast functions important for wound healing by basal JNK activity.
- Published
- 2003
- Full Text
- View/download PDF
43. Inactivation of p21WAF1 sensitizes cells to apoptosis via an increase of both p14ARF and p53 levels and an alteration of the Bax/Bcl-2 ratio.
- Author
-
Javelaud D and Besancon F
- Subjects
- Base Sequence, Colonic Neoplasms, Cyclin-Dependent Kinase Inhibitor p21, DNA Primers, Gene Deletion, Genes, Reporter, Humans, Luciferases genetics, Recombinant Proteins metabolism, Tumor Cells, Cultured, bcl-2-Associated X Protein, Apoptosis physiology, Cyclins antagonists & inhibitors, Proto-Oncogene Proteins metabolism, Proto-Oncogene Proteins c-bcl-2 metabolism, Tumor Suppressor Protein p14ARF metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
p21(WAF1) appears to be a major determinant of the cell fate in response to anticancer therapy. It was shown previously that HCT116 human colon cancer cells growing in vitro enter a stable arrest upon DNA damage, whereas cells with a defective p21(WAF1) response undergo apoptosis. Here we report that the enhanced sensitivity of HCT116/p21(-/-) cells to chemotherapeutic drug-induced apoptosis correlates with an increased expression of p53 and a modification of their Bax/Bcl-2 ratio in favor of the pro-apoptotic protein Bax. Treatment of HCT116/p21(-/-) cells with daunomycin resulted in a reduction of the mitochondrial membrane potential and in activation of caspase-9, whereas no such changes were observed in HCT116/p21(+/+) cells, providing evidence that p21(WAF1) exerts an antagonistic effect on the mitochondrial pathway of apoptosis. Moreover, the role of p53 in activation of this pathway was demonstrated by the fact that inhibition of p53 activity by pifithrin-alpha reduced the sensitivity of HCT116/p21(-/-) cells to daunomycin-induced apoptosis and restored a Bax/Bcl-2 ratio similar to that observed in HCT116p21(+/+) cells. Enhancement of p53 expression after disruption of p21(WAF1) resulted from a stabilization of p53, which correlated with an increased expression of the tumor suppressor p14(ARF), an inhibitor of the ubiquitin ligase activity of Mdm2. In accordance with the role of p14(ARF) in p53 stabilization, overexpression of p14(ARF) in HCT116/p21(+/+) cells resulted in a strong increase in p53 activity. Our results identify a novel mechanism for the anti-apoptotic effect of p21(WAF1) consisting in maintenance of mitochondrial homeostasis that occurs in consequence of a negative control of p14(ARF) expression.
- Published
- 2002
- Full Text
- View/download PDF
44. Inhibition of constitutive NF-kappa B activity suppresses tumorigenicity of Ewing sarcoma EW7 cells.
- Author
-
Javelaud D, Poupon MF, Wietzerbin J, and Besançon F
- Subjects
- Angiogenesis Inducing Agents biosynthesis, Angiogenesis Inducing Agents genetics, Animals, Apoptosis, Bone Neoplasms etiology, Bone Neoplasms pathology, Cell Division, DNA-Binding Proteins genetics, Extracellular Matrix Proteins metabolism, Female, Genes, Reporter, Humans, Kinetics, Mice, Mice, Nude, NF-KappaB Inhibitor alpha, NF-kappa B physiology, RNA, Neoplasm biosynthesis, Sarcoma, Ewing etiology, Sarcoma, Ewing pathology, Transcription, Genetic, Transfection, Tumor Cells, Cultured, Tumor Necrosis Factor-alpha pharmacology, Bone Neoplasms metabolism, I-kappa B Proteins, NF-kappa B antagonists & inhibitors, Sarcoma, Ewing metabolism
- Abstract
Ewing sarcoma is 1 of the most aggressive tumors that can affect children and young adults. Despite advances in therapy, the prognosis remains poor emphasizing the need for defining new targets for treatment. We investigated a possible role of nuclear factor-kappa B (NF-kappa B) activity of Ewing sarcoma-derived EW7 cells in their tumorigenicity. In these cells, expression of a degradation-resistant form of the inhibitory factor I kappa B alpha inhibited NF-kappa B activity without affecting their in vitro proliferation rate. It causes, however, a remarkable loss of their ability to generate tumors in nude mice that correlates with both a decrease in extracellular matrix (ECM) protein secretion and an acquisition of sensitivity to murine tumor necrosis factor alpha (TNF alpha)-induced apoptosis. These data support the concept that NF-kappa B activity plays a role in the tumorigenicity of Ewing sarcoma cells, identifying NF-kappa B as a potential target for reducing Ewing tumor progression., (Copyright 2001 Wiley‐Liss, Inc.)
- Published
- 2002
- Full Text
- View/download PDF
45. NF-kappa B activation results in rapid inactivation of JNK in TNF alpha-treated Ewing sarcoma cells: a mechanism for the anti-apoptotic effect of NF-kappa B.
- Author
-
Javelaud D and Besançon F
- Subjects
- Bone Neoplasms enzymology, Caspases physiology, Cyclin-Dependent Kinase Inhibitor p21, Cyclins metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins physiology, Fungal Proteins, Humans, Kinetics, Mitogen-Activated Protein Kinase 8, Mitogen-Activated Protein Kinases genetics, Models, Biological, Mutation, NF-KappaB Inhibitor alpha, Phosphorylation, Sarcoma, Ewing enzymology, Transfection, Tumor Cells, Cultured, p38 Mitogen-Activated Protein Kinases, Apoptosis, Bone Neoplasms metabolism, I-kappa B Proteins, Mitogen-Activated Protein Kinases metabolism, NF-kappa B metabolism, Sarcoma, Ewing metabolism, Tumor Necrosis Factor-alpha pharmacology
- Abstract
We recently reported that inhibition of NF-kappa B activation as a consequence of the overexpression of a degradation-resistant form of I kappa B alpha [I kappa B alpha(A32/36)] sensitized Ewing sarcoma cells to TNF alpha-induced killing. The c-Jun N-terminal kinases (JNK) have been shown to participate in death signaling triggered by certain stimuli and are activated by TNF alpha. To obtain insight into the mechanism of the anti-apoptotic effect of NF-kappa B, we compared the profiles of JNK activation by TNF alpha in control cells and in cells in which NF-kappa B activation was impaired. We show here that JNK activation was transient in control cells but remained elevated in I kappa B alpha(A32/36)-expressing cells. NF-kappa B repressed specifically the JNK pathway, since the kinetics of activation of the other TNF alpha-activated-MAP kinase p38 were identical in both cells. Prolongation of JNK activation in I kappa B alpha(A32/36)-expressing cells was not inhibited by the broad spectrum caspase inhibitor Z-VAD-FMK and thus was not the consequence of caspase activation. Pretreatment of control cells with the phosphatase inhibitor vanadate greatly prolonged JNK activation by TNF alpha and resulted in induction of apoptosis by this cytokine. Moreover, overexpression of a dominant-negative mutant of JNK1 decreased TNF alpha-induced apoptosis in cells expressing the super repressor of NF-kappa B, indicating that the sustained activation of JNK1 participated in death signaling triggered by TNF alpha. Our results provide evidence that the repression of JNK activation by NF-kappa B participates in the anti-apoptotic effect of this transcription factor in TNF alpha-treated Ewing sarcoma cells.
- Published
- 2001
- Full Text
- View/download PDF
46. Induction of p21Waf1/Cip1 by TNFalpha requires NF-kappaB activity and antagonizes apoptosis in Ewing tumor cells.
- Author
-
Javelaud D, Wietzerbin J, Delattre O, and Besançon F
- Subjects
- Antineoplastic Agents pharmacology, Cyclin-Dependent Kinase Inhibitor p21, DNA-Binding Proteins analysis, Humans, NF-KappaB Inhibitor alpha, Tumor Cells, Cultured, Apoptosis drug effects, Cyclins physiology, I-kappa B Proteins, NF-kappa B physiology, Sarcoma, Ewing pathology, Tumor Necrosis Factor-alpha pharmacology
- Abstract
The Ewing family of tumors is characterized by recurrent reciprocal translocations that generate chimeric proteins, either EWS - FLI-1 or EWS - ERG. These proteins are potent transcriptional activators and are responsible for maintaining the oncogenic properties of tumor cells. Since apoptosis appears to be the main mechanism whereby chemotherapy and radiation kill tumor cells, identification of events that can antagonize apoptosis in Ewing tumors is essential for improving their response to conventional therapies. Here, we report that the transcriptional factor NF-kappaB is a survival factor for Ewing tumor-derived cells. In fact, inhibition of NF-kappaB activation as a consequence of the overexpression of a degradation-resistant form of IkappaBalpha, IkappaBalpha (A32/36), sensitized these cells to TNFalpha-induced killing. Although treatment with TNFalpha did not modify the cellular expression of Bcl-2, c-IAP1, c-IAP2, p53 and EWS - FLI-1 proteins, it increased p21Waf1/Cip1 levels. This induction required NF-kappaB activation since it was not observed in the IkappaBalpha (A32/36) expressing cells. Moreover, overexpression of p21Waf1/Cip1 in these IkappaBalpha (A32/36)-expressing cells, in which NF-kappaB and consequently p21Waf1/Cip1 are no longer inducible by TNFalpha, decreased their susceptibility to TNFalpha-induced killing. Our results therefore identify p21Waf1/Cip1 as a mediator of the antiapoptotic effect of TNFalpha-induced NF-kappaB in Ewing tumor cells.
- Published
- 2000
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.