Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Oncotarget ; 9(22): 15860-15875, 2018 Mar 23.
Article in English | MEDLINE | ID: mdl-29662612

ABSTRACT

Neurofibromatosis type 1 is a disease caused by mutation of neurofibromin 1 (NF1), loss of which results in hyperactive Ras signaling and a concomitant increase in cell proliferation and survival. Patients with neurofibromatosis type 1 frequently develop tumors such as plexiform neurofibromas and malignant peripheral nerve sheath tumors. Mutation of NF1 or loss of the NF1 protein is also observed in glioblastoma, lung adenocarcinoma, and ovarian cancer among other sporadic cancers. A therapy that selectively targets NF1 deficient tumors would substantially advance our ability to treat these malignancies. To address the need for these therapeutics, we developed and conducted a synthetic lethality screen to discover molecules that target yeast lacking the homolog of NF1, IRA2. One of the lead candidates that was observed to be synthetic lethal with ira2Δ yeast is Y100. Here, we describe the mechanisms by which Y100 targets ira2Δ yeast and NF1-deficient tumor cells. Y100 treatment disrupted proteostasis, metabolic homeostasis, and induced the formation of mitochondrial superoxide in NF1-deficient cancer cells. Previous studies also indicate that NF1/Ras-dysregulated tumors may be sensitive to modulators of oxidative and ER stress. We hypothesize that the use of Y100 and molecules with related mechanisms of action represent a feasible therapeutic strategy for targeting NF1 deficient cells.

2.
Curr Protoc Immunol ; 109: 9.10.1-9.10.8, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25845565

ABSTRACT

Proteasome inhibitors are indispensable research tools in immunology and cell biology. With numerous proteasome inhibitors available commercially, choosing the appropriate compound for a biological experiment may be challenging, especially for a novice. This unit provides an overview of the proteasome inhibitors commonly used in research. It discusses how to select an appropriate highly specific inhibitor, its concentration, and length of exposure for mammalian cell culture experiments. In addition, assays that can be used to confirm proteasome inhibition are discussed.


Subject(s)
Proteasome Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Humans , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/chemistry , Proteasome Inhibitors/classification , Proteolysis , Research , Signal Transduction/drug effects , Ubiquitin/metabolism
3.
Anal Biochem ; 451: 1-3, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24486333

ABSTRACT

Proteasome-Glo is a homogeneous cell-based assay of proteasomal chymotrypsin-like, trypsin-like, and caspase-like activities using luminogenic substrates, commercially available from Promega. Here we report that the background activity from cleavage of the substrate of the trypsin-like sites by nonproteasomal proteases in multiple breast and lung cancer cell lines exceeds the activity of the proteasome. We also observed substantial background chymotrypsin-like activity in some cell lines. Thus, Proteasome-Glo assay must be used with caution, and it is necessary to include a specific proteasome inhibitor to determine the background for each proteasome activity.


Subject(s)
Luminescent Measurements , Proteasome Endopeptidase Complex/analysis , Cell Line, Tumor , Fluorescent Dyes/chemistry , HeLa Cells , Humans , Proteasome Endopeptidase Complex/metabolism , Substrate Specificity , Trypsin/analysis , Trypsin/metabolism
4.
PLoS One ; 8(2): e56132, 2013.
Article in English | MEDLINE | ID: mdl-23460792

ABSTRACT

The proteasome inhibitor bortezomib (Velcade) is prescribed for the treatment of multiple myeloma. Clinically achievable concentrations of bortezomib cause less than 85% inhibition of the chymotrypsin-like activity of the proteasome, but little attention has been paid as to whether in vitro studies are representative of this level of inhibition. Patients receive bortezomib as an intravenous or subcutaneous bolus injection, resulting in maximum proteasome inhibition within one hour followed by a gradual recovery of activity. In contrast, most in vitro studies use continuous treatment so that activity never recovers. Replacing continuous treatment with 1 h-pulse treatment increases differences in sensitivity in a panel of 7 multiple myeloma cell lines from 5.3-fold to 18-fold, and reveals that the more sensitive cell lines undergo apoptosis at faster rates. Clinically achievable inhibition of active sites was sufficient to induce cytotoxicity only in one cell line. At concentrations of bortezomib that produced similar inhibition of peptidase activities a different extent of inhibition of protein degradation was observed, providing an explanation for the differential sensitivity. The amount of protein degraded per number of active proteasomes correlated with sensitivity to bortezomib. Thus, (i) in vitro studies of proteasome inhibitors should be conducted at pharmacologically achievable concentrations and duration of treatment; (ii) a similar level of inhibition of active sites results in a different extent of inhibition of protein breakdown in different cell lines, and hence a difference in sensitivity.


Subject(s)
Boronic Acids/therapeutic use , Multiple Myeloma/drug therapy , Pyrazines/therapeutic use , Apoptosis/drug effects , Boronic Acids/pharmacology , Bortezomib , Cell Death/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Neoplasm Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Proteolysis/drug effects , Pyrazines/pharmacology , Time Factors
5.
PLoS One ; 7(11): e50066, 2012.
Article in English | MEDLINE | ID: mdl-23166821

ABSTRACT

Genetic analysis of TP63 implicates ΔNp63 isoforms in preservation of replicative capacity and cellular lifespan within adult stem cells. ΔNp63α is also an oncogene and survival factor that mediates therapeutic resistance in squamous carcinomas. These diverse activities are the result of genetic and functional interactions between TP63 and an array of morphogenic and morphostatic signals that govern tissue and tumor stasis, mitotic polarity, and cell fate; however the cellular signals that account for specific functions of TP63 are incompletely understood. To address this we sought to identify signaling pathways that regulate expression, stability or activity of ΔNp63α. An siRNA-based screen of the human kinome identified the Type 1 TGFß receptor, ALK5, as the kinase required for phosphorylation of ΔNp63α at Serine 66/68 (S66/68). This activity is TGFß-dependent and sensitive to either ALK5-directed siRNA or the ALK5 kinase inhibitor A83-01. Mechanistic studies support a model in which ALK5 is proteolytically cleaved at the internal juxtamembrane region resulting in the translocation of the C-terminal ALK5-intracellular kinase domain (ALK5(IKD)). In this study, we demonstrate that ALK5-mediated phosphorylation of ΔNp63α is required for the anti-clonogenic effects of TGFΒ and ectopic expression of ALK5(IKD) mimics these effects. Finally, we present evidence that ultraviolet irradiation-mediated phosphorylation of ΔNp63α is sensitive to ALK5 inhibitors. These findings identify a non-canonical TGFß-signaling pathway that mediates the anti-clonogenic effects of TGFß and the effects of cellular stress via ΔNp63α phosphorylation.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/metabolism , Cells, Cultured , Colony-Forming Units Assay , DNA Primers/genetics , Humans , Models, Biological , Phosphorylation , RNA, Small Interfering/genetics , Receptor, Transforming Growth Factor-beta Type I , Transfection
6.
Chem Biol ; 18(5): 608-18, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21609842

ABSTRACT

Proteasomes degrade the majority of proteins in mammalian cells, are involved in the regulation of multiple physiological functions, and are established targets of anticancer drugs. The proteasome has three types of active sites. Chymotrypsin-like sites are the most important for protein breakdown and have long been considered the only suitable targets for antineoplastic drugs; however, our recent work demonstrated that inhibitors of caspase-like sites sensitize malignant cells to inhibitors of the chymotrypsin-like sites. Here, we describe the development of specific cell-permeable inhibitors and an activity-based probe of the trypsin-like sites. These compounds selectively sensitize multiple myeloma cells to inhibitors of the chymotrypsin-like sites, including antimyeloma agents bortezomib and carfilzomib. Thus, trypsin-like sites are cotargets for anticancers drugs. Together with inhibitors of chymotrypsin- and caspase-like sites developed earlier, we provide the scientific community with a complete set of tools to separately modulate proteasome active sites in living cells.


Subject(s)
Antineoplastic Agents/therapeutic use , Boronic Acids/therapeutic use , Multiple Myeloma/drug therapy , Oligopeptides/therapeutic use , Protease Inhibitors/therapeutic use , Proteasome Inhibitors , Pyrazines/therapeutic use , Antineoplastic Agents/chemistry , Boronic Acids/chemistry , Bortezomib , Catalytic Domain , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Humans , Oligopeptides/chemistry , Protease Inhibitors/chemistry , Proteasome Endopeptidase Complex/metabolism , Pyrazines/chemistry , Trypsin/metabolism
7.
J Biol Chem ; 285(51): 40125-34, 2010 Dec 17.
Article in English | MEDLINE | ID: mdl-20937826

ABSTRACT

Proteasomes degrade most proteins in mammalian cells and are established targets of anti-cancer drugs. The majority of proteasome inhibitors are composed of short peptides with an electrophilic functionality (pharmacophore) at the C terminus. All eukaryotic proteasomes have three types of active sites as follows: chymotrypsin-like, trypsin-like, and caspase-like. It is widely believed that active site specificity of inhibitors is determined primarily by the peptide sequence and not the pharmacophore. Here, we report that active site specificity of inhibitors can also be tuned by the chemical nature of the pharmacophore. Specifically, replacement of the epoxyketone by vinyl sulfone moieties further improves the selectivity of ß5-specific inhibitors NC-005, YU-101, and PR-171 (carfilzomib). This increase in specificity is likely the basis of the decreased cytotoxicity of vinyl sulfone-based inhibitors to HeLa cells as compared with that of epoxyketone-based inhibitors.


Subject(s)
Antineoplastic Agents/chemistry , Cytotoxins/chemistry , Protease Inhibitors/chemistry , Proteasome Endopeptidase Complex/chemistry , Proteasome Inhibitors , Sulfones/chemistry , Animals , Antineoplastic Agents/pharmacology , Catalytic Domain , Cytotoxins/pharmacology , HEK293 Cells , HeLa Cells , Humans , Oligopeptides , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex/metabolism , Rabbits , Sulfones/pharmacology
8.
J Med Chem ; 53(5): 2319-23, 2010 Mar 11.
Article in English | MEDLINE | ID: mdl-20131905

ABSTRACT

Proteasomal processing is conducted by three individual catalytic subunits, namely beta1, beta2, and beta5. Subunit-specific inhibitors are useful tools in dissecting the role of these individual subunits and are leads toward the development of antitumor agents. We here report that the presence of fluorinated phenylalanine derivatives in peptide based proteasome inhibitors has a profound effect on inhibitor potency and selectivity. Specifically, compound 4a emerges as one of the most beta5 specific inhibitors known to date.


Subject(s)
Catalytic Domain/drug effects , Chymotrypsin/metabolism , Cysteine Proteinase Inhibitors/chemical synthesis , Phenylalanine/analogs & derivatives , Proteasome Endopeptidase Complex/metabolism , Chymotrypsin/antagonists & inhibitors , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/pharmacology , Inhibitory Concentration 50 , Phenylalanine/chemical synthesis , Phenylalanine/chemistry , Phenylalanine/pharmacology , Proteasome Inhibitors , Structure-Activity Relationship
9.
Chem Biol ; 16(12): 1278-89, 2009 Dec 24.
Article in English | MEDLINE | ID: mdl-20064438

ABSTRACT

Proteasomes degrade most proteins in mammalian cells and are established targets of anticancer drugs. All eukaryotic proteasomes have three types of active sites: chymotrypsin-like, trypsin-like, and caspase-like. Chymotrypsin-like sites are the most important in protein degradation and are the primary target of most proteasome inhibitors. The biological roles of trypsin-like and caspase-like sites and their potential as cotargets of antineoplastic agents are not well defined. Here we describe the development of site-specific inhibitors and active-site probes of chymotrypsin-like and caspase-like sites. Using these compounds, we show that cytotoxicity of proteasome inhibitors does not correlate with inhibition of chymotrypsin-like sites and that coinhibition of either trypsin-like and/or caspase-like sites is needed to achieve maximal cytotoxicity. Thus, caspase-like and trypsin-like sites must be considered as cotargets of anticancer drugs.


Subject(s)
Antineoplastic Agents/chemistry , Protease Inhibitors/chemistry , Proteasome Inhibitors , Antineoplastic Agents/toxicity , Caspases/metabolism , Catalytic Domain , Cell Line , Chymotrypsin/metabolism , Humans , Protease Inhibitors/toxicity , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...