Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Oxid Med Cell Longev ; 2017: 1864578, 2017.
Article in English | MEDLINE | ID: mdl-29410730

ABSTRACT

NF-E2-related factor 2 (NRF2) regulates the transcription of a battery of metabolic and cytoprotective genes. NRF2 and epidermal growth factor receptors (EGFRs/HERs) are regulators of cellular proliferation and determinants of cancer initiation and progression. NRF2 and HERs confer cancers with resistance to several therapeutic agents. Nevertheless, there is limited understanding of the regulation of HER expression and activation and the link between NRF2 and HER signalling pathways. We show that NRF2 regulates both basal and inducible expression of HER1, as treatment of ovarian cancer cells (PEO1, OVCAR3, and SKOV3) with NRF2 activator tBHQ inducing HER1, while inhibition of NRF2 by siRNA knockdown or with retinoid represses HER1. Furthermore, treatment of cells with tBHQ increased total and phosphorylated NRF2, HER1, and AKT levels and compromised the cytotoxic effect of lapatinib or erlotinib. Treatment with siRNA or retinoid antagonised the effect of tBHQ on NRF2 and HER1 levels and enhanced the sensitivity of ovarian cancer cells to lapatinib or erlotinib. Pharmacological or genetic inhibition of NRF2 and/or treatment with lapatinib or erlotinib elevated cellular ROS and depleted glutathione. This extends the understanding of NRF2 and its regulation of HER family receptors and opens a strategic target for improving cancer therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , ErbB Receptors/metabolism , Erlotinib Hydrochloride/pharmacology , NF-E2-Related Factor 2/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Quinazolines/pharmacology , Bexarotene , Cell Line, Tumor , Down-Regulation/drug effects , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Erlotinib Hydrochloride/administration & dosage , Female , Humans , Lapatinib , MCF-7 Cells , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/genetics , Ovarian Neoplasms/pathology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Quinazolines/administration & dosage , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Signal Transduction , Tetrahydronaphthalenes/administration & dosage , Tetrahydronaphthalenes/pharmacology
2.
Oncotarget ; 7(46): 75874-75901, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27713148

ABSTRACT

Nuclear erythroid related factor-2 (NRF2) is known to promote cancer therapeutic detoxification and crosstalk with growth promoting pathways. HER2 receptor tyrosine kinase is frequently overexpressed in cancers leading to uncontrolled receptor activation and signaling. A combination of HER2 targeting monoclonal antibodies shows greater anticancer efficacy than the single targeting antibodies, however, its mechanism of action is largely unclear. Here we report novel actions of anti-HER2 drugs, Trastuzumab and Pertuzumab, involving NRF2.HER2 targeting by antibodies inhibited growth in association with persistent generation of reactive oxygen species (ROS), glutathione (GSH) depletion, reduction in NRF2 levels and inhibition of NRF2 function in ovarian cancer cell lines. The combination of antibodies produced more potent effects than single antibody alone; downregulated NRF2 substrates by repressing the Antioxidant Response (AR) pathway with concomitant transcriptional inhibition of NRF2. We showed the antibody combination produced increased methylation at the NRF2 promoter consistent with repression of NRF2 antioxidant function, as HDAC and methylation inhibitors reversed such produced transcriptional effects. These findings demonstrate a novel mechanism and role for NRF2 in mediating the response of cancer cells to the combination of Trastuzumab and Pertuzumab and reinforce the importance of NRF2 in drug resistance and as a key anticancer target.


Subject(s)
NF-E2-Related Factor 2/metabolism , Ovarian Neoplasms/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers , Cell Line, Tumor , Cell Proliferation/drug effects , Computational Biology/methods , CpG Islands , DNA Methylation , Drug Synergism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks , Glutathione/metabolism , Histone Deacetylases/metabolism , Humans , Immunotherapy , NF-E2-Related Factor 2/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Phosphorylation , Promoter Regions, Genetic , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Reactive Oxygen Species/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction/drug effects
3.
Oxid Med Cell Longev ; 2016: 4148791, 2016.
Article in English | MEDLINE | ID: mdl-26770651

ABSTRACT

NF-E2 related factor-2 (NRF2) is an essential transcription factor for multiple genes encoding antioxidants and detoxification enzymes. NRF2 is implicated in promoting cancer therapeutic resistance by its detoxification function and crosstalk with proproliferative pathways. However, the exact mechanism of this intricate connectivity between NRF2 and growth factor induced proliferative pathway remains elusive. Here, we have demonstrated that pharmacological activation of NRF2 by tert-butylhydroquinone (tBHQ) upregulates the HER family receptors, HER2 and HER3 expression, elevates pAKT levels, and enhances the proliferation of ovarian cancer cells. Preactivation of NRF2 also attenuates the combined growth inhibitory effects of HER2 targeting monoclonal antibodies, Pertuzumab and Trastuzumab. Further, tBHQ caused transcriptional induction of HER2 and HER3, while SiRNA-mediated knockdown of NRF2 prevented this and further caused transcriptional repression and enhanced cytotoxicity of the HER2 inhibitors. Hence, NRF2 regulates both HER2 and HER3 receptors to influence cellular responses to HER2 targeting monoclonal antibodies. This deciphered crosstalk mechanism reinforces the role of NRF2 in drug resistance and as a relevant anticancer target.


Subject(s)
Immunotherapy , Molecular Targeted Therapy , NF-E2-Related Factor 2/metabolism , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Signal Transduction , Antibodies, Monoclonal, Humanized/pharmacology , Antioxidant Response Elements/genetics , Base Sequence , Cell Line, Tumor , Cell Proliferation/drug effects , Computer Simulation , Cytoprotection/drug effects , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Heme Oxygenase-1/metabolism , Humans , Hydroquinones/pharmacology , Molecular Sequence Data , Phosphorylation/drug effects , Phosphoserine/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-3/genetics , Signal Transduction/drug effects , Trastuzumab/pharmacology
4.
J Biotechnol ; 202: 40-9, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25747275

ABSTRACT

Seliciclib (R-Roscovitine) was identified as an inhibitor of CDKs and has undergone drug development and clinical testing as an anticancer agent. In this review, the authors describe the discovery of Seliciclib and give a brief summary of the biology of the CDKs Seliciclib inhibits. An overview of the published in vitro and in vivo work supporting the development as an anti-cancer agent, from in vitro experiments to animal model studies ending with a summary of the clinical trial results and trials underway is presented. In addition some potential non-oncology applications are explored and the potential mode of action of Seliciclib in these areas is described. Finally the authors argue that optimisation of the therapeutic effects of kinase inhibitors such as Seliciclib could be enhanced using a systems biology approach involving mathematical modelling of the molecular pathways regulating cell growth and division.


Subject(s)
Drug Repositioning/methods , Protein Kinase Inhibitors/therapeutic use , Purines/therapeutic use , Systems Biology/methods , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Proliferation/drug effects , Clinical Trials as Topic , Cyclin-Dependent Kinases/antagonists & inhibitors , Humans , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Roscovitine
5.
J Biotechnol ; 202: 12-30, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25449014

ABSTRACT

Cells are constantly exposed to Reactive Oxygen Species (ROS) produced both endogenously to meet physiological requirements and from exogenous sources. While endogenous ROS are considered as important signalling molecules, high uncontrollable ROS are detrimental. It is unclear how cells can achieve a balance between maintaining physiological redox homeostasis and robustly activate the antioxidant system to remove exogenous ROS. We have utilised a Systems Biology approach to understand how this robust adaptive system fulfils homeostatic requirements of maintaining steady-state ROS and growth rate, while undergoing rapid readjustment under challenged conditions. Using a panel of human ovarian and normal cell lines, we experimentally quantified and established interrelationships between key elements of ROS homeostasis. The basal levels of NRF2 and KEAP1 were cell line specific and maintained in tight correlation with their growth rates and ROS. Furthermore, perturbation of this balance triggered cell specific kinetics of NRF2 nuclear-cytoplasmic relocalisation and sequestration of exogenous ROS. Our experimental data were employed to parameterise a mathematical model of the NRF2 pathway that elucidated key response mechanisms of redox regulation and showed that the dynamics of NRF2-H2O2 regulation defines a relationship between half-life, total and nuclear NRF2 level and endogenous H2O2 that is cell line specific.


Subject(s)
Hydrogen Peroxide/pharmacokinetics , Intracellular Signaling Peptides and Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Ovarian Neoplasms/pathology , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Female , Gene Regulatory Networks , Humans , Hydrogen Peroxide/metabolism , Kelch-Like ECH-Associated Protein 1 , Models, Genetic , Ovarian Neoplasms/metabolism , Oxidation-Reduction , Signal Transduction , Systems Biology
6.
Cells ; 3(2): 563-91, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24918976

ABSTRACT

The receptor tyrosine kinases (RTKs) are key drivers of cancer progression and targets for drug therapy. A major challenge in anti-RTK treatment is the dependence of drug effectiveness on co-expression of multiple RTKs which defines resistance to single drug therapy. Reprogramming of the RTK network leading to alteration in RTK co-expression in response to drug intervention is a dynamic mechanism of acquired resistance to single drug therapy in many cancers. One route to overcome this resistance is combination therapy. We describe the results of a joint in silico, in vitro, and in vivo investigations on the efficacy of trastuzumab, pertuzumab and their combination to target the HER2 receptors. Computational modelling revealed that these two drugs alone and in combination differentially suppressed RTK network activation depending on RTK co-expression. Analyses of mRNA expression in SKOV3 ovarian tumour xenograft showed up-regulation of HER3 following treatment. Considering this in a computational model revealed that HER3 up-regulation reprograms RTK kinetics from HER2 homodimerisation to HER3/HER2 heterodimerisation. The results showed synergy of the trastuzumab and pertuzumab combination treatment of the HER2 overexpressing tumour can be due to an independence of the combination effect on HER3/HER2 composition when it changes due to drug-induced RTK reprogramming.

7.
Exp Biol Med (Maywood) ; 237(6): 622-34, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22728709

ABSTRACT

Ataxia-telangiectasia mutated (ATM) kinase is a component of a signalling mechanism that determines the process of decision-making in response to DNA damage and involves the participation of multiple proteins. ATM is activated by DNA double-strand breaks (DSBs) through the Mre11-Rad50-Nbs1 (MRN) DNA repair complex, and orchestrates signalling cascades that initiate the DNA damage response. Cells lacking ATM are hypersensitive to insults, particularly genotoxic stress, induced through radiation or radiomimetic drugs. Here, we investigate the degree of ATM activation during time-dependent treatment with genotoxic agents and the effects of ATM on phospho-induction and localization of its downstream substrates. Additionally, we have demonstrated a new cell-cycle-independent mechanism of ATM gene regulation following ATM kinase inhibition with KU5593. Inhibition of ATM activity causes induction of ATM protein followed by oscillation and this mechanism is governed at the transcriptional level. Furthermore, this autoregulatory induction of ATM is also accompanied by a transient upregulation of p53, pATR and E2F1 levels. Since ATM inhibition is believed to sensitize cancer cells to genotoxic agents, this novel insight into the mechanism of ATM regulation might be useful for designing more precise strategies for modulation of ATM activity in cancer therapy.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/drug effects , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/drug effects , Enzyme Inhibitors/pharmacology , Morpholines/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/drug effects , Pyrones/pharmacology , Transcriptional Activation/drug effects , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/drug effects , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Ataxia Telangiectasia Mutated Proteins , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Cell Line, Tumor , Cells, Cultured , DNA Damage , DNA-Binding Proteins/genetics , E2F1 Transcription Factor/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...