69 results on '"Elizalde PV"'
Search Results
2. Abstract P6-20-14: Neutralizing soluble tumor necrosis factor alpha overcomes trastuzumab-resistant breast cancer immune evasion by downregulating mucin 4, improving NK cell function and decreasing myeloid-derived suppressor cells in tumor microenvironment
- Author
-
Schillaci, R, primary, Bruni, S, additional, De Martino, M, additional, Mercogliano, MF, additional, Inurrigarro, G, additional, Frahm, I, additional, Proietti, CJ, additional, and Elizalde, PV, additional
- Published
- 2019
- Full Text
- View/download PDF
3. Abstract P5-03-02: Progestin Activation of p42/p44 MAPK Induces Stat3 Phosphorylation at Serine 727 Promoting Breast Cancer Growth
- Author
-
Tkach, M, primary, Rosemblit, C, additional, Béguelin, W, additional, Proietti, C, additional, Rivas, MA, additional, Diaz Flaqué, MC, additional, Cayrol, F, additional, Charreau, EH, additional, Elizalde, PV, additional, and Schillaci, R., 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. Correction: Stat3 regulates ErbB-2 expression and co-opts ErbB-2 nuclear function to induce miR-21 expression, PDCD4 downregulation and breast cancer metastasis.
- Author
-
Venturutti L, Romero LV, Urtreger AJ, Chervo MF, Russo RIC, Mercogliano MF, Inurrigarro G, Pereyra MG, Proietti CJ, Izzo F, Díaz Flaqué MC, Sundblad V, Roa JC, Guzmán P, de Kier Joffé EDB, Charreau EH, Schillaci R, and Elizalde PV
- Published
- 2024
- Full Text
- View/download PDF
6. Correction: Halting ErbB-2 isoforms retrograde transport to the nucleus as a new theragnostic approach for triple-negative breast cancer.
- Author
-
Madera S, Izzo F, Chervo MF, Dupont A, Chiauzzi VA, Bruni S, Petrillo E, Merin SS, De Martino M, Montero D, Levit C, Lebersztein G, Anfuso F, Roldán Deamicis A, Mercogliano MF, Proietti CJ, Schillaci R, Elizalde PV, and Cordo Russo RI
- Published
- 2023
- Full Text
- View/download PDF
7. Corrigendum to "Novel role of signal transducer and activator of transcription 3 as a progesterone receptor coactivator in breast cancer" [Steroids 76 (2011) 381-392].
- Author
-
Proietti CJ, Béguelin W, Díaz Flaqué MC, Cayrol F, Rivas MA, Tkach M, Charreau EH, Schillaci R, and Elizalde PV
- Published
- 2023
- Full Text
- View/download PDF
8. Correction: MiR-16 mediates trastuzumab and lapatinib response in ErbB-2-positive breast and gastric cancer via its novel targets CCNJ and FUBP1.
- Author
-
Venturutti L, Russo RIC, Rivas MA, Mercogliano MF, Izzo F, Oakley RH, Pereyra MG, De Martino M, Proietti CJ, Yankilevich P, Roa JC, Guzmán P, Cortese E, Allemand DH, Huang TH, Charreau EH, Cidlowski JA, Schillaci R, and Elizalde PV
- Published
- 2023
- Full Text
- View/download PDF
9. Retraction Note: Progesterone receptor assembly of a transcriptional complex along with activator protein 1, signal transducer and activator of transcription 3 and ErbB-2 governs breast cancer growth and predicts response to endocrine therapy.
- Author
-
Flaqué MCD, Galigniana NM, Béguelin W, Vicario R, Proietti CJ, Russo RC, Rivas MA, Tkach M, Guzmán P, Roa JC, Maronna E, Pineda V, Muñoz S, Mercogliano MF, Charreau EH, Yankilevich P, Schillaci R, and Elizalde PV
- Published
- 2023
- Full Text
- View/download PDF
10. Correction: Targeting ErbB-2 nuclear localization and function inhibits breast cancer growth and overcomes trastuzumab resistance.
- Author
-
Russo RIC, Béguelin W, Flaqué MCD, Proietti CJ, Venturutti L, Galigniana N, Tkach M, Guzmán P, Roa JC, O'Brien NA, Charreau EH, Schillaci R, and Elizalde PV
- Published
- 2023
- Full Text
- View/download PDF
11. Correction: Activation of ErbB-2 via a hierarchical interaction between ErbB-2 and type I insulin-like growth factor receptor in mammary tumor cells.
- Author
-
Balañá ME, Labriola L, Salatino M, Movsichoff F, Peters G, Charreau EH, and Elizalde PV
- Published
- 2023
- Full Text
- View/download PDF
12. Blocking soluble TNFα sensitizes HER2-positive breast cancer to trastuzumab through MUC4 downregulation and subverts immunosuppression.
- Author
-
Bruni S, Mauro FL, Proietti CJ, Cordo-Russo RI, Rivas MA, Inurrigarro G, Dupont A, Rocha D, Fernández EA, Deza EG, Lopez Della Vecchia D, Barchuk S, Figurelli S, Lasso D, Friedrich AD, Santilli MC, Regge MV, Lebersztein G, Levit C, Anfuso F, Castiglione T, Elizalde PV, Mercogliano MF, and Schillaci R
- Subjects
- Mice, Animals, Trastuzumab pharmacology, Trastuzumab therapeutic use, Down-Regulation, Tumor Necrosis Factor-alpha metabolism, Receptor, ErbB-2, Cell Line, Tumor, Immunosuppression Therapy, Mucin-4 genetics, Mucin-4 metabolism, Neoplasms drug therapy
- Abstract
Background: The success of HER2-positive (HER2+) breast cancer treatment with trastuzumab, an antibody that targets HER2, relies on immune response. We demonstrated that TNFα induces mucin 4 (MUC4) expression, which shields the trastuzumab epitope on the HER2 molecule decreasing its therapeutic effect. Here, we used mouse models and samples from HER2+ breast cancer patients to unravel MUC4 participation in hindering trastuzumab effect by fostering immune evasion., Methods: We used a dominant negative TNFα inhibitor (DN) selective for soluble TNFα (sTNFα) together with trastuzumab. Preclinical experiments were performed using two models of conditionally MUC4-silenced tumors to characterize the immune cell infiltration. A cohort of 91 patients treated with trastuzumab was used to correlate tumor MUC4 with tumor-infiltrating lymphocytes., Results: In mice bearing de novo trastuzumab-resistant HER2+ breast tumors, neutralizing sTNFα with DN induced MUC4 downregulation. Using the conditionally MUC4-silenced tumor models, the antitumor effect of trastuzumab was reinstated and the addition of TNFα-blocking agents did not further decrease tumor burden. DN administration with trastuzumab modifies the immunosuppressive tumor milieu through M1-like phenotype macrophage polarization and NK cells degranulation. Depletion experiments revealed a cross-talk between macrophages and NK cells necessary for trastuzumab antitumor effect. In addition, tumor cells treated with DN are more susceptible to trastuzumab-dependent cellular phagocytosis. Finally, MUC4 expression in HER2+ breast cancer is associated with immune desert tumors., Conclusions: These findings provide rationale to pursue sTNFα blockade combined with trastuzumab or trastuzumab drug conjugates for MUC4+ and HER2+ breast cancer patients to overcome trastuzumab resistance., Competing Interests: Competing interests: RS is a consultant for and a research grant from INmune Bio. All other authors declare they have no competing interests., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
13. Halting ErbB-2 isoforms retrograde transport to the nucleus as a new theragnostic approach for triple-negative breast cancer.
- Author
-
Madera S, Izzo F, Chervo MF, Dupont A, Chiauzzi VA, Bruni S, Petrillo E, Merin SS, De Martino M, Montero D, Levit C, Lebersztein G, Anfuso F, Roldán Deamicis A, Mercogliano MF, Proietti CJ, Schillaci R, Elizalde PV, and Cordo Russo RI
- Subjects
- Cell Nucleus metabolism, Humans, Protein Isoforms genetics, Protein Isoforms metabolism, Receptor, ErbB-2 metabolism, Receptors, Progesterone metabolism, Triple Negative Breast Neoplasms pathology
- Abstract
Triple-negative breast cancer (TNBC) is clinically defined by the absence of estrogen and progesterone receptors and the lack of membrane overexpression or gene amplification of receptor tyrosine kinase ErbB-2/HER2. Due to TNBC heterogeneity, clinical biomarkers and targeted therapies for this disease remain elusive. We demonstrated that ErbB-2 is localized in the nucleus (NErbB-2) of TNBC cells and primary tumors, from where it drives growth. We also discovered that TNBC expresses both wild-type ErbB-2 (WTErbB-2) and alternative ErbB-2 isoform c (ErbB-2c). Here, we revealed that the inhibitors of the retrograde transport Retro-2 and its cyclic derivative Retro-2.1 evict both WTErbB-2 and ErbB-2c from the nucleus of BC cells and tumors. Using BC cells from several molecular subtypes, as well as normal breast cells, we demonstrated that Retro-2 specifically blocks proliferation of BC cells expressing NErbB-2. Importantly, Retro-2 eviction of both ErbB-2 isoforms from the nucleus resulted in a striking growth abrogation in multiple TNBC preclinical models, including tumor explants and xenografts. Our mechanistic studies in TNBC cells revealed that Retro-2 induces a differential accumulation of WTErbB-2 at the early endosomes and the plasma membrane, and of ErbB-2c at the Golgi, shedding new light both on Retro-2 action on endogenous protein cargoes undergoing retrograde transport, and on the biology of ErbB-2 splicing variants. In addition, we revealed that the presence of a functional signal peptide and a nuclear export signal (NES), both located at the N-terminus of WTErbB-2, and absent in ErbB-2c, accounts for the differential subcellular distribution of ErbB-2 isoforms upon Retro-2 treatment. Our present discoveries provide evidence for the rational repurposing of Retro-2 as a novel therapeutic agent for TNBC., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
14. Regulation of telomere homeostasis and genomic stability in cancer by N 6 -adenosine methylation (m 6 A).
- Author
-
Lee JH, Hong J, Zhang Z, de la Peña Avalos B, Proietti CJ, Deamicis AR, Guzmán G P, Lam HM, Garcia J, Roudier MP, Sisk AE, De La Rosa R, Vu K, Yang M, Liao Y, Scheirer J, Pechacek D, Yadav P, Rao MK, Zheng S, Johnson-Pais TL, Leach RJ, Elizalde PV, Dray E, and Xu K
- Abstract
The role of RNA methylation on N
6 -adenosine (m6 A) in cancer has been acknowledged, but the underlying mechanisms remain obscure. Here, we identified homeobox containing 1 ( HMBOX1 ) as an authentic target mRNA of m6 A machinery, which is highly methylated in malignant cells compared to the normal counterparts and subject to expedited degradation upon the modification. m6 A-mediated down-regulation of HMBOX1 causes telomere dysfunction and inactivation of p53 signaling, which leads to chromosome abnormalities and aggressive phenotypes. CRISPR-based, m6 A-editing tools further prove that the methyl groups on HMBOX1 per se contribute to the generation of altered cancer genome. In multiple types of human cancers, expression of the RNA methyltransferase METTL3 is negatively correlated with the telomere length but favorably with fractions of altered cancer genome, whereas HMBOX1 mRNA levels show the opposite patterns. Our work suggests that the cancer-driving genomic alterations may potentially be fixed by rectifying particular epitranscriptomic program., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution NonCommercial License 4.0 (CC BY-NC).)- Published
- 2021
- Full Text
- View/download PDF
15. Harnessing Tumor Necrosis Factor Alpha to Achieve Effective Cancer Immunotherapy.
- Author
-
Mercogliano MF, Bruni S, Mauro F, Elizalde PV, and Schillaci R
- Abstract
Tumor necrosis factor alpha (TNFα) is a pleiotropic cytokine known to have contradictory roles in oncoimmunology. Indeed, TNFα has a central role in the onset of the immune response, inducing both activation and the effector function of macrophages, dendritic cells, natural killer (NK) cells, and B and T lymphocytes. Within the tumor microenvironment, however, TNFα is one of the main mediators of cancer-related inflammation. It is involved in the recruitment and differentiation of immune suppressor cells, leading to evasion of tumor immune surveillance. These characteristics turn TNFα into an attractive target to overcome therapy resistance and tackle cancer. This review focuses on the diverse molecular mechanisms that place TNFα as a source of resistance to immunotherapy such as monoclonal antibodies against cancer cells or immune checkpoints and adoptive cell therapy. We also expose the benefits of TNFα blocking strategies in combination with immunotherapy to improve the antitumor effect and prevent or treat adverse immune-related effects.
- Published
- 2021
- Full Text
- View/download PDF
16. Nuclear PDCD4 Expression Defines a Subset of Luminal B-Like Breast Cancers with Good Prognosis.
- Author
-
Madera S, Chervo MF, Chiauzzi VA, Pereyra MG, Venturutti L, Izzo F, Roldán Deamicis A, Guzman P, Dupont A, Roa JC, Cenciarini ME, Barchuk S, Figurelli S, Lopez Della Vecchia D, Levit C, Lebersztein G, Anfuso F, Castiglioni T, Cortese E, Ares S, Deza EG, Gercovich FG, Proietti CJ, Schillaci R, Cordo Russo RI, and Elizalde PV
- Subjects
- Breast Neoplasms pathology, Female, Humans, Prognosis, Apoptosis Regulatory Proteins metabolism, Breast Neoplasms genetics, RNA-Binding Proteins metabolism
- Abstract
The hormone receptor-positive (estrogen and/or progesterone receptor (PR)-positive) and HER2-negative breast cancer (BC) subtype is a biologically heterogeneous entity that includes luminal A-like (LumA-like) and luminal B-like (LumB-like) subtypes. Decreased PR levels is a distinctive biological feature of LumB-like tumors. These tumors also show reduced sensitivity to endocrine therapies and poorer prognosis than LumA-like tumors. Identification of biomarkers to accurately predict disease relapse in these subtypes is crucial in order to select effective therapies. We identified the tumor suppressor PDCD4 (programmed cell death 4), located in the nucleus (NPDCD4), as an independent prognostic factor of good clinical outcome in LumA-like and LumB-like subtypes. NPDCD4-positive LumB-like tumors presented overall and disease-free survival rates comparable to those of NPDCD4-positive LumA-like tumors, indicating that NPDCD4 improves the outcome of LumB-like patients. In contrast, NPDCD4 loss increased the risk of disease recurrence and death in LumB-like compared with LumA-like tumors. This, along with our results showing that LumB-like tumors present lower NPDCD4 positivity than LumA-like tumors, suggests that NPDCD4 loss contributes to endocrine therapy resistance in LumB-like BCs. We also revealed that PR induces PDCD4 transcription in LumB-like BC, providing a mechanistic explanation to the low PDCD4 levels in LumB-like BCs lacking PR. Finally, PDCD4 silencing enhanced BC cell survival in a patient-derived explant model of LumB-like disease. Our discoveries highlight NPDCD4 as a novel biomarker in LumA- and LumB-like subtypes, which could be included in the panel of immunohistochemical markers used in the clinic to accurately predict the prognosis of LumB-like tumors.
- Published
- 2020
- Full Text
- View/download PDF
17. Canonical ErbB-2 isoform and ErbB-2 variant c located in the nucleus drive triple negative breast cancer growth.
- Author
-
Chervo MF, Cordo Russo RI, Petrillo E, Izzo F, De Martino M, Bellora N, Cenciarini ME, Chiauzzi VA, Santa María de la Parra L, Pereyra MG, Güttlein LN, Podhajcer OL, Daniotti JL, Dupont A, Barchuk S, Figurelli S, Lopez Della Vecchia D, Roa JC, Guzmán P, Proietti CJ, Schillaci R, and Elizalde PV
- Subjects
- Cell Line, Tumor, Cell Nucleus enzymology, Cell Nucleus metabolism, Cell Proliferation physiology, Female, Humans, Mitogen-Activated Protein Kinase 7 biosynthesis, Mitogen-Activated Protein Kinase 7 genetics, Paraffin Embedding, Protein Isoforms, Receptor, ErbB-2 antagonists & inhibitors, Receptor, ErbB-2 biosynthesis, Receptor, ErbB-2 genetics, Triple Negative Breast Neoplasms enzymology, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms pathology, Receptor, ErbB-2 metabolism, Triple Negative Breast Neoplasms metabolism
- Abstract
Triple negative breast cancer (TNBC) refers to tumors that do not express clinically significant levels of estrogen and progesterone receptors, and lack membrane overexpression or gene amplification of ErbB-2/HER2, a receptor tyrosine kinase. Transcriptome and proteome heterogeneity of TNBC poses a major challenge to precision medicine. Clinical biomarkers and targeted therapies for this disease remain elusive, so chemotherapy has been the standard of care for early and metastatic TNBC. Our present findings placed ErbB-2 in an unanticipated scenario: the nucleus of TNBC (NErbB-2). Our study on ErbB-2 alternative splicing events, using a PCR-sequencing approach combined with an RNA interference strategy, revealed that TNBC cells express either the canonical (wild-type) ErbB-2, encoded by transcript variant 1, or the non-canonical ErbB-2 isoform c, encoded by alternative variant 3 (RefSeq), or both. These ErbB-2 isoforms function in the nucleus as transcription factors. Evicting both from the nucleus or silencing isoform c only, blocks TN cell and tumor growth. This reveals not only NErbB-2 canonical and alternative isoforms role as targets of therapy in TNBC, but also isoform c dominant oncogenic potential. Furthermore, we validated our findings in the clinic and observed that NErbB-2 correlates with poor prognosis in primary TN tumors, disclosing NErbB-2 as a novel biomarker for TNBC. Our discoveries challenge the present scenario of drug development for personalized BC medicine that focuses on wild-type RefSeq proteins, which conserve the canonical domains and are located in their classical cellular compartments.
- Published
- 2020
- Full Text
- View/download PDF
18. Tumor Necrosis Factor α Blockade: An Opportunity to Tackle Breast Cancer.
- Author
-
Mercogliano MF, Bruni S, Elizalde PV, and Schillaci R
- Abstract
Breast cancer is the most frequently diagnosed cancer and the principal cause of mortality by malignancy in women and represents a main problem for public health worldwide. Tumor necrosis factor α (TNFα) is a pro-inflammatory cytokine whose expression is increased in a variety of cancers. In particular, in breast cancer it correlates with augmented tumor cell proliferation, higher malignancy grade, increased occurrence of metastasis and general poor prognosis for the patient. These characteristics highlight TNFα as an attractive therapeutic target, and consequently, the study of soluble and transmembrane TNFα effects and its receptors in breast cancer is an area of active research. In this review we summarize the recent findings on TNFα participation in luminal, HER2-positive and triple negative breast cancer progression and metastasis. Also, we describe TNFα role in immune response against tumors and in chemotherapy, hormone therapy, HER2-targeted therapy and anti-immune checkpoint therapy resistance in breast cancer. Furthermore, we discuss the use of TNFα blocking strategies as potential therapies and their clinical relevance for breast cancer. These TNFα blocking agents have long been used in the clinical setting to treat inflammatory and autoimmune diseases. TNFα blockade can be achieved by monoclonal antibodies (such as infliximab, adalimumab, etc.), fusion proteins (etanercept) and dominant negative proteins (INB03). Here we address the different effects of each compound and also analyze the use of potential biomarkers in the selection of patients who would benefit from a combination of TNFα blocking agents with HER2-targeted treatments to prevent or overcome therapy resistance in breast cancer., (Copyright © 2020 Mercogliano, Bruni, Elizalde and Schillaci.)
- Published
- 2020
- Full Text
- View/download PDF
19. Steroid hormone receptors: A South American perspective.
- Author
-
Elizalde PV and Proietti CJ
- Subjects
- Humans, Latin America, Receptors, Steroid chemistry, Receptors, Steroid metabolism
- Published
- 2020
- Full Text
- View/download PDF
20. Blockade of Stat3 oncogene addiction induces cellular senescence and reveals a cell-nonautonomous activity suitable for cancer immunotherapy.
- Author
-
De Martino M, Tkach M, Bruni S, Rocha D, Mercogliano MF, Cenciarini ME, Chervo MF, Proietti CJ, Dingli F, Loew D, Fernández EA, Elizalde PV, Piaggio E, and Schillaci R
- Subjects
- Animals, Cellular Senescence, Humans, Immunotherapy, Mice, Oncogene Addiction, STAT3 Transcription Factor genetics, Melanoma, Triple Negative Breast Neoplasms
- Abstract
Stat3 is constitutively activated in several tumor types and plays an essential role in maintaining their malignant phenotype and immunosupression. To take advantage of the promising antitumor activity of Stat3 targeting, it is vital to understand the mechanism by which Stat3 regulates both cell autonomous and non-autonomous processes. Here, we demonstrated that turning off Stat3 constitutive activation in different cancer cell types induces senescence, thus revealing their Stat3 addiction. Taking advantage of the senescence-associated secretory phenotype (SASP) induced by Stat3 silencing (SASP-siStat3), we designed an immunotherapy. The administration of SASP-siStat3 immunotherapy induced a strong inhibition of triple-negative breast cancer and melanoma growth associated with activation of CD4 + T and NK cells. Combining this immunotherapy with anti-PD-1 antibody resulted in survival improvement in mice bearing melanoma. The characterization of the SASP components revealed that type I IFN-related mediators, triggered by the activation of the cyclic GMP-AMP synthase DNA sensing pathway, are important for its immunosurveillance activity. Overall, our findings provided evidence that administration of SASP-siStat3 or low dose of Stat3-blocking agents would benefit patients with Stat3-addicted tumors to unleash an antitumor immune response and to improve the effectiveness of immune checkpoint inhibitors., (© 2020 The Author(s). Published with license by Taylor & Francis Group, LLC.)
- Published
- 2020
- Full Text
- View/download PDF
21. Nuclear ErbB-2: a Novel Therapeutic Target in ErbB-2-Positive Breast Cancer?
- Author
-
Cordo Russo RI, Chervo MF, Madera S, Charreau EH, and Elizalde PV
- Subjects
- Antineoplastic Agents therapeutic use, Apoptosis, Biomarkers, Biomarkers, Tumor metabolism, Breast Neoplasms drug therapy, Cell Nucleus metabolism, Cell Survival, Drug Resistance, Neoplasm, Female, Humans, Neoplasm Metastasis, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction, Treatment Outcome, Breast Neoplasms metabolism, Molecular Targeted Therapy, Receptor, ErbB-2 metabolism
- Abstract
Membrane overexpression of ErbB-2 (MErbB-2), a member of the ErbB family of receptor tyrosine kinases, occurs in 15-20% of breast cancers (BC) and constitutes a therapeutic target in this BC subtype (ErbB-2-positive). Although MErbB-2-targeted therapies have significantly improved patients' clinical outcome, resistance to available drugs is still a major issue in the clinic. Lack of accurate biomarkers for predicting responses to anti-ErbB-2 drugs at the time of diagnosis is also an important unresolved issue. Hence, a better understanding of the ErbB-2 signaling pathway constitutes a critical task in the battle against BC. In its canonical mechanism of action, MErbB-2 activates downstream signaling pathways, which transduce its proliferative effects in BC. The dogma of ErbB-2 mechanism of action has been challenged by the demonstration that MErbB-2 migrates to the nucleus, where it acts as a transcriptional regulator. Accumulating findings demonstrate that nuclear ErbB-2 (NErbB-2) is involved in BC growth and metastasis. Emerging evidence also reveal a role of NErbB-2 in the response to available anti-MErbB-2 agents. Here, we will review NErbB-2 function in BC and will particularly discuss the role of NErbB-2 as a novel target for therapy in ErbB-2-positive BC.
- Published
- 2019
- Full Text
- View/download PDF
22. Revisiting progesterone receptor (PR) actions in breast cancer: Insights into PR repressive functions.
- Author
-
Proietti CJ, Cenciarini ME, and Elizalde PV
- Subjects
- Animals, Breast Neoplasms genetics, Chromatin metabolism, Humans, Transcription, Genetic, Breast Neoplasms metabolism, Receptors, Progesterone metabolism
- Abstract
Progesterone receptor (PR) is a master regulator in female reproductive tissues that controls developmental processes and proliferation and differentiation during the reproductive cycle and pregnancy. PR also plays a role in progression of endocrine-dependent breast cancer. As a member of the nuclear receptor family of ligand-dependent transcription factors, the main action of PR is to regulate networks of target gene expression in response to binding its cognate steroid hormone, progesterone. Liganded-PR transcriptional activation has been thoroughly studied and associated mechanisms have been described while progesterone-mediated repression has remained less explored. The present work summarizes recent advances in the understanding of how PR-mediated repression is accomplished in breast cancer cells and highlights the significance of fully understanding the determinants of context-dependent PR action., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
23. Tamoxifen Resistance in Breast Cancer Is Regulated by the EZH2-ERα-GREB1 Transcriptional Axis.
- Author
-
Wu Y, Zhang Z, Cenciarini ME, Proietti CJ, Amasino M, Hong T, Yang M, Liao Y, Chiang HC, Kaklamani VG, Jeselsohn R, Vadlamudi RK, Huang TH, Li R, De Angelis C, Fu X, Elizalde PV, Schiff R, Brown M, and Xu K
- Subjects
- Animals, Apoptosis, Biomarkers, Tumor, Breast Neoplasms drug therapy, Breast Neoplasms pathology, Carcinogenesis, Cell Proliferation, DNA Methylation, Enhancer of Zeste Homolog 2 Protein genetics, Estrogen Antagonists pharmacology, Estrogen Receptor alpha genetics, Female, Follow-Up Studies, Gene Expression Regulation, Neoplastic, Humans, Mice, Mice, Nude, Neoplasm Proteins genetics, Prognosis, Promoter Regions, Genetic, Transcription, Genetic, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Breast Neoplasms genetics, Drug Resistance, Neoplasm genetics, Enhancer of Zeste Homolog 2 Protein metabolism, Epigenesis, Genetic, Estrogen Receptor alpha metabolism, Neoplasm Proteins metabolism, Tamoxifen pharmacology
- Abstract
Resistance to cancer treatment can be driven by epigenetic reprogramming of specific transcriptomes in favor of the refractory phenotypes. Here we discover that tamoxifen resistance in breast cancer is driven by a regulatory axis consisting of a master transcription factor, its cofactor, and an epigenetic regulator. The oncogenic histone methyltransferase EZH2 conferred tamoxifen resistance by silencing the expression of the estrogen receptor α (ERα) cofactor GREB1. In clinical specimens, induction of DNA methylation of a particular CpG-enriched region at the GREB1 promoter negatively correlated with GREB1 levels and cell sensitivity to endocrine agents. GREB1 also ensured proper cellular reactions to different ligands by recruiting distinct sets of ERα cofactors to cis -regulatory elements, which explains the contradictory biological effects of GREB1 on breast cancer cell growth in response to estrogen or antiestrogen. In refractory cells, EZH2-dependent repression of GREB1 triggered chromatin reallocation of ERα coregulators, converting the antiestrogen into an agonist. In clinical specimens from patients receiving adjuvant tamoxifen treatment, expression levels of EZH2 and GREB1 were correlated negatively, and taken together better predicted patient responses to endocrine therapy. Overall, our work suggests a new strategy to overcome endocrine resistance in metastatic breast cancer by targeting a particular epigenetic program. Significance: This study suggests a new strategy to overcome endocrine resistance in metastatic breast cancer by targeting a particular epigenetic program defined within. Cancer Res; 78(3); 671-84. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
24. Invasive micropapillary carcinoma of the breast overexpresses MUC4 and is associated with poor outcome to adjuvant trastuzumab in HER2-positive breast cancer.
- Author
-
Mercogliano MF, Inurrigarro G, De Martino M, Venturutti L, Rivas MA, Cordo-Russo R, Proietti CJ, Fernández EA, Frahm I, Barchuk S, Allemand DH, Figurelli S, Deza EG, Ares S, Gercovich FG, Cortese E, Amasino M, Guzmán P, Roa JC, Elizalde PV, and Schillaci R
- Subjects
- Adult, Aged, Antineoplastic Agents, Immunological, Biomarkers, Tumor metabolism, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Breast Neoplasms pathology, Carcinoma, Ductal, Breast drug therapy, Carcinoma, Ductal, Breast metabolism, Carcinoma, Ductal, Breast pathology, Carcinoma, Papillary drug therapy, Carcinoma, Papillary metabolism, Carcinoma, Papillary pathology, Case-Control Studies, Chemotherapy, Adjuvant, Female, Follow-Up Studies, Humans, Middle Aged, Neoplasm Invasiveness, Prognosis, Receptor, ErbB-2 antagonists & inhibitors, Receptor, ErbB-2 immunology, Retrospective Studies, Survival Rate, Breast Neoplasms mortality, Carcinoma, Ductal, Breast mortality, Carcinoma, Papillary mortality, Mucin-4 metabolism, Receptor, ErbB-2 metabolism, Trastuzumab pharmacology
- Abstract
Background: Invasive micropapillary carcinoma of the breast (IMPC) is a histological tumor variant that occurs with low frequency characterized by an inside-out formation of tumor clusters with a pseudopapillary arrangement. IMPC is an aggressive tumor with poor clinical outcome. In addition, this histological subtype usually expresses human epidermal growth factor receptor 2 (HER2) which also correlates with a more aggressive tumor. In this work we studied the clinical significance of IMPC in HER2-positive breast cancer patients treated with adjuvant trastuzumab. We also analyzed mucin 4 (MUC4) expression as a novel biomarker to identify IMPC., Methods: We retrospectively studied 86 HER2-positive breast cancer patients treated with trastuzumab and chemotherapy in the adjuvant setting. We explored the association of the IMPC component with clinicopathological parameters at diagnosis and its prognostic value. We compared MUC4 expression in IMPC with respect to other histological breast cancer subtypes by immunohistochemistry., Results: IMPC, either as a pure entity or associated with invasive ductal carcinoma (IDC), was present in 18.6% of HER2-positive cases. It was positively correlated with estrogen receptor expression and tumor size and inversely correlated with patient's age. Disease-free survival was significantly lower in patients with IMPC (hazard ratio = 2.6; 95%, confidence interval 1.1-6.1, P = 0.0340). MUC4, a glycoprotein associated with metastasis, was strongly expressed in all IMPC cases tested. IMPC appeared as the histological breast cancer subtype with the highest MUC4 expression compared to IDC, lobular and mucinous carcinoma., Conclusion: In HER2-positive breast cancer, the presence of IMPC should be carefully examined. As it is often not informed, because it is relatively difficult to identify or altogether overlooked, we propose MUC4 expression as a useful biomarker to highlight IMPC presence. Patients with MUC4-positive tumors with IMPC component should be more frequently monitored and/or receive additional therapies.
- Published
- 2017
- Full Text
- View/download PDF
25. Inhibition of MHC-I by Brucella abortus is an early event during infection and involves EGFR pathway.
- Author
-
Velásquez LN, Milillo MA, Delpino MV, Trotta A, Mercogliano MF, Pozner RG, Schillaci R, Elizalde PV, Giambartolomei GH, and Barrionuevo P
- Subjects
- Animals, Brucella abortus pathogenicity, Brucellosis microbiology, CD8-Positive T-Lymphocytes microbiology, Extracellular Signal-Regulated MAP Kinases metabolism, Female, Histocompatibility Antigens Class I genetics, Humans, Immune Evasion, Mice, Mice, Inbred C57BL, Microbiology, Signal Transduction, THP-1 Cells, Up-Regulation, Brucella abortus immunology, Brucellosis immunology, CD8-Positive T-Lymphocytes immunology, ErbB Receptors metabolism, Histocompatibility Antigens Class I metabolism, Monocytes immunology
- Abstract
Brucella abortus is able to persist inside the host despite the development of potent CD8
+ T-cell responses. We have recently reported the ability of B. abortus to inhibit the interferon-γ-induced major histocompatibility complex (MHC)-I cell surface expression on human monocytes. This phenomenon was due to the B. abortus-mediated retention of MHC-I molecules within the Golgi apparatus and was dependent on bacterial viability. However, the implications of bacterial virulence or replicative capacity and the signaling pathways remained unknown. Here we demonstrated that the B. abortus mutant strains RB51 and virB10- are able to inhibit MHC-I expression in the same manner as wild-type B. abortus, even though they are unable to persist inside human monocytes for a long period of time. Consistent with this, the phenomenon was triggered early in time and could be observed at 8 h postinfection. At 24 and 48 h, it was even stronger. Regarding the signaling pathway, targeting epidermal growth factor (EGF) receptor (EGFR), ErbB2 (HER2) or inhibition of tumor necrosis factor-α-converting enzyme, one of the enzymes which generates soluble EGF-like ligands, resulted in partial recovery of MHC-I surface expression. Moreover, recombinant EGF and transforming growth factor-α as well as the combination of both were also able to reproduce the B. abortus-induced MHC-I downmodulation. Finally, when infection was performed in the presence of an extracellular signal-regulated kinase 1/2 (Erk1/2) inhibitor, MHC-I surface expression was significantly recovered. Overall, these results describe how B. abortus evades CD8+ T-cell responses early during infection and exploits the EGFR-ERK signaling pathway to escape from the immune system and favor chronicity.- Published
- 2017
- Full Text
- View/download PDF
26. TNFα-Induced Mucin 4 Expression Elicits Trastuzumab Resistance in HER2-Positive Breast Cancer.
- Author
-
Mercogliano MF, De Martino M, Venturutti L, Rivas MA, Proietti CJ, Inurrigarro G, Frahm I, Allemand DH, Deza EG, Ares S, Gercovich FG, Guzmán P, Roa JC, Elizalde PV, and Schillaci R
- Subjects
- Ado-Trastuzumab Emtansine, Animals, Antibody-Dependent Cell Cytotoxicity, Antineoplastic Agents, Immunological metabolism, Antineoplastic Agents, Immunological therapeutic use, Breast Neoplasms mortality, Breast Neoplasms pathology, Cell Line, Tumor, Disease-Free Survival, Drug Resistance, Neoplasm drug effects, Drug Synergism, Female, Humans, Immunoconjugates pharmacology, Maytansine analogs & derivatives, Maytansine pharmacology, Mice, Mice, Nude, Mucin-4 biosynthesis, Mucin-4 genetics, Neoplasm Proteins antagonists & inhibitors, RNA Interference, Receptor, ErbB-2 antagonists & inhibitors, Recombinant Fusion Proteins metabolism, Signal Transduction drug effects, Stomach Neoplasms pathology, Trastuzumab metabolism, Trastuzumab therapeutic use, Tumor Necrosis Factor-alpha antagonists & inhibitors, Tumor Necrosis Factor-alpha genetics, Tumor Necrosis Factor-alpha immunology, Up-Regulation drug effects, Xenograft Model Antitumor Assays, Antineoplastic Agents, Immunological pharmacology, Breast Neoplasms drug therapy, Drug Resistance, Neoplasm physiology, Gene Expression Regulation, Neoplastic drug effects, Mucin-4 physiology, Neoplasm Proteins analysis, Receptor, ErbB-2 analysis, Trastuzumab pharmacology, Tumor Necrosis Factor-alpha physiology
- Abstract
Purpose: Although trastuzumab administration improved the outcome of HER2-positive breast cancer patients, resistance events hamper its clinical benefits. We demonstrated that TNFα stimulation in vitro induces trastuzumab resistance in HER2-positive breast cancer cell lines. Here, we explored the mechanism of TNFα-induced trastuzumab resistance and the therapeutic strategies to overcome it., Experimental Design: Trastuzumab-sensitive breast cancer cells, genetically engineered to stably overexpress TNFα, and de novo trastuzumab-resistant tumors, were used to evaluate trastuzumab response and TNFα-blocking antibodies effectiveness respectively. Immunohistochemistry and antibody-dependent cell cytotoxicity (ADCC), together with siRNA strategy, were used to explore TNFα influence on the expression and function of its downstream target, mucin 4 (MUC4). The clinical relevance of MUC4 expression was studied in a cohort of 78 HER2-positive breast cancer patients treated with adjuvant trastuzumab., Results: TNFα overexpression turned trastuzumab-sensitive cells and tumors into resistant ones. Histopathologic findings revealed mucin foci in TNFα-producing tumors. TNFα induced upregulation of MUC4 that reduced trastuzumab binding to its epitope and impaired ADCC. Silencing MUC4 enhanced trastuzumab binding, increased ADCC, and overcame trastuzumab and trastuzumab-emtansine antiproliferative effects in TNFα-overexpressing cells. Accordingly, administration of TNFα-blocking antibodies downregulated MUC4 and sensitized de novo trastuzumab-resistant breast cancer cells and tumors to trastuzumab. In HER2-positive breast cancer samples, MUC4 expression was found to be an independent predictor of poor disease-free survival (P = 0.008)., Conclusions: We identified TNFα-induced MUC4 expression as a novel trastuzumab resistance mechanism. We propose MUC4 expression as a predictive biomarker of trastuzumab efficacy and a guide to combination therapy of TNFα-blocking antibodies with trastuzumab. Clin Cancer Res; 23(3); 636-48. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
27. MiR-16 mediates trastuzumab and lapatinib response in ErbB-2-positive breast and gastric cancer via its novel targets CCNJ and FUBP1.
- Author
-
Venturutti L, Cordo Russo RI, Rivas MA, Mercogliano MF, Izzo F, Oakley RH, Pereyra MG, De Martino M, Proietti CJ, Yankilevich P, Roa JC, Guzmán P, Cortese E, Allemand DH, Huang TH, Charreau EH, Cidlowski JA, Schillaci R, and Elizalde PV
- Subjects
- 3' Untranslated Regions, Animals, Antineoplastic Agents pharmacology, Binding Sites, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Cell Line, Tumor, Cell Proliferation, Disease Models, Animal, Drug Resistance, Neoplasm genetics, Female, Genes, Tumor Suppressor, Humans, Lapatinib, Male, Mice, Models, Biological, Promoter Regions, Genetic, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-myc metabolism, RNA Interference, RNA-Binding Proteins, Receptor, ErbB-2 antagonists & inhibitors, Receptor, ErbB-2 metabolism, Stomach Neoplasms drug therapy, Stomach Neoplasms metabolism, Breast Neoplasms genetics, Cyclins genetics, DNA Helicases genetics, DNA-Binding Proteins genetics, Gene Expression Regulation, Neoplastic drug effects, MicroRNAs genetics, Quinazolines pharmacology, Stomach Neoplasms genetics, Trastuzumab pharmacology
- Abstract
ErbB-2 amplification/overexpression accounts for an aggressive breast cancer (BC) subtype (ErbB-2-positive). Enhanced ErbB-2 expression was also found in gastric cancer (GC) and has been correlated with poor clinical outcome. The ErbB-2-targeted therapies trastuzumab (TZ), a monoclonal antibody, and lapatinib, a tyrosine kinase inhibitor, have proved highly beneficial. However, resistance to such therapies remains a major clinical challenge. We here revealed a novel mechanism underlying the antiproliferative effects of both agents in ErbB-2-positive BC and GC. TZ and lapatinib ability to block extracellular signal-regulated kinases 1/2 and phosphatidylinositol-3 kinase (PI3K)/AKT in sensitive cells inhibits c-Myc activation, which results in upregulation of miR-16. Forced expression of miR-16 inhibited in vitro proliferation in BC and GC cells, both sensitive and resistant to TZ and lapatinib, as well as in a preclinical BC model resistant to these agents. This reveals miR-16 role as tumor suppressor in ErbB-2-positive BC and GC. Using genome-wide expression studies and miRNA target prediction algorithms, we identified cyclin J and far upstream element-binding protein 1 (FUBP1) as novel miR-16 targets, which mediate miR-16 antiproliferative effects. Supporting the clinical relevance of our results, we found that high levels of miR-16 and low or null FUBP1 expression correlate with TZ response in ErbB-2-positive primary BCs. These findings highlight a potential role of miR-16 and FUBP1 as biomarkers of sensitivity to TZ therapy. Furthermore, we revealed miR-16 as an innovative therapeutic agent for TZ- and lapatinib-resistant ErbB-2-positive BC and GC.
- Published
- 2016
- Full Text
- View/download PDF
28. ErbB-2 nuclear function in breast cancer growth, metastasis and resistance to therapy.
- Author
-
Elizalde PV, Cordo Russo RI, Chervo MF, and Schillaci R
- Subjects
- Animals, Antineoplastic Agents therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Female, Gene Expression Regulation, Neoplastic, Humans, Neoplasm Metastasis, Receptor, ErbB-2 genetics, Signal Transduction genetics, Breast Neoplasms pathology, Drug Resistance, Neoplasm genetics, Receptor, ErbB-2 physiology
- Abstract
Approximately 15-20% of breast cancers (BC) show either membrane overexpression of ErbB-2 (MErbB-2), a member of the ErbBs family of receptor tyrosine kinases, or ERBB2 gene amplification. Until the development of MErbB-2-targeted therapies, this BC subtype, called ErbB-2-positive, was associated with increased metastatic potential and poor prognosis. Although these therapies have significantly improved overall survival and cure rates, resistance to available drugs is still a major clinical issue. In its classical mechanism, MErbB-2 activates downstream signaling cascades, which transduce its effects in BC. The fact that ErbB-2 is also present in the nucleus of BC cells was discovered over twenty years ago. Also, compelling evidence revealed a non-canonical function of nuclear ErbB-2 as a transcriptional regulator. As a deeper understanding of nuclear ErbB-2 actions would be crucial to the disclosure of its role as a biomarker and a target of therapy in BC, we will here review its function in BC, in particular, its role in growth, metastatic spreading and response to currently available MErbB-2-positive BC therapies., (© 2016 Society for Endocrinology.)
- Published
- 2016
- Full Text
- View/download PDF
29. Stat3 regulates ErbB-2 expression and co-opts ErbB-2 nuclear function to induce miR-21 expression, PDCD4 downregulation and breast cancer metastasis.
- Author
-
Venturutti L, Romero LV, Urtreger AJ, Chervo MF, Cordo Russo RI, Mercogliano MF, Inurrigarro G, Pereyra MG, Proietti CJ, Izzo F, Díaz Flaqué MC, Sundblad V, Roa JC, Guzmán P, Bal de Kier Joffé ED, Charreau EH, Schillaci R, and Elizalde PV
- Subjects
- Adolescent, Adult, Aged, Apoptosis Regulatory Proteins genetics, Breast Neoplasms pathology, Cell Line, Tumor, Female, Gene Expression Regulation, Neoplastic, Humans, MicroRNAs genetics, Middle Aged, Neoplasm Metastasis, RNA-Binding Proteins genetics, Receptor, ErbB-2 genetics, Signal Transduction, Transcriptional Activation genetics, Transfection, Apoptosis Regulatory Proteins biosynthesis, Breast Neoplasms genetics, MicroRNAs biosynthesis, RNA-Binding Proteins biosynthesis, Receptor, ErbB-2 biosynthesis, STAT3 Transcription Factor genetics
- Abstract
Membrane overexpression of the receptor tyrosine kinase ErbB-2 (MErbB-2) accounts for a clinically aggressive breast cancer (BC) subtype (ErbB-2-positive) with increased incidence of metastases. We and others demonstrated that nuclear ErbB-2 (NErbB-2) also plays a key role in BC and is a poor prognostic factor in ErbB-2-positive tumors. The signal transducer and activator of transcription 3 (Stat3), another player in BC, has been recognized as a downstream mediator of MErbB-2 action in BC metastasis. Here, we revealed an unanticipated novel direction of the ErbB-2 and Stat3 interaction underlying BC metastasis. We found that Stat3 binds to its response elements (GAS) at the ErbB-2 promoter to upregulate ErbB-2 transcription in metastatic, ErbB-2-positive BC. We validated these results in several BC subtypes displaying metastatic and non-metastatic ability, highlighting Stat3 general role as upstream regulator of ErbB-2 expression in BC. Moreover, we showed that Stat3 co-opts NErbB-2 function by recruiting ErbB-2 as its coactivator at the GAS sites in the promoter of microRNA-21 (miR-21), a metastasis-promoting microRNA (miRNA). Using an ErbB-2 nuclear localization domain mutant and a constitutively activated ErbB-2 variant, we found that NErbB-2 role as a Stat3 coactivator and also its direct role as transcription factor upregulate miR-21 in BC. This reveals a novel function of NErbB-2 as a regulator of miRNAs expression. Increased levels of miR-21, in turn, downregulate the expression of the metastasis-suppressor protein programmed cell death 4 (PDCD4), a validated miR-21 target. Using an in vivo model of metastatic ErbB-2-postive BC, in which we silenced Stat3 and reconstituted ErbB-2 or miR-21 expression, we showed that both are downstream mediators of Stat3-driven metastasis. Supporting the clinical relevance of our results, we found an inverse correlation between ErbB-2/Stat3 nuclear co-expression and PDCD4 expression in ErbB-2-positive primary invasive BCs. Our findings identify Stat3 and NErbB-2 as novel therapeutic targets to inhibit ErbB-2-positive BC metastasis.
- Published
- 2016
- Full Text
- View/download PDF
30. Heregulin Co-opts PR Transcriptional Action Via Stat3 Role As a Coregulator to Drive Cancer Growth.
- Author
-
Proietti CJ, Izzo F, Díaz Flaqué MC, Cordo Russo R, Venturutti L, Mercogliano MF, De Martino M, Pineda V, Muñoz S, Guzmán P, Roa JC, Schillaci R, and Elizalde PV
- Subjects
- Animals, Base Sequence, Binding Sites, Cell Nucleus drug effects, Cell Nucleus metabolism, Cell Proliferation drug effects, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Female, Gene Expression Regulation, Neoplastic drug effects, Mammary Tumor Virus, Mouse genetics, Mice, Inbred BALB C, Molecular Sequence Data, Promoter Regions, Genetic, Protein Binding drug effects, Receptor, ErbB-2 metabolism, Signal Transduction drug effects, Signal Transduction genetics, Transcriptional Activation drug effects, bcl-X Protein genetics, bcl-X Protein metabolism, Breast Neoplasms genetics, Breast Neoplasms pathology, Neuregulin-1 pharmacology, Receptors, Progesterone genetics, STAT3 Transcription Factor metabolism, Transcriptional Activation genetics
- Abstract
Accumulated findings have demonstrated the presence of bidirectional interactions between progesterone receptor (PR) and the ErbB family of receptor tyrosine kinases signaling pathways in breast cancer. We previously revealed signal transducer and activator of transcription 3 (Stat3) as a nodal convergence point between said signaling pathways proving that Stat3 is activated by one of the ErbBs' ligands, heregulin (HRG)β1 via ErbB2 and through the co-option of PR as a signaling molecule. Here, we found that HRGβ1 induced Stat3 recruitment to the promoters of the progestin-regulated cell cycle modulators Bcl-XL and p21(CIP1) and also stimulated Stat3 binding to the mouse mammary tumor virus promoter, which carries consensus progesterone response elements. Interestingly, HRGβ1-activated Stat3 displayed differential functions on PR activity depending on the promoter bound. Indeed, Stat3 was required for PR binding in bcl-X, p21(CIP1), and c-myc promoters while exerting a PR coactivator function on the mouse mammary tumor virus promoter. Stat3 also proved to be necessary for HRGβ1-induced in vivo tumor growth. Our results endow Stat3 a novel function as a coregulator of HRGβ1-activated PR to promote breast cancer growth. These findings underscore the importance of understanding the complex interactions between PR and other regulatory factors, such as Stat3, that contribute to determine the context-dependent transcriptional actions of PR.
- Published
- 2015
- Full Text
- View/download PDF
31. Targeting ErbB-2 nuclear localization and function inhibits breast cancer growth and overcomes trastuzumab resistance.
- Author
-
Cordo Russo RI, Béguelin W, Díaz Flaqué MC, Proietti CJ, Venturutti L, Galigniana N, Tkach M, Guzmán P, Roa JC, O'Brien NA, Charreau EH, Schillaci R, and Elizalde PV
- Subjects
- Active Transport, Cell Nucleus drug effects, Animals, Cell Nucleus drug effects, Cell Nucleus metabolism, Cell Proliferation genetics, Drug Resistance, Neoplasm genetics, Drug Synergism, Female, Genes, Dominant physiology, Humans, Mice, Inbred BALB C, Mice, Nude, Mutant Proteins therapeutic use, Protein Isoforms pharmacology, Protein Isoforms therapeutic use, Protein Transport drug effects, Receptor, ErbB-2 genetics, Receptor, ErbB-2 metabolism, Receptor, ErbB-2 physiology, Trastuzumab, Tumor Cells, Cultured, Antibodies, Monoclonal, Humanized therapeutic use, Breast Neoplasms drug therapy, Breast Neoplasms pathology, Cell Proliferation drug effects, Drug Resistance, Neoplasm drug effects, Molecular Targeted Therapy methods, Mutant Proteins pharmacology, Receptor, ErbB-2 antagonists & inhibitors
- Abstract
Membrane overexpression of ErbB-2/HER2 receptor tyrosine kinase (membrane ErbB-2 (MErbB-2)) has a critical role in breast cancer (BC). We and others have also shown the role of nuclear ErbB-2 (NErbB-2) in BC, whose presence we identified as a poor prognostic factor in MErbB-2-positive tumors. Current anti-ErbB-2 therapies, as with the antibody trastuzumab (Ttzm), target only MErbB-2. Here, we found that blockade of NErbB-2 action abrogates growth of BC cells, sensitive and resistant to Ttzm, in a scenario in which ErbB-2, ErbB-3 and Akt are phosphorylated, and ErbB-2/ErbB-3 dimers are formed. Also, inhibition of NErbB-2 presence suppresses growth of a preclinical BC model resistant to Ttzm. We showed that at the cyclin D1 promoter, ErbB-2 assembles a transcriptional complex with Stat3 (signal transducer and activator of transcription 3) and ErbB-3, another member of the ErbB family, which reveals the first nuclear function of ErbB-2/ErbB-3 dimer. We identified NErbB-2 as the major proliferation driver in Ttzm-resistant BC, and demonstrated that Ttzm inability to disrupt the Stat3/ErbB-2/ErbB-3 complex underlies its failure to inhibit growth. Furthermore, our results in the clinic revealed that nuclear interaction between ErbB-2 and Stat3 correlates with poor overall survival in primary breast tumors. Our findings challenge the paradigm of anti-ErbB-2 drug design and highlight NErbB-2 as a novel target to overcome Ttzm resistance.
- Published
- 2015
- Full Text
- View/download PDF
32. Progesterone-induced stimulation of mammary tumorigenesis is due to the progesterone metabolite, 5α-dihydroprogesterone (5αP) and can be suppressed by the 5α-reductase inhibitor, finasteride.
- Author
-
Wiebe JP, Rivas MA, Mercogliano MF, Elizalde PV, and Schillaci R
- Subjects
- Animals, Breast metabolism, Breast pathology, Breast Neoplasms metabolism, Breast Neoplasms pathology, Carcinogenesis metabolism, Carcinogenesis pathology, Cell Line, Tumor, Cell Proliferation drug effects, Cholestenone 5 alpha-Reductase analysis, Female, Humans, Mice, Mice, Inbred BALB C, Progesterone metabolism, 5-alpha Reductase Inhibitors therapeutic use, 5-alpha-Dihydroprogesterone metabolism, Breast drug effects, Breast Neoplasms prevention & control, Carcinogenesis drug effects, Cholestenone 5 alpha-Reductase metabolism, Finasteride therapeutic use
- Abstract
Progesterone has long been linked to breast cancer but its actual role as a cancer promoter has remained in dispute. Previous in vitro studies have shown that progesterone is converted to 5α-dihydroprogesterone (5αP) in breast tissue and human breast cell lines by the action of 5α-reductase, and that 5αP acts as a cancer-promoter hormone. Also studies with human breast cell lines in which the conversion of progesterone to 5αP is blocked by a 5α-reductase inhibitor, have shown that the in vitro stimulation in cell proliferation with progesterone treatments are not due to progesterone itself but to the metabolite 5αP. No similar in vivo study has been previously reported. The objective of the current studies was to determine in an in vivo mouse model if the presumptive progesterone-induced mammary tumorigenesis is due to the progesterone metabolite, 5αP. BALB/c mice were challenged with C4HD murine mammary cells, which have been shown to form tumors when treated with progesterone or the progestin, medroxyprogesterone acetate. Cells and mice were treated with various doses and combinations of progesterone, 5αP and/or the 5α-reductase inhibitor, finasteride, and the effects on cell proliferation and induction and growth of tumors were monitored. Hormone levels in serum and tumors were measured by specific RIA and ELISA tests. Proliferation of C4HD cells and induction and growth of tumors was stimulated by treatment with either progesterone or 5αP. The progesterone-induced stimulation was blocked by finasteride and reinstated by concomitant treatment with 5αP. The 5αP-induced tumors expressed high levels of ER, PR and ErbB-2. Hormone measurements showed significantly higher levels of 5αP in serum from mice with tumors than from mice without tumors, regardless of treatments, and 5αP levels were significantly higher (about 4-fold) in tumors than in respective sera, while progesterone levels did not differ between the compartments. The results indicate that the stimulation of C4HD tumor growth in BALB/c mice treated with progesterone is due to the progesterone metabolite 5αP formed at elevated levels in mammary cells as a result of the 5α-reductase action on progesterone. The results provide the first in vivo demonstration that stimulation of breast cell tumorigenesis and tumor growth accompanying progesterone treatment is due to the progesterone metabolite 5αP, and that breast tumorigenesis can be blocked with the 5α-reductase inhibitor, finasteride., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
33. Progesterone receptor activation downregulates GATA3 by transcriptional repression and increased protein turnover promoting breast tumor growth.
- Author
-
Izzo F, Mercogliano F, Venturutti L, Tkach M, Inurrigarro G, Schillaci R, Cerchietti L, Elizalde PV, and Proietti CJ
- Subjects
- Animals, Breast Neoplasms metabolism, Cell Line, Tumor, Cyclin A2 metabolism, Down-Regulation, Female, GATA3 Transcription Factor metabolism, Humans, Mammary Neoplasms, Animal metabolism, Mice, Phosphorylation, Receptors, Estrogen, Breast Neoplasms genetics, Cyclin A2 genetics, GATA3 Transcription Factor genetics, Gene Expression Regulation, Neoplastic, Mammary Neoplasms, Animal genetics, Progestins metabolism, Receptors, Progesterone metabolism
- Abstract
Introduction: The transcription factor GATA3 is involved in mammary gland development and is crucial for the maintenance of the differentiated status of luminal epithelial cells. The role of GATA3 in breast cancer as a tumor suppressor has been established, although insights into the mechanism of GATA3 expression loss are still required., Methods: Chromatin immunoprecipitation assays were conducted to study progestin modulation of recruitment of transcription factors to GATA3 promoter. We performed western blot and reverse RT-qPCR experiments to explore progestin regulation of GATA3 protein and mRNA expression respectively. Confocal microscopy and in vitro phosphorylation studies were conducted to examine progestin capacity to induce GATA3 serine phosphorylation in its 308 residue. GATA3 participation in progestin-induced breast cancer growth was addressed in in vitro proliferation and in vivo tumor growth experiments., Results: In this study, we demonstrate that progestin-activated progesterone receptor (PR) reduces GATA3 expression through regulation at the transcriptional and post-translational levels in breast cancer cells. In the former mechanism, the histone methyltransferase enhancer of zeste homolog 2 is co-recruited with activated PR to a putative progesterone response element in the GATA3 proximal promoter, increasing H3K27me3 levels and inducing chromatin compaction, resulting in decreased GATA3 mRNA levels. This transcriptional regulation is coupled with increased GATA3 protein turnover through progestin-induced GATA3 phosphorylation at serine 308 followed by 26S proteasome-mediated degradation. Both molecular mechanisms converge to accomplish decreased GATA3 expression levels in breast cancer cells upon PR activation. In addition, we demonstrated that decreased GATA3 levels are required for progestin-induced upregulation of cyclin A2, which mediates the G1 to S phase transition of the cell cycle and was reported to be associated with poor prognosis in breast cancer. Finally, we showed that downregulation of GATA3 is required for progestin stimulation of both in vitro cell proliferation and in vivo tumor growth., Conclusions: In the present study, we reveal that progestin-induced PR activation leads to loss of GATA3 expression in breast cancer cells through transcriptional and post-translational regulation. Importantly, we demonstrate that GATA3 downregulation is required for progestin-induced upregulation of cyclin A2 and for progestin-induced in vitro and in vivo breast cancer cell growth.
- Published
- 2014
- Full Text
- View/download PDF
34. Progesterone receptor assembly of a transcriptional complex along with activator protein 1, signal transducer and activator of transcription 3 and ErbB-2 governs breast cancer growth and predicts response to endocrine therapy.
- Author
-
Díaz Flaqué MC, Galigniana NM, Béguelin W, Vicario R, Proietti CJ, Russo R, Rivas MA, Tkach M, Guzmán P, Roa JC, Maronna E, Pineda V, Muñoz S, Mercogliano M, Charreau EH, Yankilevich P, Schillaci R, and Elizalde PV
- Subjects
- Animals, Breast Neoplasms mortality, Breast Neoplasms pathology, Cell Nucleus drug effects, Cyclin D1 genetics, Cyclin D1 metabolism, Female, Follow-Up Studies, Humans, Medroxyprogesterone Acetate pharmacology, Mice, Inbred BALB C, Phosphorylation drug effects, Promoter Regions, Genetic, Receptor, ErbB-2 genetics, Retrospective Studies, Selective Estrogen Receptor Modulators therapeutic use, Tamoxifen therapeutic use, Treatment Outcome, Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Receptor, ErbB-2 metabolism, Receptors, Progesterone metabolism, STAT3 Transcription Factor metabolism, Transcription Factor AP-1 metabolism
- Abstract
Introduction: The role of the progesterone receptor (PR) in breast cancer remains a major clinical challenge. Although PR induces mammary tumor growth, its presence in breast tumors is a marker of good prognosis. We investigated coordinated PR rapid and nonclassical transcriptional effects governing breast cancer growth and endocrine therapy resistance., Methods: We used breast cancer cell lines expressing wild-type and mutant PRs, cells sensitive and resistant to endocrine therapy, a variety of molecular and cellular biology approaches, in vitro proliferation studies and preclinical models to explore PR regulation of cyclin D1 expression, tumor growth, and response to endocrine therapy. We investigated the clinical significance of activator protein 1 (AP-1) and PR interaction in a cohort of 99 PR-positive breast tumors by an immunofluorescence protocol we developed. The prognostic value of AP-1/PR nuclear colocalization in overall survival (OS) was evaluated using Kaplan-Meier method, and Cox model was used to explore said colocalization as an independent prognostic factor for OS., Results: We demonstrated that at the cyclin D1 promoter and through coordinated rapid and transcriptional effects, progestin induces the assembly of a transcriptional complex among AP-1, Stat3, PR, and ErbB-2 which functions as an enhanceosome to drive breast cancer growth. Our studies in a cohort of human breast tumors identified PR and AP-1 nuclear interaction as a marker of good prognosis and better OS in patients treated with tamoxifen (Tam), an anti-estrogen receptor therapy. Rationale for this finding was provided by our demonstration that Tam inhibits rapid and genomic PR effects, rendering breast cancer cells sensitive to its antiproliferative effects., Conclusions: We here provided novel insight into the paradox of PR action as well as new tools to identify the subgroup of ER+/PR + patients unlikely to respond to ER-targeted therapies.
- Published
- 2013
- Full Text
- View/download PDF
35. Progestin drives breast cancer growth by inducing p21(CIP1) expression through the assembly of a transcriptional complex among Stat3, progesterone receptor and ErbB-2.
- Author
-
Diaz Flaqué MC, Vicario R, Proietti CJ, Izzo F, Schillaci R, and Elizalde PV
- Subjects
- Animals, Breast Neoplasms genetics, Cyclin-Dependent Kinase Inhibitor p21 biosynthesis, Female, Mice, Mice, Inbred BALB C, Transcription, Genetic genetics, Tumor Cells, Cultured, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cyclin-Dependent Kinase Inhibitor p21 genetics, Progestins pharmacology, Receptor, ErbB-2 metabolism, Receptors, Progesterone metabolism, STAT3 Transcription Factor metabolism, Transcription, Genetic drug effects
- Abstract
Cell cycle regulator p21(CIP1) has controversial biological effects in breast cancer since in spite of its role as cell cycle inhibitor and promoter of cellular senescence, it also induces cell proliferation and chemoteraphy resistance. We here explored the molecular mechanisms involved in progestin regulation of p21(CIP1) expression. We also investigated the biological effects of p21(CIP1) in breast cancer cells. We found that the synthetic progestin medroxyprogesterone acetate (MPA) upregulates p21(CIP1) protein expression via c-Src, signal transducer and activator of transcription 3 (Stat3) and ErbB-2 phosphorylation. Notably, we also found that ErbB-2 nuclear function plays a key role in MPA-induction of p21(CIP1) expression. Interestingly, we determined that progestin drives p21(CIP1) transcriptional activation via a novel nonclassical transcriptional mechanism in which progesterone receptor is recruited along with Stat3 and ErbB-2 to a Stat3 binding site at p21(CIP1) promoter. Our findings revealed that ErbB-2 functions as a coactivator of Stat3 in progestin induction of p21(CIP1) transcriptional activation. Furthermore, we demonstrated that blockage of p21(CIP1) expression strongly inhibited in vitro and in vivo progestin-induced breast cancer cell proliferation. These results further support the hypothesis that according to cell context and type of stimulus, p21(CIP1) is capable of inducing cell cycle progression. Moreover, we provided evidence that Stat3 and nuclear ErbB-2 are key players in progestin-induced p21(CIP1) regulation., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
36. 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
37. 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
38. Downregulation of the tumor-suppressor miR-16 via progestin-mediated oncogenic signaling contributes to breast cancer development.
- Author
-
Rivas MA, Venturutti L, Huang YW, Schillaci R, Huang TH, and Elizalde PV
- Subjects
- 3' Untranslated Regions genetics, Animals, Cell Line, Tumor, Cell Proliferation, Cyclin E genetics, Cyclin E metabolism, Down-Regulation, Female, Gene Expression Regulation, Neoplastic, Genes, Tumor Suppressor, Genome-Wide Association Study, Humans, Mice, Mice, Inbred BALB C, Oncogene Proteins genetics, Oncogene Proteins metabolism, Proteins metabolism, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins c-myc metabolism, RNA Interference, RNA, Small Interfering, Receptors, Progesterone genetics, Receptors, Progesterone metabolism, STAT3 Transcription Factor genetics, STAT3 Transcription Factor metabolism, Signal Transduction, Up-Regulation, Histidine-Rich Glycoprotein, Breast Neoplasms genetics, Breast Neoplasms metabolism, MicroRNAs metabolism, Progestins metabolism
- Abstract
Introduction: Experimental and clinical evidence points to a critical role of progesterone and the nuclear progesterone receptor (PR) in controlling mammary gland tumorigenesis. However, the molecular mechanisms of progesterone action in breast cancer still remain elusive. On the other hand, micro RNAs (miRNAs) are short ribonucleic acids which have also been found to play a pivotal role in cancer pathogenesis. The role of miRNA in progestin-induced breast cancer is poorly explored. In this study we explored progestin modulation of miRNA expression in mammary tumorigenesis., Methods: We performed a genome-wide study to explore progestin-mediated regulation of miRNA expression in breast cancer. miR-16 expression was studied by RT-qPCR in cancer cell lines with silenced PR, signal transducer and activator of transcription 3 (Stat3) or c-Myc, treated or not with progestins. Breast cancer cells were transfected with the precursor of miR-16 and proliferation assays, Western blots or in vivo experiments were performed. Target genes of miR-16 were searched through a bioinformatical approach, and the study was focused on cyclin E. Reporter gene assays were performed to confirm that cyclin E 3'UTR is a direct target of miR-16., Results: We found that nine miRNAs were upregulated and seven were downregulated by progestin in mammary tumor cells. miR-16, whose function as a tumor suppressor in leukemia has already been shown, was identified as one of the downregulated miRNAs in murine and human breast cancer cells. Progestin induced a decrease in miR-16 levels via the classical PR and through a hierarchical interplay between Stat3 and the oncogenic transcription factor c-Myc. A search for miR-16 targets showed that the CCNE1 gene, encoding the cell cycle regulator cyclin E, contains conserved putative miR-16 target sites in its mRNA 3' UTR region. We found that, similar to the molecular mechanism underlying progestin-modulated miR-16 expression, Stat3 and c-Myc participated in the induction of cyclin E expression by progestin. Moreover, overexpression of miR-16 abrogated the ability of progestin to induce cyclin E upregulation, revealing that cyclin E is a novel target of miR-16 in breast cancer. Overexpression of miR-16 also inhibited progestin-induced breast tumor growth in vitro and in vivo, demonstrating for the first time, a role for miR-16 as a tumor suppressor in mammary tumorigenesis. We also found that the ErbB ligand heregulin (HRG) downregulated the expression of miR-16, which then participates in the proliferative activity of HRG in breast tumor cells., Conclusions: In this study, we reveal the first progestin-regulated miRNA expression profile and identify a novel role for miR-16 as a tumor suppressor in progestin- and growth factor-induced growth in breast cancer.
- Published
- 2012
- Full Text
- View/download PDF
39. Influence of conformationally restricted pyrimidines on the activity of 10-23 DNAzymes.
- Author
-
Robaldo L, Izzo F, Dellafiore M, Proietti C, Elizalde PV, Montserrat JM, and Iribarren AM
- Subjects
- Antineoplastic Agents chemistry, Biocatalysis, Cell Proliferation drug effects, DNA, Catalytic metabolism, DNA, Single-Stranded metabolism, Dose-Response Relationship, Drug, Drug Screening Assays, Antitumor, Enzyme Activation drug effects, Humans, Molecular Conformation, Pyrimidines chemistry, STAT3 Transcription Factor antagonists & inhibitors, STAT3 Transcription Factor genetics, STAT3 Transcription Factor metabolism, Structure-Activity Relationship, Tumor Cells, Cultured, Antineoplastic Agents pharmacology, DNA, Catalytic antagonists & inhibitors, DNA, Single-Stranded antagonists & inhibitors, Pyrimidines pharmacology
- Abstract
The catalytic core of a 10-23 DNAzyme was modified introducing conformationally restricted nucleosides such as (2'R)-, (2'S)-2'-deoxy-2'-C-methyluridine, (2'R)-, (2'S)-2'-deoxy-2'-C-methylcytidine, 2,2'-anhydrouridine and LNA-C, in one, two or three positions. Catalytic activities under pseudo first order conditions were compared at different Mg(2+) concentrations using a short RNA substrate. At low Mg(2+) concentrations, triple modified DNAzymes with similar kinetic performance to that displayed by the non-modified control were identified. In the search for a partial explanation of the obtained results, in silico studies were carried out in order to explore the conformational behavior of 2'-deoxy-2'-C-methylpyrimidines in the context of a loop structure, suggesting that at least partial flexibility is needed for the maintenance of activity. Finally, the modified 2'-C-methyl DNAzyme activity was tested assessing the inhibition of Stat3 expression and the decrease in cell proliferation using the human breast cancer cell line T47D., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
40. The molecular basis of progesterone receptor action in breast carcinogenesis.
- Author
-
Elizalde PV and Proietti CJ
- Abstract
Abstract Progesterone plays an essential role in the regulation of cell proliferation and differentiation in the mammary gland. In addition, experimental and clinical evidence points to a critical role of progesterone and the nuclear progesterone receptor (PR) in controlling mammary gland tumorigenesis. However, the molecular mechanisms of progesterone action in breast cancer still remain elusive. In its classical mechanism of action, PR acts as a ligand-induced transcription factor (TF) interacting directly with specific progesterone response elements (PREs) in the promoter of target genes. In addition to its transcriptional effects, PR activates signal transduction pathways through a rapid or non-genomic mechanism. Interestingly, progestin induces the expression of key genes involved in breast cancer growth, which lack PREs in their promoters, via a non-classical PR transcriptional mechanism through PR tethering to other TFs. Recent findings on steroid hormone receptor modulation of target genes raise the most exciting possibility that progestin may also induce long-range transcriptional control of gene expression via PR binding to cis-regulatory elements (PREs or half PREs) located far upstream or downstream from the trascriptional start site. This review will focus on the involvement and interplay of the different PR actions in breast cancer.
- Published
- 2012
- Full Text
- View/download PDF
41. Clinical relevance of ErbB-2/HER2 nuclear expression in breast cancer.
- Author
-
Schillaci R, Guzmán P, Cayrol F, Beguelin W, Díaz Flaqué MC, Proietti CJ, Pineda V, Palazzi J, Frahm I, Charreau EH, Maronna E, Roa JC, and Elizalde PV
- Subjects
- Adult, Aged, Biomarkers, Tumor analysis, Breast Neoplasms chemistry, Carcinoma chemistry, Chile, Cohort Studies, Female, Humans, Immunohistochemistry, Kaplan-Meier Estimate, Membrane Proteins analysis, Membrane Proteins metabolism, Microarray Analysis, Middle Aged, Nuclear Proteins analysis, Prognosis, Proportional Hazards Models, Receptor, ErbB-2 analysis, Biomarkers, Tumor metabolism, Breast Neoplasms metabolism, Carcinoma metabolism, Nuclear Proteins metabolism, Receptor, ErbB-2 metabolism
- 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
42. Small interfering RNA targeted to IGF-IR delays tumor growth and induces proinflammatory cytokines in a mouse breast cancer model.
- Author
-
Durfort T, Tkach M, Meschaninova MI, Rivas MA, Elizalde PV, Venyaminova AG, Schillaci R, and François JC
- Subjects
- Animals, Cell Cycle Checkpoints genetics, Cell Cycle Checkpoints immunology, Cell Line, Tumor, Cell Proliferation, Female, Gene Silencing, Humans, Inflammation metabolism, Mammary Neoplasms, Experimental genetics, Mammary Neoplasms, Experimental immunology, Mice, Signal Transduction genetics, Signal Transduction immunology, Transfection, Cytokines metabolism, Inflammation Mediators metabolism, Mammary Neoplasms, Experimental metabolism, Mammary Neoplasms, Experimental pathology, RNA, Small Interfering genetics, Receptor, IGF Type 1 deficiency, Receptor, IGF Type 1 genetics
- Abstract
Insulin-like growth factor I (IGF-I) and its type I receptor (IGF-IR) play significant roles in tumorigenesis and in immune response. Here, we wanted to know whether an RNA interference approach targeted to IGF-IR could be used for specific antitumor immunostimulation in a breast cancer model. For that, we evaluated short interfering RNA (siRNAs) for inhibition of in vivo tumor growth and immunological stimulation in immunocompetent mice. We designed 2'-O-methyl-modified siRNAs to inhibit expression of IGF-IR in two murine breast cancer cell lines (EMT6, C4HD). Cell transfection of IGF-IR siRNAs decreased proliferation, diminished phosphorylation of downstream signaling pathway proteins, AKT and ERK, and caused a G0/G1 cell cycle block. The IGF-IR silencing also induced secretion of two proinflammatory cytokines, TNF- α and IFN-γ. When we transfected C4HD cells with siRNAs targeting IGF-IR, mammary tumor growth was strongly delayed in syngenic mice. Histology of developing tumors in mice grafted with IGF-IR siRNA treated C4HD cells revealed a low mitotic index, and infiltration of lymphocytes and polymorphonuclear neutrophils, suggesting activation of an antitumor immune response. When we used C4HD cells treated with siRNA as an immunogen, we observed an increase in delayed-type hypersensitivity and the presence of cytotoxic splenocytes against wild-type C4HD cells, indicative of evolving immune response. Our findings show that silencing IGF-IR using synthetic siRNA bearing 2'-O-methyl nucleotides may offer a new clinical approach for treatment of mammary tumors expressing IGF-IR. Interestingly, our work also suggests that crosstalk between IGF-I axis and antitumor immune response can mobilize proinflammatory cytokines.
- Published
- 2012
- Full Text
- View/download PDF
43. Novel role of signal transducer and activator of transcription 3 as a progesterone receptor coactivator in breast cancer.
- Author
-
Proietti CJ, Béguelin W, Flaqué MC, Cayrol F, Rivas MA, Tkach M, Charreau EH, Schillaci R, and Elizalde PV
- Subjects
- Animals, Cell Line, Tumor, Cell Nucleus metabolism, Chromatin Immunoprecipitation, Cyclin-Dependent Kinase Inhibitor p21 genetics, Cyclin-Dependent Kinase Inhibitor p21 metabolism, Female, Mice, Mice, Inbred BALB C, Protein Binding, Receptors, Progesterone agonists, Response Elements, Transcriptional Activation, Up-Regulation, bcl-X Protein genetics, bcl-X Protein metabolism, Adenocarcinoma metabolism, Mammary Neoplasms, Experimental metabolism, Receptors, Progesterone metabolism, STAT3 Transcription Factor metabolism
- Abstract
Interactions between progesterone receptor (PR) and signal transducer and activator of transcription 3 (Stat3)-mediated signaling pathways have already been described. In the present study, we explored the capacity of Stat3 to functionally interact with progesterone receptor (PR) and modulate PR transcriptional activation in breast cancer cells. We found that the synthetic progestin medroxyprogesterone acetate (MPA) induced the association of a PR/Stat3 complex in which Stat3 acts as a coactivator of PR. We demonstrated that Stat3 activation is required for MPA modulation of the endogenous genes bcl-X and p21(CIP1) which are involved in MPA-induced cell cycle regulation. Stat3 activity as a coactivator of PR was observed in both the classical and nonclassical ligand activated-PR transcriptional mechanisms, since the effects described were identified in the bcl-X promoter which contains a progesterone responsive element and in the p21(CIP1) promoter which carries Sp1 binding sites where PR is recruited via the transcription factor Sp1. The data herein presented identifies a potential therapeutic intervention for PR-positive breast tumors consisting of targeting Stat3 function or PR/Stat3 interaction which will result in the inhibition of PR function., (Copyright © 2010 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
44. Progesterone receptor induces ErbB-2 nuclear translocation to promote breast cancer growth via a novel transcriptional effect: ErbB-2 function as a coactivator of Stat3.
- Author
-
Béguelin W, Díaz Flaqué MC, Proietti CJ, Cayrol F, Rivas MA, Tkach M, Rosemblit C, Tocci JM, Charreau EH, Schillaci R, and Elizalde PV
- Subjects
- Active Transport, Cell Nucleus drug effects, Animals, Base Sequence, Breast Neoplasms genetics, Breast Neoplasms pathology, Cell Line, Tumor, Cell Proliferation, Female, Gene Knockdown Techniques, Genes, bcl-1, Humans, Mammary Neoplasms, Experimental genetics, Mammary Neoplasms, Experimental pathology, Medroxyprogesterone Acetate toxicity, Mice, Mice, Inbred BALB C, Progestins toxicity, Promoter Regions, Genetic, RNA, Small Interfering genetics, Receptor, ErbB-2 antagonists & inhibitors, Receptor, ErbB-2 genetics, Receptors, Progesterone antagonists & inhibitors, Receptors, Progesterone genetics, STAT3 Transcription Factor antagonists & inhibitors, STAT3 Transcription Factor genetics, Signal Transduction, Transcription, Genetic drug effects, Breast Neoplasms etiology, Breast Neoplasms metabolism, Mammary Neoplasms, Experimental etiology, Mammary Neoplasms, Experimental metabolism, Receptor, ErbB-2 metabolism, Receptors, Progesterone metabolism, STAT3 Transcription Factor metabolism, Trans-Activators metabolism
- Abstract
Progesterone receptor (PR) and ErbB-2 bidirectional cross talk participates in breast cancer development. Here, we identified a new mechanism of the PR and ErbB-2 interaction involving the PR induction of ErbB-2 nuclear translocation and the assembly of a transcriptional complex in which ErbB-2 acts as a coactivator of Stat3. We also highlighted that the function of ErbB-2 as a Stat3 coactivator drives progestin-induced cyclin D1 promoter activation. Notably, PR is also recruited together with Stat3 and ErbB-2 to the cyclin D1 promoter, unraveling a new and unexpected nonclassical PR genomic mechanism. The assembly of the nuclear Stat3/ErbB-2 transcriptional complex plays a key role in the proliferation of breast tumors with functional PR and ErbB-2. Our findings reveal a novel therapeutic intervention for PR- and ErbB-2-positive breast tumors via the specific blockage of ErbB-2 nuclear translocation.
- Published
- 2010
- Full Text
- View/download PDF
45. 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
46. 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
47. 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
48. Progestin effects on breast cancer cell proliferation, proteases activation, and in vivo development of metastatic phenotype all depend on progesterone receptor capacity to activate cytoplasmic signaling pathways.
- Author
-
Carnevale RP, Proietti CJ, Salatino M, Urtreger A, Peluffo G, Edwards DP, Boonyaratanakornkit V, Charreau EH, Bal de Kier Joffé E, Schillaci R, and Elizalde PV
- Subjects
- Animals, Breast Neoplasms genetics, Cell Line, Tumor, Cell Proliferation drug effects, Cytoplasm metabolism, Matrix Metalloproteinase 2 metabolism, Matrix Metalloproteinase 9 metabolism, Mice, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Mitogen-Activated Protein Kinase Kinases metabolism, Neoplasm Metastasis, Phosphatidylinositol 3-Kinases metabolism, Phosphoinositide-3 Kinase Inhibitors, Receptors, Progesterone genetics, Signal Transduction, Urokinase-Type Plasminogen Activator metabolism, Breast Neoplasms metabolism, Breast Neoplasms pathology, Peptide Hydrolases metabolism, Progestins pharmacology, Receptors, Progesterone metabolism
- Abstract
Accumulating evidence indicates that progestins are involved in controlling mammary gland tumorigenesis. Here, we assessed the molecular mechanisms of progestin action in breast cancer models with different phenotypes. We examined C4HD cells, an estrogen (ER) and progesterone (PR) receptor-positive murine breast cancer model in which progestins exert sustained proliferative response, the LM3 murine metastatic mammary tumor cell line, which lacks PR and ER expression, and human PR null T47D-Y breast cancer cells. In addition to acting as a transcription factor, PR can also function as an activator of signaling pathways. To explore which of these two functions were involved in progestin responses, reconstitution experiments in the PR-negative models were performed with wild-type PR-B, with a DNA binding mutant C587A-PR, and with mutant PR-BmPro, which lacks the ability to activate cytoplasm signaling pathways. We found that in a cell context either ER-positive or -negative, progestins induced cell growth and modulation of matrix metalloproteinases-9 (MMP-9) and -2 (MMP-2), and urokinase-type plasminogen activator (uPA) activities, via MAPK and phosphatidylinositol 3-kinase/Akt pathways, in cells expressing wild-type PR-B or DNA binding mutant C587A-PR. In contrast, in cells expressing mutant PR-BmPro, progestins did not induce growth. We also found that unliganded PR expression conferred breast cancer cells an in vitro less proliferative phenotype, as compared with cells lacking PR expression. Modulation of this behavior occurred when PR was functioning either as transcription factor or as signaling activator. Finally, we for the first time demonstrated that progestins favor development of breast tumor metastasis via PR function as activator of signaling pathways. Our present findings provide mechanistic support to the design of a novel therapeutic intervention in PR-positive breast tumors involving blockage of PR capacity to activate cytoplasmic signaling.
- Published
- 2007
- Full Text
- View/download PDF
49. Progestin-induced caveolin-1 expression mediates breast cancer cell proliferation.
- Author
-
Salatino M, Beguelin W, Peters MG, Carnevale R, Proietti CJ, Galigniana MD, Vedoy CG, Schillaci R, Charreau EH, Sogayar MC, and Elizalde PV
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Caveolin 1 genetics, Female, MAP Kinase Signaling System, Mice, Mice, Inbred BALB C, Molecular Sequence Data, Phosphatidylinositol 3-Kinases physiology, Phosphorylation, Promoter Regions, Genetic, Receptors, Progesterone drug effects, Receptors, Progesterone physiology, src-Family Kinases physiology, Caveolin 1 physiology, Gene Expression Regulation, Neoplastic drug effects, Mammary Neoplasms, Experimental pathology, Medroxyprogesterone Acetate pharmacology
- Abstract
Progestin regulation of gene expression was assessed in the progestin-dependent murine tumor line C4HD which requires MPA, a synthetic progestin, for in vivo growth and expresses high levels of progesterone receptor (PR). By using suppressive subtractive hybridization, caveolin-1 was identified as a gene whose expression was increased with in vivo MPA treatment. By Northern and Western blot analysis, we further confirmed that caveolin-1 mRNA and protein expression increased in MPA-treated tumors as compared with untreated tumors. When primary cultures of C4HD cells were treated in vitro with MPA, caveolin-1 levels also increased, effect that was abolished by pre-treatment with progestin antagonist RU486. In addition, MPA promoted strong caveolin-1 promoter transcriptional activation both in mouse and human breast cancer cells. We also showed that MPA regulation of caveolin-1 expression involved in activation of two signaling pathways: MAPK and PI-3K. Short-term MPA treatment of C4HD cells led to tyrosine phosphorylation of caveolin-1 protein, where Src was the kinase involved. Additionally, we showed that MPA-induced association of caveolin-1 and PR, which was detected by coimmunoprecipitation and by confocal microscopy. Finally, we proved that MPA-induced proliferation of C4HD cells was inhibited by suppression of caveolin-1 expression with antisense oligodeoxynucleotides to caveolin-1 mRNA. Furthermore, we observed that inhibition of caveolin-1 expression abrogated PR capacity to induced luciferase activity from a progesterone response element-driven reporter plasmid. Comprehensively, our results demonstrated for the first time that caveolin-1 expression is upregulated by progestin in breast cancer. We also demonstrated that caveolin-1 is a downstream effector of MPA that is partially responsible for the stimulation of growth of breast cancer cells.
- Published
- 2006
- Full Text
- View/download PDF
50. Immunization with murine breast cancer cells treated with antisense oligodeoxynucleotides to type I insulin-like growth factor receptor induced an antitumoral effect mediated by a CD8+ response involving Fas/Fas ligand cytotoxic pathway.
- Author
-
Schillaci R, Salatino M, Cassataro J, Proietti CJ, Giambartolomei GH, Rivas MA, Carnevale RP, Charreau EH, and Elizalde PV
- Subjects
- Animals, B7-2 Antigen metabolism, Breast Neoplasms genetics, Breast Neoplasms metabolism, Cancer Vaccines immunology, Cell Proliferation, Cells, Cultured, Fas Ligand Protein, Female, HSP70 Heat-Shock Proteins metabolism, Immunization, Interferon-gamma biosynthesis, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Neoplasm Transplantation, Oligodeoxyribonucleotides, Antisense genetics, Phenotype, Receptor, IGF Type 1 genetics, Receptor, IGF Type 1 metabolism, T-Lymphocytes, Cytotoxic metabolism, Apoptosis, Breast Neoplasms immunology, Breast Neoplasms pathology, Membrane Glycoproteins metabolism, Receptor, IGF Type 1 deficiency, T-Lymphocytes, Cytotoxic immunology, Tumor Necrosis Factors metabolism, fas Receptor metabolism
- Abstract
We have demonstrated that in vivo administration of phosphorothioate antisense oligodeoxynucleotides (AS[S]ODNs) to type I insulin-like growth factor receptor (IGF-IR) mRNA resulted in inhibition of C4HD breast cancer growth in BALB/c mice. The present study focused on whether in vivo administration of C4HD tumor cells pretreated with IGF-IR AS[S]ODN and irradiated could provide protection against C4HD wild-type tumor challenge and also on elucidating the mechanism mediating this effect. Our results showed that mice immunized with IGF-IR AS[S]ODN-treated C4HD cells experienced a growth inhibition of 53.4%, 61.6%, and 60.2% when compared with PBS-treated mice, wild-type C4HD cell-injected mice, or phosphorothioate sense oligodeoxynucleotide-treated C4HD cell-injected mice, respectively. The protective effect was C4HD-specific, because no cross-protection was observed against other syngeneic mammary tumor lines. The lack of protection against tumor formation in nude mice indicated that T cells were involved in the antitumoral response. Furthermore, cytotoxicity and splenocyte proliferation assays demonstrated that a cellular CD8(+)-dependent immune response, acting through the Fas/Fas ligand death pathway, could be mediating the antitumor effect induced by immunization with AS[S]ODN-treated cells. Immunization also induced splenocytes to produce Ag-dependent IFN-gamma, indicating the presence of a type 1 response. We demonstrated for the first time that IGF-IR AS[S]ODN treatment of breast cancer cells induced expression of CD86 and heat shock protein 70 molecules, both involved in the induction of the immunogenic phenotype. Immunization with these tumor immunogens imparted protection against parental tumor growth through activation of a specific immune response.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.