Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Oncotarget ; 10(41): 4249-4251, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31289622

ABSTRACT

[This corrects the article DOI: 10.18632/oncotarget.12858.].

2.
Oncotarget ; 7(47): 77205-77224, 2016 Nov 22.
Article in English | MEDLINE | ID: mdl-27791205

ABSTRACT

Activation of the IFN/STAT1 pathway is closely associated with drug response and recurrence of breast cancer treated by chemotherapy. The aim of the current study was to elucidate the molecular mechanisms involved upstream and downstream of this pathway in order to identify distinct entities that might be manipulated to improve treatment efficacy. Four breast cancer cell lines (T-47D, MCF7, MDA-MB-231 and HBCx-19 established from the eponymous PDX) were treated in vitro with mafosfamide, a DNA damage inducer. In two of these cell lines (MCF7 and HBCx-19), genotoxic treatment upregulated type I IFN expression leading to paracrine activation of IFN/STAT1 signaling pathway after 6-8 days. We show that STING, a well-characterized inducer of IFN in immune cells, is rapidly triggered in MCF7 cells under genotoxic stress and forms nuclear foci that co-localize with phosphorylated IRF-3 and γH2AX. STING silencing abrogated chemotherapy-induced type I IFN production and signaling and potentiated genotoxic treatment efficacy as it promoted cell death extent and delayed cell colony regrowth. Similar results were obtained after silencing PARP12, one selected gene of the IFN/STAT1 pathway fingerprint. In summary, this study provides the first demonstration of STING activation in breast cancer cells. Our data suggest that genotoxic-induced, STING-mediated type I IFN signaling is a cell-intrinsic mechanism of breast cancer cell survival and regrowth.


Subject(s)
Breast Neoplasms/genetics , Cyclophosphamide/analogs & derivatives , Membrane Proteins/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Survival , Cyclophosphamide/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interferons/metabolism , MCF-7 Cells , Paracrine Communication/drug effects , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , Up-Regulation
3.
Br J Cancer ; 114(2): 177-87, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26695443

ABSTRACT

BACKGROUND: Oestrogen receptor-negative (ER-) breast cancer is intrinsically sensitive to chemotherapy. However, tumour response is often incomplete, and relapse occurs with high frequency. The aim of this work was to analyse the molecular characteristics of residual tumours and early response to chemotherapy in patient-derived xenografts (PDXs) of breast cancer. METHODS: Gene and protein expression profiles were analysed in a panel of ER- breast cancer PDXs before and after chemotherapy treatment. Tumour and stromal interferon-gamma expression was measured in xenografts lysates by human and mouse cytokine arrays, respectively. RESULTS: The analysis of residual tumour cells in chemo-responder PDX revealed a strong overexpression of IFN-inducible genes, induced early after AC treatment and associated with increased STAT1 phosphorylation, DNA-damage and apoptosis. No increase in IFN-inducible gene expression was observed in chemo-resistant PDXs upon chemotherapy. Overexpression of IFN-related genes was associated with human IFN-γ secretion by tumour cells. CONCLUSIONS: Treatment-induced activation of the IFN/STAT1 pathway in tumour cells is associated with chemotherapy response in ER- breast cancer. Further validations in prospective clinical trials will aim to evaluate the usefulness of this signature to assist therapeutic strategies in the clinical setting.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Interferon-gamma/drug effects , Receptors, Estrogen/metabolism , STAT1 Transcription Factor/drug effects , Adaptor Proteins, Signal Transducing , Animals , Antigens/drug effects , Antigens/genetics , Antigens/metabolism , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Capecitabine/pharmacology , Carrier Proteins/drug effects , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 3/drug effects , Caspase 3/genetics , Caspase 3/metabolism , Caspase 7/drug effects , Caspase 7/genetics , Caspase 7/metabolism , Cisplatin/pharmacology , Cytokines/drug effects , Cytokines/genetics , Cytokines/metabolism , Cytoskeletal Proteins/drug effects , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Female , Gene Expression Profiling , Humans , Immunohistochemistry , In Situ Hybridization , Interferon-beta/drug effects , Interferon-beta/genetics , Interferon-beta/metabolism , Interferon-gamma/genetics , Interferon-gamma/metabolism , Intracellular Signaling Peptides and Proteins/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Nude , Mitochondrial Proteins/drug effects , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Myxovirus Resistance Proteins/drug effects , Myxovirus Resistance Proteins/genetics , Myxovirus Resistance Proteins/metabolism , Neoplasm Transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...