13 results on '"Beguelin W"'
Search Results
2. Progestin-induced caveolin-1 expression mediates breast cancer cell proliferation
- Author
-
Salatino, M, Beguelin, W, Peters, M G, Carnevale, R, Proietti, C J, Galigniana, M D, Vedoy, C G, Schillaci, R, Charreau, E H, Sogayar, M C, and Elizalde, P V
- Published
- 2006
- Full Text
- View/download PDF
3. Progestins Induce Breast Cancer Cell Proliferation through the AP-1 Transcription Factor.
- Author
-
Flaque, MC Diaz, primary, Beguelin, W, additional, Proietti, CJ, additional, Rivas, MA, additional, Tkach, M, additional, Charreau, EH, additional, Schillaci, R, additional, and Elizalde, PV, additional
- Published
- 2010
- Full Text
- View/download PDF
4. Tumor necrosis factor transactivates ErbB2 in breast cancer cells.
- Author
-
Rivas, MA, primary, Tkach, M, additional, Proietti, CJ, additional, Rosemblit, C, additional, Beguelin, W, additional, Sundblad, V, additional, Díaz Flaqué, MC, additional, Charreau, EH, additional, Elizalde, PV, additional, and Schillaci, R, additional
- Published
- 2009
- Full Text
- View/download PDF
5. Clinical relevance of ErbB-2/HER2 nuclear expression in breast cancer
- Author
-
Schillaci Roxana, Guzmán Pablo, Cayrol Florencia, Beguelin Wendy, Díaz Flaqué María C, Proietti Cecilia J, Pineda Viviana, Palazzi Jorge, Frahm Isabel, Charreau Eduardo H, Maronna Esteban, Roa Juan C, and Elizalde Patricia V
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background The biological relevance of nuclear ErbB-2/HER2 (NuclErbB-2) presence in breast tumors remains unexplored. In this study we assessed the clinical significance of ErbB-2 nuclear localization in primary invasive breast cancer. The reporting recommendations for tumor marker prognostic studies (REMARK) guidelines were used as reference. Methods Tissue microarrays from a cohort of 273 primary invasive breast carcinomas from women living in Chile, a Latin American country, were examined for membrane (MembErbB-2) and NuclErbB-2 expression by an immunofluorescence (IF) protocol we developed. ErbB-2 expression was also evaluated by immunohistochemistry (IHC) with a series of antibodies. Correlation between NuclErbB-2 and MembErbB-2, and between NuclErbB-2 and clinicopathological characteristics of tumors was studied. The prognostic value of NuclErbB-2 in overall survival (OS) was evaluated using Kaplan-Meier method, and Cox model was used to explore NuclErbB-2 as independent prognostic factor for OS. Results The IF protocol we developed showed significantly higher sensitivity for detection of NuclErbB-2 than IHC procedures, while its specificity and sensitivity to detect MembErbB-2 were comparable to those of IHC procedures. We found 33.6% NuclErbB-2 positivity, 14.2% MembErbB-2 overexpression by IF, and 13.0% MembErbB-2 prevalence by IHC in our cohort. We identified NuclErbB-2 positivity as a significant independent predictor of worse OS in patients with MembErbB-2 overexpression. NuclErbB-2 was also a biomarker of lower OS in tumors that overexpress MembErbB-2 and lack steroid hormone receptors. Conclusions We revealed a novel role for NuclErbB-2 as an independent prognostic factor of poor clinical outcome in MembErbB-2-positive breast tumors. Our work indicates that patients presenting NuclErbB-2 may need new therapeutic strategies involving specific blockage of ErbB-2 nuclear migration.
- Published
- 2012
- Full Text
- View/download PDF
6. Modular Immune Organoids with Integrin Ligand Specificity Differentially Regulate Ex Vivo B Cell Activation.
- Author
-
Purwada A, Shah SB, Beguelin W, Melnick AM, and Singh A
- Abstract
Germinal centers are dynamic structures within lymphoid tissues, which develop once B cells receive activating signals from surrounding immune cells. Germinal center B cells are small in number, heterogeneous, and prone to rapid apoptosis unless selected by the body to form memory B cells. Despite extensive research in the B cell differentiation process, the role of the lymphoid niche, in particular integrin ligands, in the development of early germinal center-like phenotype remains unclear. Here, we report a biomaterials-based modular immune organoid that enables development of early germinal-center phenotype in an integrin ligand-specific manner. We demonstrate the differential role of integrin α4β1- and αvβ3-binding ligands in the induction of GL7+ (GC-like) and GL7- (non-GC-like) phenotype in differentiating B cells while in the presence of CD40 ligand and interleukin-4. We further demonstrate the role of integrin ligand specificities in clustering of β3 integrin and B cell receptor on the surface of differentiated B cells in 3D organoids as compared to the classic 2D cocultures. The study demonstrates that biomaterials-based immune organoids represent an ex vivo platform technology, which recapitulates certain aspects of GC biology to understand the process of B cell differentiation and induction of immunological responses. This platform is particularly useful in understanding the role of selective biomolecular signals and the temporal dependency of immune responses to these signals.
- Published
- 2017
- Full Text
- View/download PDF
7. Rationally designed BCL6 inhibitors target activated B cell diffuse large B cell lymphoma.
- Author
-
Cardenas MG, Yu W, Beguelin W, Teater MR, Geng H, Goldstein RL, Oswald E, Hatzi K, Yang SN, Cohen J, Shaknovich R, Vanommeslaeghe K, Cheng H, Liang D, Cho HJ, Abbott J, Tam W, Du W, Leonard JP, Elemento O, Cerchietti L, Cierpicki T, Xue F, MacKerell AD Jr, and Melnick AM
- Subjects
- Animals, Cell Line, Tumor, Doxorubicin pharmacology, Drug Screening Assays, Antitumor, HEK293 Cells, Humans, Indoles pharmacology, Ligands, Lymphoma, Large B-Cell, Diffuse pathology, Magnetic Resonance Spectroscopy, Male, Mice, Mice, SCID, Neoplasm Transplantation, Protein Binding, Proto-Oncogene Proteins c-bcl-6 metabolism, Thiazolidinediones pharmacology, Translocation, Genetic, Antineoplastic Agents pharmacology, Drug Design, Gene Expression Regulation, Neoplastic, Lymphoma, Large B-Cell, Diffuse drug therapy, Proto-Oncogene Proteins c-bcl-6 antagonists & inhibitors
- Abstract
Diffuse large B cell lymphomas (DLBCLs) arise from proliferating B cells transiting different stages of the germinal center reaction. In activated B cell DLBCLs (ABC-DLBCLs), a class of DLBCLs that respond poorly to current therapies, chromosomal translocations and amplification lead to constitutive expression of the B cell lymphoma 6 (BCL6) oncogene. The role of BCL6 in maintaining these lymphomas has not been investigated. Here, we designed small-molecule inhibitors that display higher affinity for BCL6 than its endogenous corepressor ligands to evaluate their therapeutic efficacy for targeting ABC-DLBCL. We used an in silico drug design functional-group mapping approach called SILCS to create a specific BCL6 inhibitor called FX1 that has 10-fold greater potency than endogenous corepressors and binds an essential region of the BCL6 lateral groove. FX1 disrupted formation of the BCL6 repression complex, reactivated BCL6 target genes, and mimicked the phenotype of mice engineered to express BCL6 with corepressor binding site mutations. Low doses of FX1 induced regression of established tumors in mice bearing DLBCL xenografts. Furthermore, FX1 suppressed ABC-DLBCL cells in vitro and in vivo, as well as primary human ABC-DLBCL specimens ex vivo. These findings indicate that ABC-DLBCL is a BCL6-dependent disease that can be targeted by rationally designed inhibitors that exceed the binding affinity of natural BCL6 ligands.
- Published
- 2016
- Full Text
- View/download PDF
8. Hematopoietic stem cell origin of BRAFV600E mutations in hairy cell leukemia.
- Author
-
Chung SS, Kim E, Park JH, Chung YR, Lito P, Teruya-Feldstein J, Hu W, Beguelin W, Monette S, Duy C, Rampal R, Telis L, Patel M, Kim MK, Huberman K, Bouvier N, Berger MF, Melnick AM, Rosen N, Tallman MS, Park CY, and Abdel-Wahab O
- Subjects
- Animals, Humans, Leukemia, Hairy Cell pathology, Mice, Hematopoietic Stem Cells pathology, Leukemia, Hairy Cell genetics, Mutation, Proto-Oncogene Proteins B-raf genetics
- Abstract
Hairy cell leukemia (HCL) is a chronic lymphoproliferative disorder characterized by somatic BRAFV600E mutations. The malignant cell in HCL has immunophenotypic features of a mature B cell, but no normal counterpart along the continuum of developing B lymphocytes has been delineated as the cell of origin. We find that the BRAFV600E mutation is present in hematopoietic stem cells (HSCs) in HCL patients, and that these patients exhibit marked alterations in hematopoietic stem/progenitor cell (HSPC) frequencies. Quantitative sequencing analysis revealed a mean BRAFV600E-mutant allele frequency of 4.97% in HSCs from HCL patients. Moreover, transplantation of BRAFV600E-mutant HSCs from an HCL patient into immunodeficient mice resulted in stable engraftment of BRAFV600E-mutant human hematopoietic cells, revealing the functional self-renewal capacity of HCL HSCs. Consistent with the human genetic data, expression of BRafV600E in murine HSPCs resulted in a lethal hematopoietic disorder characterized by splenomegaly, anemia, thrombocytopenia, increased circulating soluble CD25, and increased clonogenic capacity of B lineage cells-all classic features of human HCL. In contrast, restricting expression of BRafV600E to the mature B cell compartment did not result in disease. Treatment of HCL patients with vemurafenib, an inhibitor of mutated BRAF, resulted in normalization of HSPC frequencies and increased myeloid and erythroid output from HSPCs. These findings link the pathogenesis of HCL to somatic mutations that arise in HSPCs and further suggest that chronic lymphoid malignancies may be initiated by aberrant HSCs., (Copyright © 2014, American Association for the Advancement of Science.)
- Published
- 2014
- Full Text
- View/download PDF
9. p42/p44 MAPK-mediated Stat3Ser727 phosphorylation is required for progestin-induced full activation of Stat3 and breast cancer growth.
- Author
-
Tkach M, Rosemblit C, Rivas MA, Proietti CJ, Díaz Flaqué MC, Mercogliano MF, Beguelin W, Maronna E, Guzmán P, Gercovich FG, Deza EG, Elizalde PV, and Schillaci R
- Subjects
- Animals, Breast Neoplasms metabolism, Cell Proliferation, Cyclin D1 metabolism, Female, Humans, Mice, Mice, Inbred BALB C, Phosphorylation, STAT3 Transcription Factor genetics, Medroxyprogesterone Acetate pharmacology, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3 metabolism, STAT3 Transcription Factor metabolism
- Abstract
Stat3 is a signaling node for multiple oncogenic pathways and is therefore frequently active in breast cancer. As experimental and clinical evidence reveals that progestins are key players in controlling mammary gland tumorigenesis, we studied Stat3 participation in this event. We have previously shown that progestins induce Stat3Tyr705 phosphorylation and its transcriptional activation in breast cancer cells. In this study, we demonstrate that progestins also induce Stat3 phosphorylation at Ser727 residue, which occurs via activation of c-Src/p42/p44 MAPK pathways in murine progestin-dependent C4HD cells and in T-47D cells. Expression of a Stat3S727A vector, which carries a serine-to-alanine substitution at codon 727, shows that Stat3Ser727 phosphorylation is required for full transcriptional activation of cyclin D1 gene expression by progestins and for in vivo Stat3 recruitment on cyclin D1 promoter. Transfection of Stat3S727A in murine and human breast cancer cells abolished progestin-induced in vitro and in vivo growth. Moreover, we found a positive correlation between progesterone receptor expression and nuclear localization of Stat3Ser727 phosphorylation in breast cancer biopsies. These data highlight Stat3 phosphorylation in Ser727 residue as a nongenomic action by progestins, necessary to promote breast cancer growth.
- Published
- 2013
- Full Text
- View/download PDF
10. Targeting Stat3 induces senescence in tumor cells and elicits prophylactic and therapeutic immune responses against breast cancer growth mediated by NK cells and CD4+ T cells.
- Author
-
Tkach M, Coria L, Rosemblit C, Rivas MA, Proietti CJ, Díaz Flaqué MC, Beguelin W, Frahm I, Charreau EH, Cassataro J, Elizalde PV, and Schillaci R
- Subjects
- Animals, CD4-Positive T-Lymphocytes metabolism, CD4-Positive T-Lymphocytes pathology, Cell Line, Tumor, Chemokines metabolism, Cytokines metabolism, Disease Models, Animal, Female, Killer Cells, Natural metabolism, Killer Cells, Natural pathology, Mammary Neoplasms, Animal pathology, Mice, Mice, Inbred BALB C, Mice, Nude, Primary Cell Culture, STAT3 Transcription Factor, CD4-Positive T-Lymphocytes immunology, Cellular Senescence immunology, Gene Targeting methods, Killer Cells, Natural immunology, Mammary Neoplasms, Animal immunology, Mammary Neoplasms, Animal therapy
- Abstract
Aberrant Stat3 activation and signaling contribute to malignant transformation by promoting cell cycle progression, inhibiting apoptosis, and mediating tumor immune evasion. Stat3 inhibition in tumor cells induces the expression of chemokines and proinflammatory cytokines, so we proposed to apply Stat3-inhibited breast cancer cells as a source of immunogens to induce an antitumor immune response. Studies were performed in two murine breast cancer models in which Stat3 is activated: progestin-dependent C4HD cells and 4T1 cells. We immunized BALB/c mice with irradiated cancer cells previously transfected with a dominant-negative Stat3 vector (Stat3Y705F) in either a prophylactic or a therapeutic manner. Prophylactic administration of breast cancer cells transfected with Stat3Y705F (Stat3Y705F-breast cancer cells) inhibited primary tumor growth compared with administration of empty vector-transfected cells in both models. In the 4T1 model, 50% of the challenged mice were tumor free, and the incidence of metastasis decreased by 90%. In vivo assays of C4HD tumors showed that the antitumor immune response involves the participation of CD4(+) T cells and cytotoxic NK cells. Therapeutic immunization with Stat3Y705F-breast cancer cells inhibited tumor growth, promoted tumor cell differentiation, and decreased metastasis. Furthermore, inhibition of Stat3 activation in breast cancer cells induced cellular senescence, contributing to their immunogenic phenotype. In this work, we provide preclinical proof of concept that ablating Stat3 signaling in breast cancer cells results in an effective immunotherapy against breast cancer growth and metastasis. Moreover, our findings showing that Stat3 inactivation results in induction of a cellular senescence program disclose a potential mechanism for immunotherapy research.
- Published
- 2012
- Full Text
- View/download PDF
11. Transactivation of ErbB-2 induced by tumor necrosis factor alpha promotes NF-kappaB activation and breast cancer cell proliferation.
- Author
-
Rivas MA, Tkach M, Beguelin W, Proietti CJ, Rosemblit C, Charreau EH, Elizalde PV, and Schillaci R
- Subjects
- Animals, Breast Neoplasms pathology, Cell Division, Cell Line, Tumor drug effects, Cell Line, Tumor metabolism, Dimerization, Female, Humans, Mammary Neoplasms, Experimental genetics, Mammary Neoplasms, Experimental metabolism, Mammary Neoplasms, Experimental pathology, Mice, Mice, Inbred BALB C, Neoplasm Invasiveness, Neoplasm Proteins genetics, Phosphorylation, Protein Kinases physiology, Protein Processing, Post-Translational, RNA, Small Interfering pharmacology, Receptor, ErbB-2 genetics, Signal Transduction drug effects, Signal Transduction genetics, Breast Neoplasms genetics, Genes, erbB-2, NF-kappa B metabolism, Neoplasm Proteins biosynthesis, Receptor, ErbB-2 biosynthesis, Receptor, ErbB-2 physiology, Transcriptional Activation, Tumor Necrosis Factor-alpha physiology
- Abstract
Tumor necrosis factor alpha (TNFalpha) is a pleiotropic cytokine which, acting locally, induces tumor growth. Accumulating evidence, including our findings, showed that TNFalpha is mitogenic in breast cancer cells in vitro and in vivo. In the present study, we explored TNFalpha involvement on highly aggressive ErbB-2-overexpressing breast cancer cells. We found that TNFalpha induces ErbB-2 phosphorylation in mouse breast cancer C4HD cells and in the human breast cancer cell lines SK-BR-3 and BT-474. ErbB-2 phosphorylation at Tyr877 residue was mediated by TNFalpha-induced c-Src activation. Moreover, TNFalpha promoted ErbB-2/ErbB-3 heterocomplex formation, Akt activation and NF-kappaB transcriptional activation. Inhibition of ErbB-2 by addition of AG825, an epidermal growth factor receptor/ErbB-2-tyrosine kinase inhibitor, or knockdown of ErbB-2 by RNA interference strategy, blocked TNFalpha-induced NF-kappaB activation and proliferation. However, the humanized monoclonal antibody anti-ErbB-2 Herceptin could not inhibit TNFalpha ability to promote breast cancer growth. Interestingly, our work disclosed that TNFalpha is able to transactivate ErbB-2 and use it as an obligatory downstream signaling molecule in the generation of mitogenic signals. As TNFalpha has been shown to be present in the tumor microenvironment of a significant proportion of human infiltrating breast cancers, our findings would have clinical implication in ErbB-2-positive breast cancer treatment.
- Published
- 2010
- Full Text
- View/download PDF
12. Activation of Stat3 by heregulin/ErbB-2 through the co-option of progesterone receptor signaling drives breast cancer growth.
- Author
-
Proietti CJ, Rosemblit C, Beguelin W, Rivas MA, Díaz Flaqué MC, Charreau EH, Schillaci R, and Elizalde PV
- Subjects
- Animals, Female, Mammary Neoplasms, Animal etiology, Mice, Mice, Inbred BALB C, Signal Transduction, Cell Proliferation, Mammary Neoplasms, Animal pathology, Neuregulin-1 metabolism, Receptor, ErbB-2 metabolism, Receptors, Progesterone metabolism, STAT3 Transcription Factor metabolism
- Abstract
Cross talk between the steroid hormone receptors for estrogen and progesterone (PR) and the ErbB family of receptor tyrosine kinases appears to be a hallmark of breast cancer growth, but its underlying mechanism remains poorly explored. Here we have highlighted signal transducer and activator of transcription 3 (Stat3) as a key protein activated by heregulin (HRG), a ligand of the ErbB receptors, through co-opted, ligand-independent PR function as a signaling molecule. Stat3 activation was an absolute requirement in HRG-induced mammary tumor growth, and targeting Stat3 effectively inhibited growth of breast cancer cells with activated HRG/ErbB-2 and PR. Our findings unravel a novel potential therapeutic intervention in PR- and ErbB-2-positive breast tumors, involving the specific blockage of PR signaling activity.
- Published
- 2009
- Full Text
- View/download PDF
13. TNF alpha acting on TNFR1 promotes breast cancer growth via p42/P44 MAPK, JNK, Akt and NF-kappa B-dependent pathways.
- Author
-
Rivas MA, Carnevale RP, Proietti CJ, Rosemblit C, Beguelin W, Salatino M, Charreau EH, Frahm I, Sapia S, Brouckaert P, Elizalde PV, and Schillaci R
- Subjects
- Animals, Apoptosis Regulatory Proteins drug effects, Apoptosis Regulatory Proteins genetics, Apoptosis Regulatory Proteins metabolism, Carcinogens, Carcinoma, Ductal, Breast chemically induced, Carcinoma, Ductal, Breast drug therapy, Cell Line, Tumor, Female, Humans, JNK Mitogen-Activated Protein Kinases drug effects, JNK Mitogen-Activated Protein Kinases metabolism, Mammary Neoplasms, Experimental chemically induced, Mammary Neoplasms, Experimental drug therapy, Medroxyprogesterone Acetate, Mice, Mice, Inbred BALB C, Mitogen-Activated Protein Kinase 1 drug effects, Mitogen-Activated Protein Kinase 1 metabolism, Mitogen-Activated Protein Kinase 3 drug effects, Mitogen-Activated Protein Kinase 3 metabolism, NF-kappa B antagonists & inhibitors, NF-kappa B metabolism, Neoplasms, Hormone-Dependent chemically induced, Neoplasms, Hormone-Dependent drug therapy, Nitriles pharmacology, Proto-Oncogene Proteins c-akt drug effects, Proto-Oncogene Proteins c-akt metabolism, Receptors, Tumor Necrosis Factor, Type I immunology, Signal Transduction immunology, Sulfones pharmacology, Transcriptional Activation drug effects, Transcriptional Activation immunology, Carcinoma, Ductal, Breast physiopathology, Cell Proliferation drug effects, Mammary Neoplasms, Experimental physiopathology, Neoplasms, Hormone-Dependent physiopathology, Receptors, Tumor Necrosis Factor, Type I drug effects, Signal Transduction drug effects, Tumor Necrosis Factor-alpha pharmacology
- Abstract
Tumor necrosis factor alpha (TNF alpha) enhances proliferation of chemically-induced mammary tumors and of T47D human cell line through not fully understood pathways. Here, we explored the intracellular signaling pathways triggered by TNF alpha, the participation of TNF alpha receptor (TNFR) 1 and TNFR2 and the molecular mechanism leading to breast cancer growth. We demonstrate that TNFalpha induced proliferation of C4HD murine mammary tumor cells and of T47D cells through the activation of p42/p44 MAPK, JNK, PI3-K/Akt pathways and nuclear factor-kappa B (NF-kappa B) transcriptional activation. A TNF alpha-specific mutein selectively binding to TNFR1 induced p42/p44 MAPK, JNK, Akt activation, NF-kappa B transcriptional activation and cell proliferation, just like wild-type TNF alpha, while a mutein selective for TNFR2 induced only p42/p44 MAPK activation. Interestingly, blockage of TNFR1 or TNFR2 with specific antibodies was enough to impair TNF alpha signaling and biological effect. Moreover, in vivo TNF alpha administration supported C4HD tumor growth. We also demonstrated, for the first time, that injection of a selective inhibitor of NF-kappa B activity, Bay 11-7082, resulted in regression of TNF alpha-promoted tumor. Bay 11-7082 blocked TNF alpha capacity to induce cell proliferation and up-regulation of cyclin D1 and of Bcl-xLin vivo and in vitro. Our results reveal evidence for TNF alpha as a breast tumor promoter, and provide novel data for a future therapeutic approach using TNF alpha antagonists and NF-kappa B pharmacological inhibitors in established breast cancer treatment.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.