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.
Oncotarget ; 6(19): 17314-27, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-26015393

ABSTRACT

There is a growing body of evidence supporting the use of epigenetic therapies in the treatment of multiple myeloma. We show the novel HDAC inhibitor CHR-3996 induces apoptosis in myeloma cells at concentrations in the nanomolar range and with apoptosis mediated by p53 and caspase pathways. In addition, HDAC inhibitors are highly synergistic, both in vitro and in vivo, with the aminopeptidase inhibitor tosedostat (CHR-2797). We demonstrate that the basis for this synergy is a consequence of changes in the levels of NFκB regulators BIRC3/cIAP2, A20, CYLD, and IκB, which were markedly affected by the combination. When co-administered the HDAC and aminopeptidase inhibitors caused rapid nuclear translocation of NFκB family members p65 and p52, following activation of both canonical and non-canonical NFκB signalling pathways. The subsequent up-regulation of inhibitors of NFκB activation (most significantly BIRC3/cIAP2) turned off the cytoprotective effects of the NFκB signalling response in a negative feedback loop. These results provide a rationale for combining HDAC and aminopeptidase inhibitors clinically for the treatment of myeloma patients and support the disruption of the NFκB signalling pathway as a therapeutic strategy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Azabicyclo Compounds/administration & dosage , Glycine/analogs & derivatives , Hydroxamic Acids/administration & dosage , Multiple Myeloma/pathology , Pyrimidines/administration & dosage , Signal Transduction/drug effects , Aminopeptidases/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Enzyme Inhibitors/administration & dosage , Glycine/administration & dosage , Histone Deacetylase Inhibitors/administration & dosage , Humans , Mice , Mice, Inbred NOD , Mice, SCID , NF-kappa B/drug effects , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays
2.
EMBO J ; 30(5): 894-905, 2011 Mar 02.
Article in English | MEDLINE | ID: mdl-21317875

ABSTRACT

Ire1 (Ern1) is an unusual transmembrane protein kinase essential for the endoplasmic reticulum (ER) unfolded protein response (UPR). Activation of Ire1 by association of its N-terminal ER luminal domains promotes autophosphorylation by its cytoplasmic kinase domain, leading to activation of the C-terminal ribonuclease domain, which splices Xbp1 mRNA generating an active Xbp1s transcriptional activator. We have determined the crystal structure of the cytoplasmic portion of dephosphorylated human Ire1α bound to ADP, revealing the 'phosphoryl-transfer' competent dimeric face-to-face complex, which precedes and is distinct from the back-to-back RNase 'active' conformation described for yeast Ire1. We show that the Xbp1-specific ribonuclease activity depends on autophosphorylation, and that ATP-competitive inhibitors staurosporin and sunitinib, which inhibit autophosphorylation in vitro, also inhibit Xbp1 splicing in vivo. Furthermore, we demonstrate that activated Ire1α is a competent protein kinase, able to phosphorylate a heterologous peptide substrate. These studies identify human Ire1α as a target for development of ATP-competitive inhibitors that will modulate the UPR in human cells, which has particular relevance for myeloma and other secretory malignancies.


Subject(s)
Cell Nucleus/genetics , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum/metabolism , Endoribonucleases/chemistry , Endoribonucleases/metabolism , Gene Expression Regulation , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , RNA Splicing , Transcription Factors/metabolism , Unfolded Protein Response/physiology , Blotting, Western , Crystallography, X-Ray , Cytoplasm , DNA-Binding Proteins/genetics , Endoribonucleases/genetics , Humans , Membrane Proteins/genetics , Phosphorylation , Protein Folding , Protein Multimerization , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , Regulatory Factor X Transcription Factors , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription, Genetic , X-Box Binding Protein 1
3.
Blood ; 116(2): 250-3, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20421453

ABSTRACT

Immunoglobulin production by myeloma plasma cells depends on the unfolded protein response for protein production and folding. Recent studies have highlighted the importance of IRE1alpha and X box binding protein 1 (XBP1), key members of this pathway, in normal B-plasma cell development. We have determined the gene expression levels of IRE1alpha, XBP1, XBP1UNSPLICED (XBP1u), and XBP1SPLICED (XBP1s) in a series of patients with myeloma and correlated findings with clinical outcome. We show that IRE1alpha and XBP1 are highly expressed and that patients with low XBP1s/u ratios have a significantly better overall survival. XBP1s is an independent prognostic marker and can be used with beta2 microglobulin and t(4;14) to identify a group of patients with a poor outcome. Furthermore, we show the beneficial therapeutic effects of thalidomide in patients with low XBP1s/u ratios. This study highlights the importance of XBP1 in myeloma and its significance as an independent prognostic marker and as a predictor of thalidomide response.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , DNA-Binding Proteins/biosynthesis , Drug Resistance, Neoplasm/genetics , Multiple Myeloma/metabolism , Thalidomide/therapeutic use , Transcription Factors/biosynthesis , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Cyclophosphamide/administration & dosage , Cyclophosphamide/therapeutic use , DNA-Binding Proteins/genetics , Dexamethasone/administration & dosage , Dexamethasone/therapeutic use , Disease-Free Survival , Doxorubicin/therapeutic use , Female , Gene Expression , Gene Expression Profiling , Humans , Kaplan-Meier Estimate , Male , Melphalan/therapeutic use , Middle Aged , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Prednisone/therapeutic use , Prognosis , Proportional Hazards Models , Regulatory Factor X Transcription Factors , Transcription Factors/genetics , Vincristine/therapeutic use , X-Box Binding Protein 1
5.
Mol Cancer Ther ; 8(4): 762-70, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19372548

ABSTRACT

Myeloma cells are highly dependent on the unfolded protein response to assemble folded immunoglobulins correctly. Therefore, targeting protein handling within a myeloma cell by inhibiting the aminopeptidase enzyme system, which catalyses the hydrolysis of amino acids from the proteins NH2 terminus, represents a therapeutic approach. CHR-2797, a novel aminopeptidase inhibitor, is able to inhibit proliferation and induce growth arrest and apoptosis in myeloma cells, including cells resistant to conventional chemotherapeutics. It causes minimal inhibition of bone marrow stromal cell (BMSC) proliferation but is able to overcome the microenvironmental protective effects, inhibiting the proliferation of myeloma cells bound to BMSCs and the increase in vascular endothelial growth factor levels seen when myeloma cells and BMSCs are bound together. Additive and synergistic effects are seen with bortezomib, melphalan, and dexamethasone. Apoptosis occurs via both caspase-dependent and non-caspase-dependent pathways with an increase in Noxa, cleavage of Mcl-1, and activation of the unfolded protein response. Autophagy is also seen. CHR-2797 causes an up-regulation of genes involved in the proteasome/ubiquitin pathway, as well as aminopeptidases, and amino acid deprivation response genes. In conclusion, inhibiting protein turnover using the aminopeptidase inhibitor CHR-2797 results in myeloma cell apoptosis and represents a novel therapeutic approach that warrants further investigation in the clinical setting.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Cell Proliferation/drug effects , Glycine/analogs & derivatives , Hydroxamic Acids/pharmacology , Multiple Myeloma/drug therapy , Multiple Myeloma/enzymology , Aminopeptidases/metabolism , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Bone Marrow Cells/enzymology , Bone Marrow Cells/pathology , Caspases/metabolism , Cell Cycle/drug effects , Glycine/pharmacology , Humans , Immunoblotting , Stromal Cells/enzymology , Stromal Cells/pathology , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/metabolism
6.
Cell Cycle ; 7(7): 865-9, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18414035

ABSTRACT

Resistance to current cancer therapies has forced scientists to investigate new avenues of therapy distinct from those aimed at single targets, to strategies based on targeting families of proteins, on which cancers rely for their ability to survive stress. Two such protein families are the heat shock proteins (HSP), especially the HSP90 family, and proteins involved in mediating the unfolded protein response (UPR). HSP90 stabilises key survival factors in cancer cells including AKT, ERB2 and HIF1alpha, which alone makes HSP90 inhibitors extremely interesting as potential therapies. In addition targeting HSP90 can destabilise the UPR inducing cell death. A broad range of cancer-types rely on the UPR to correctly fold key signalling proteins properly, as well as to allow the cell to cope with the hypoxic environment associated with tumour development. These associations suggest that a range of tumours may be targeted using HSP90 inhibitors and that the development of specific inhibitors of the UPR may be of interest. In this article, based on work in multiple myeloma, we highlight the importance of targeting multiple signalling pathways simultaneously, using the UPR and heat shock proteins as examples, as a means of effectively killing cancer cells.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Models, Biological , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Protein Folding , Signal Transduction/genetics , Activating Transcription Factor 6/metabolism , Endoribonucleases/metabolism , Humans , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , eIF-2 Kinase/metabolism
7.
Blood ; 110(7): 2641-9, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17525289

ABSTRACT

Plasma cells producing high levels of paraprotein are dependent on the unfolded protein response (UPR) and chaperone proteins to ensure correct protein folding and cell survival. We hypothesized that disrupting client-chaperone interactions using heat shock protein 90 (Hsp90) inhibitors would result in an inability to handle immunoglobulin production with the induction of the UPR and myeloma cell death. To study this, myeloma cells were treated with Hsp90 inhibitors as well as known endoplasmic reticulum stress inducers and proteasome inhibitors. Treatment with thapsigargin and tunicamycin led to the activation of all 3 branches of the UPR, with early splicing of XBP1 indicative of IRE1 activation, upregulation of CHOP consistent with ER resident kinase (PERK) activation, and activating transcription factor 6 (ATF6) splicing. 17-AAG and radicicol also induced splicing of XBP1, with the induction of CHOP and activation of ATF6, whereas bortezomib resulted in the induction of CHOP and activation of ATF6 with minimal effects on XBP1. After treatment with all drugs, expression levels of the molecular chaperones BiP and GRP94 were increased. All drugs inhibited proliferation and induced cell death with activation of JNK and caspase cleavage. In conclusion, Hsp90 inhibitors induce myeloma cell death at least in part via endoplasmic reticulum stress and the UPR death pathway.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Multiple Myeloma/metabolism , Protein Folding , Apoptosis , Caspases/metabolism , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Humans , Inclusion Bodies/drug effects , Multiple Myeloma/pathology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Thapsigargin/pharmacology , Tunicamycin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...