Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Chembiochem ; 21(22): 3220-3224, 2020 11 16.
Article in English | MEDLINE | ID: mdl-32598532

ABSTRACT

Proteasomes are established therapeutic targets for hematological cancers and promising targets for autoimmune diseases. In the past, we have designed and synthesized mechanism-based proteasome inhibitors that are selective for the individual catalytic activities of human constitutive proteasomes and immunoproteasomes: ß1c, ß1i, ß2c, ß2i, ß5c and ß5i. We show here that by taking the oligopeptide recognition element and substituting the electrophile for a fluorogenic leaving group, fluorogenic substrates are obtained that report on the proteasome catalytic activity also targeted by the parent inhibitor. Though not generally applicable (ß5c and ß2i substrates showing low activity), effective fluorogenic substrates reporting on the individual activity of ß1c, ß1i, ß2c and ß5i subunits in Raji (human B cell) lysates and purified 20S proteasome were identified in this manner. Our work thus adds to the expanding proteasome research toolbox through the identification of new and/or more effective subunit-selective fluorogenic substrates.


Subject(s)
Fluorescent Dyes/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Humans , Hydrolysis , Molecular Structure , Proteasome Endopeptidase Complex/isolation & purification , Proteasome Inhibitors/chemical synthesis , Proteasome Inhibitors/chemistry , Protein Subunits , Substrate Specificity
2.
Angew Chem Int Ed Engl ; 55(13): 4199-203, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26511210

ABSTRACT

Proteasomes are therapeutic targets for various cancers and autoimmune diseases. Constitutively expressed proteasomes have three active sites, ß1c, ß2c, and ß5c. Lymphoid tissues also express the immunoproteasome subunits ß1i, ß2i, and ß5i. Rapid and simultaneous measurement of the activity of these catalytic subunits would assist in the discovery of new inhibitors, improve analysis of proteasome inhibitors in clinical trials, and simplify analysis of subunit expression. In this work, we present a cocktail of activity-based probes that enables simultaneous gel-based detection of all six catalytic human proteasome subunits. We used this cocktail to develop specific inhibitors for ß1c, ß2c, ß5c, and ß2i, to compare the active-site specificity of clinical proteasome inhibitors, and to demonstrate that many hematologic malignancies predominantly express immunoproteasomes. Furthermore, we show that selective and complete inhibition of ß5i and ß1i is cytotoxic to primary cells from acute lymphocytic leukemia (ALL) patients.


Subject(s)
Molecular Probes/chemistry , Proteasome Endopeptidase Complex/metabolism , Catalytic Domain , Humans
3.
Haematologica ; 100(10): 1350-60, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26069288

ABSTRACT

Proteasome inhibitor resistance is a challenge for myeloma therapy. Bortezomib targets the ß5 and ß1 activity, but not the ß2 activity of the proteasome. Bortezomib-resistant myeloma cells down-regulate the activation status of the unfolded protein response, and up-regulate ß2 proteasome activity. To improve proteasome inhibition in bortezomib-resistant myeloma and to achieve more efficient UPR activation, we have developed LU-102, a selective inhibitor of the ß2 proteasome activity. LU-102 inhibited the ß2 activity in intact myeloma cells at low micromolar concentrations without relevant co-inhibition of ß1 and ß5 proteasome subunits. In proteasome inhibitor-resistant myeloma cells, significantly more potent proteasome inhibition was achieved by bortezomib or carfilzomib in combination with LU-102, compared to bortezomib/carfilzomib alone, resulting in highly synergistic cytotoxic activity of the drug combination via endoplasmatic reticulum stress-induced apoptosis. Combining bortezomib/carfilzomib with LU-102 significantly prolonged proteasome inhibition and increased activation of the unfolded protein response and IRE1-a activity. IRE1-α has recently been shown to control myeloma cell differentiation and bortezomib sensitivity (Leung-Hagesteijn, Cancer Cell 24:3, 289-304). Thus, ß2-selective proteasome inhibition by LU-102 in combination with bortezomib or carfilzomib results in synergistic proteasome inhibition, activation of the unfolded protein response, and cytotoxicity, and overcomes bortezomib/carfilzomib resistance in myeloma cells in vitro.


Subject(s)
Antineoplastic Agents/pharmacology , Bortezomib/pharmacology , Drug Resistance, Neoplasm , Oligopeptides/pharmacology , Proteasome Inhibitors/pharmacology , Animals , Cell Line, Tumor , Disease Models, Animal , Drug Synergism , Humans , Mice , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Xenograft Model Antitumor Assays
4.
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
5.
J Immunol ; 189(4): 1868-77, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22772448

ABSTRACT

The proteasome is responsible for the generation of most epitopes presented on MHC class I molecules. Treatment of cells with IFN-γ leads to the replacement of the constitutive catalytic subunits ß1, ß2, and ß5 by the inducible subunits low molecular mass polypeptide (LMP) 2 (ß1i), multicatalytic endopeptidase complex-like-1 (ß2i), and LMP7 (ß5i), respectively. The incorporation of these subunits is required for the production of numerous MHC class I-restricted T cell epitopes. The structural features rather than the proteolytic activity of an immunoproteasome subunit are needed for the generation of some epitopes, but the underlying mechanisms have remained elusive. Experiments with LMP2-deficient splenocytes revealed that the generation of the male HY-derived CTL-epitope UTY(246-254) was dependent on LMP2. Treatment of male splenocytes with an LMP2-selective inhibitor did not reduce UTY(246-254) presentation, whereas silencing of ß1 activity increased presentation of UTY(246-254). In vitro degradation experiments showed that the caspase-like activity of ß1 was responsible for the destruction of this CTL epitope, whereas it was preserved when LMP2 replaced ß1. Moreover, inhibition of the ß5 subunit rescued the presentation of the influenza matrix 58-66 epitope, thus suggesting that a similar mechanism can apply to the exchange of ß5 by LMP7. Taken together, our data provide a rationale why the structural property of an immunoproteasome subunit rather than its activity is required for the generation of a CTL epitope.


Subject(s)
Antigen Presentation/immunology , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/immunology , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/immunology , Animals , Antigens, Viral/immunology , Blotting, Western , Electrophoresis, Gel, Two-Dimensional , Epitopes, T-Lymphocyte/immunology , Humans , Immunoprecipitation , Male , Mass Spectrometry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Reverse Transcriptase Polymerase Chain Reaction , Transfection
6.
Elife ; 122024 Apr 15.
Article in English | MEDLINE | ID: mdl-38619391

ABSTRACT

Rapid recovery of proteasome activity may contribute to intrinsic and acquired resistance to FDA-approved proteasome inhibitors. Previous studies have demonstrated that the expression of proteasome genes in cells treated with sub-lethal concentrations of proteasome inhibitors is upregulated by the transcription factor Nrf1 (NFE2L1), which is activated by a DDI2 protease. Here, we demonstrate that the recovery of proteasome activity is DDI2-independent and occurs before transcription of proteasomal genes is upregulated but requires protein translation. Thus, mammalian cells possess an additional DDI2 and transcription-independent pathway for the rapid recovery of proteasome activity after proteasome inhibition.


Subject(s)
Proteasome Endopeptidase Complex , Proteasome Inhibitors , Animals , Endopeptidases , Mammals , Proteasome Inhibitors/pharmacology
7.
bioRxiv ; 2023 Nov 28.
Article in English | MEDLINE | ID: mdl-37577495

ABSTRACT

Rapid recovery of proteasome activity may contribute to intrinsic and acquired resistance to FDA-approved proteasome inhibitors. Previous studies have demonstrated that the expression of proteasome genes in cells treated with sub-lethal concentrations of proteasome inhibitors is upregulated by the transcription factor Nrf1 (NFE2L1), which is activated by a DDI2 protease. Here, we demonstrate that the recovery of proteasome activity is DDI2-independent and occurs before transcription of proteasomal genes is upregulated but requires protein translation. Thus, mammalian cells possess an additional DDI2 and transcription-independent pathway for the rapid recovery of proteasome activity after proteasome inhibition.

8.
Sci Rep ; 13(1): 5174, 2023 03 30.
Article in English | MEDLINE | ID: mdl-36997601

ABSTRACT

Anti-neoplastic effect of DNA cross-linking agents such as cisplatin, mitomycin C, and psoralen is attributed to their ability to induce DNA interstrand cross-links (ICLs), which block replication, transcription, and linear repair pathways by preventing DNA strand separation and trigger apoptosis. It is generally agreed that the Fanconi anemia (FA) pathway orchestrates the removal of ICLs by the combined actions of various DNA repair pathways. Recently, attention has been focused on the ability of the NEIL3-initiated base excision repair pathway to resolve psoralen- and abasic site-induced ICLs in an FA-independent manner. Intriguingly, overexpression of NEIL3 is associated with chemo-resistance and poor prognosis in many solid tumors. Here, using loss- and gain-of-function approaches, we demonstrate that NEIL3 confers resistance to cisplatin and participates in the removal of cisplatin-DNA adducts. Proteomic studies reveal that the NEIL3 protein interacts with the 26S proteasome in a cisplatin-dependent manner. NEIL3 mediates proteasomal degradation of WRNIP1, a protein involved in the early step of ICL repair. We propose that NEIL3 participates in the repair of ICL-stalled replication fork by recruitment of the proteasome to ensure a timely transition from lesion recognition to repair via the degradation of early-step vanguard proteins.


Subject(s)
Cisplatin , Proteomics , Humans , Cisplatin/pharmacology , Cross-Linking Reagents , DNA , DNA Damage , DNA Repair , DNA Replication , Ficusin/pharmacology
9.
J Biol Chem ; 286(28): 24882-95, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21628457

ABSTRACT

BH3 mimetics are small molecules designed or discovered to mimic the binding of BH3-only proteins to the hydrophobic groove of antiapoptotic BCL2 proteins. The selectivity of these molecules for BCL2, BCL-X(L), or MCL1 has been established in vitro; whether they inhibit these proteins in cells has not been rigorously investigated. In this study, we used a panel of leukemia cell lines to assess the ability of seven putative BH3 mimetics to inhibit antiapoptotic proteins in a cell-based system. We show that ABT-737 is the only BH3 mimetic that inhibits BCL2 as assessed by displacement of BAD and BIM from BCL2. The other six BH3 mimetics activate the endoplasmic reticulum stress response inducing ATF4, ATF3, and NOXA, which can then bind to and inhibit MCL1. In most cancer cells, inhibition of one antiapoptotic protein does not acutely induce apoptosis. However, by combining two BH3 mimetics, one that inhibits BCL2 and one that induces NOXA, apoptosis is induced within 6 h in a BAX/BAK-dependent manner. Because MCL1 is a major mechanism of resistance to ABT-737, these results suggest a novel strategy to overcome this resistance. Our findings highlight a novel signaling pathway through which many BH3 mimetics inhibit MCL1 and suggest the potential use of these agents as adjuvants in combination with various chemotherapy strategies.


Subject(s)
Biphenyl Compounds/pharmacology , Endoplasmic Reticulum Stress/drug effects , Nitrophenols/pharmacology , Peptide Fragments/pharmacology , Peptidomimetics/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins/pharmacology , Sulfonamides/pharmacology , Up-Regulation/drug effects , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Endoplasmic Reticulum Stress/genetics , Humans , K562 Cells , Membrane Proteins/genetics , Membrane Proteins/metabolism , Myeloid Cell Leukemia Sequence 1 Protein , Piperazines/pharmacology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Up-Regulation/genetics , bcl-Associated Death Protein/genetics , bcl-Associated Death Protein/metabolism
10.
Org Biomol Chem ; 10(1): 181-94, 2012 Jan 07.
Article in English | MEDLINE | ID: mdl-22105930

ABSTRACT

Syringolins, a class of natural products, potently and selectively inhibit the proteasome and show promising antitumour activity. To gain insight in the mode of action of syringolins, the ureido structural element present in syringolins is incorporated in oligopeptide vinyl sulfones and peptide epoxyketones yielding a focused library of potent new proteasome inhibitors. The distance of the ureido linkage with respect to the electrophilic trap strongly influences subunit selectivity within the proteasome. Compounds 13 and 15 are ß5 selective and their potency exceeds that of syringolin A. In contrast, 5 may well be the most potent ß1 selective compound active in living cells reported to date.


Subject(s)
Ketones/pharmacology , Peptides/chemistry , Proteasome Inhibitors , Sulfones/pharmacology , Urea/chemistry , Cell Line , Humans , Ketones/chemistry , Sulfones/chemistry
11.
Sci Rep ; 12(1): 12788, 2022 07 27.
Article in English | MEDLINE | ID: mdl-35896610

ABSTRACT

Proteasome inhibitors bortezomib and carfilzomib are the backbones of treatments of multiple myeloma, which remains incurable despite many recent advances. With many patients relapsing despite high initial response rates to proteasome inhibitor-containing regimens, it is critical to understand the process of acquired resistance. In vitro generated resistant cell lines are important tools in this process. The majority of previously developed bortezomib-resistant cell lines bear mutations in the proteasome PSMB5 sites, the prime target of bortezomib and carfilzomib, which are rarely observed in patients. Here we present a novel bortezomib-resistant derivative of the KMS-12-BM multiple myeloma cell line, KMS-12-BM-BPR. Unlike previously published bortezomib-resistant cell lines, it was created using clinically relevant twice-weekly pulse treatments with bortezomib instead of continuous incubation. It does not contain mutations in the PSMB5 site and retains its sensitivity to carfilzomib. Reduced load on proteasome due to decreased protein synthesis appears to be the main cause of resistance. In addition, KMS-12-BM-BPR cells are more sensitive to Bcl-2 inhibitor venetoclax. Overall, this study demonstrates the feasibility of creating a proteasome inhibitor resistant myeloma cell lines by using clinically relevant pulse treatments and provides a novel model of acquired resistance.


Subject(s)
Antineoplastic Agents , Multiple Myeloma , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Bortezomib/pharmacology , Bortezomib/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Mutation , Neoplasm Recurrence, Local , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Sulfonamides
12.
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
13.
Biomolecules ; 12(1)2021 12 31.
Article in English | MEDLINE | ID: mdl-35053202

ABSTRACT

Proteasome is a multi-subunit protein degradation machine, which plays a key role in the maintenance of protein homeostasis and, through degradation of regulatory proteins, in the regulation of numerous cell functions. Proteasome inhibitors are essential tools for biomedical research. Three proteasome inhibitors, bortezomib, carfilzomib, and ixazomib are approved by the FDA for the treatment of multiple myeloma; another inhibitor, marizomib, is undergoing clinical trials. The proteolytic core of the proteasome has three pairs of active sites, ß5, ß2, and ß1. All clinical inhibitors and inhibitors that are widely used as research tools (e.g., epoxomicin, MG-132) inhibit multiple active sites and have been extensively reviewed in the past. In the past decade, highly specific inhibitors of individual active sites and the distinct active sites of the lymphoid tissue-specific immunoproteasome have been developed. Here, we provide a comprehensive review of these site-specific inhibitors of mammalian proteasomes and describe their utilization in the studies of the biology of the active sites and their roles as drug targets for the treatment of different diseases.


Subject(s)
Antineoplastic Agents , Multiple Myeloma , Animals , Antineoplastic Agents/pharmacology , Bortezomib/pharmacology , Mammals/metabolism , Multiple Myeloma/drug therapy , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Proteasome Inhibitors/therapeutic use , Proteolysis
14.
Sci Rep ; 11(1): 10883, 2021 05 25.
Article in English | MEDLINE | ID: mdl-34035431

ABSTRACT

Proteasome inhibitors bortezomib and carfilzomib are approved for the treatment of multiple myeloma and mantle cell lymphoma and have demonstrated clinical efficacy for the treatment of acute lymphoblastic leukemia (ALL). The t(4;11)(q21;q23) chromosomal translocation that leads to the expression of MLL-AF4 fusion protein and confers a poor prognosis, is the major cause of infant ALL. This translocation sensitizes tumor cells to proteasome inhibitors, but toxicities of bortezomib and carfilzomib may limit their use in pediatric patients. Many of these toxicities are caused by on-target inhibition of proteasomes in non-lymphoid tissues (e.g., heart muscle, gut, testicles). We found that MLL-AF4 cells express high levels of lymphoid tissue-specific immunoproteasomes and are sensitive to pharmacologically relevant concentrations of specific immunoproteasome inhibitor ONX-0914, even in the presence of stromal cells. Inhibition of multiple active sites of the immunoproteasomes was required to achieve cytotoxicity against ALL. ONX-0914, an inhibitor of LMP7 (ß5i) and LMP2 (ß1i) sites of the immunoproteasome, and LU-102, inhibitor of proteasome ß2 sites, exhibited synergistic cytotoxicity. Treatment with ONX-0914 significantly delayed the growth of orthotopic ALL xenograft tumors in mice. T-cell ALL lines were also sensitive to pharmacologically relevant concentrations of ONX-0914. This study provides a strong rationale for testing clinical stage immunoproteasome inhibitors KZ-616 and M3258 in ALL.


Subject(s)
Myeloid-Lymphoid Leukemia Protein/genetics , Oligopeptides/administration & dosage , Oncogene Proteins, Fusion/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Proteasome Inhibitors/administration & dosage , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Oligopeptides/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proteasome Inhibitors/pharmacology , Random Allocation , Xenograft Model Antitumor Assays
15.
Org Biomol Chem ; 8(12): 2719-27, 2010 Jun 21.
Article in English | MEDLINE | ID: mdl-20449511

ABSTRACT

Mammals express seven different catalytically active proteasome subunits. In order to determine the roles of the different proteolytically active subunits in antigen presentation and other cellular processes, highly specific inhibitors and activity-based probes that selectively target specific active sites are needed. In this work we present the development of fluorescent activity-based probes that selectively target the beta1 and beta5 sites of the constitutive proteasome.


Subject(s)
Fluorescent Dyes/chemistry , Proteasome Endopeptidase Complex/metabolism , Protein Subunits/chemistry , Binding Sites , Cells, Cultured , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Proteasome Endopeptidase Complex/chemistry , Proteasome Inhibitors , Protein Subunits/antagonists & inhibitors , Protein Subunits/metabolism , Substrate Specificity
16.
Front Cell Dev Biol ; 8: 607045, 2020.
Article in English | MEDLINE | ID: mdl-33409281

ABSTRACT

The nucleosome is a stretch of DNA wrapped around a histone octamer. Electrostatic interactions and hydrogen bonds between histones and DNA are vital for the stable organization of nucleosome core particles, and for the folding of chromatin into more compact structures, which regulate gene expression via controlled access to DNA. As a drawback of tight association, under genotoxic stress, DNA can accidentally cross-link to histone in a covalent manner, generating a highly toxic DNA-histone cross-link (DHC). DHC is a bulky lesion that can impede DNA transcription, replication, and repair, often with lethal consequences. The chemotherapeutic agent cisplatin, as well as ionizing and ultraviolet irradiations and endogenously occurring reactive aldehydes, generate DHCs by forming either stable or transient covalent bonds between DNA and side-chain amino groups of histone lysine residues. The mechanisms of DHC repair start to unravel, and certain common principles of DNA-protein cross-link (DPC) repair mechanisms that participate in the removal of cross-linked histones from DNA have been described. In general, DPC is removed via a two-step repair mechanism. First, cross-linked proteins are degraded by specific DPC proteases or by the proteasome, relieving steric hindrance. Second, the remaining DNA-peptide cross-links are eliminated in various DNA repair pathways. Delineating the molecular mechanisms of DHC repair would help target specific DNA repair proteins for therapeutic intervention to combat tumor resistance to chemotherapy and radiotherapy.

17.
Chem Biol ; 15(5): 419-21, 2008 May.
Article in English | MEDLINE | ID: mdl-18482693

ABSTRACT

An article in this issue of Chemistry & Biology (Hines et al., 2008) and a recent study in Nature (Groll et al., 2008) establish three natural products as novel proteasome inhibitors. These inhibitors, discovered in an unusual way, reveal a different mechanism of proteasome inhibition and suggest new therapeutic application of its inhibitors.


Subject(s)
Cysteine Proteinase Inhibitors/pharmacology , Proteasome Inhibitors , Cysteine Proteinase Inhibitors/chemistry , Drug Design , X-Ray Diffraction
18.
J Med Chem ; 62(3): 1626-1642, 2019 02 14.
Article in English | MEDLINE | ID: mdl-30657666

ABSTRACT

Subunit-selective proteasome inhibitors are valuable tools to assess the biological and medicinal relevance of individual proteasome active sites. Whereas the inhibitors for the ß1c, ß1i, ß5c, and ß5i subunits exploit the differences in the substrate-binding channels identified by X-ray crystallography, compounds selectively targeting ß2c or ß2i could not yet be rationally designed because of the high structural similarity of these two subunits. Here, we report the development, chemical synthesis, and biological screening of a compound library that led to the identification of the ß2c- and ß2i-selective compounds LU-002c (4; IC50 ß2c: 8 nM, IC50 ß2i/ß2c: 40-fold) and LU-002i (5; IC50 ß2i: 220 nM, IC50 ß2c/ß2i: 45-fold), respectively. Co-crystal structures with ß2 humanized yeast proteasomes visualize protein-ligand interactions crucial for subunit specificity. Altogether, organic syntheses, activity-based protein profiling, yeast mutagenesis, and structural biology allowed us to decipher significant differences of ß2 substrate-binding channels and to complete the set of subunit-selective proteasome inhibitors.


Subject(s)
Oligopeptides/pharmacology , Proteasome Inhibitors/pharmacology , Protein Subunits/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Catalytic Domain , Cell Line, Tumor , Crystallography, X-Ray , Drug Design , Humans , Mice , Mutation , Oligopeptides/chemical synthesis , Oligopeptides/metabolism , Peptide Library , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/chemical synthesis , Proteasome Inhibitors/metabolism , Protein Binding , Protein Engineering , Protein Subunits/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/metabolism , Stereoisomerism
19.
Blood Adv ; 2(19): 2443-2451, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30266819

ABSTRACT

Proteasome inhibitors bortezomib, carfilzomib and ixazomib (approved by the US Food and Drug Administration [FDA]) induce remissions in patients with multiple myeloma (MM), but most patients eventually become resistant. MM and other hematologic malignancies express ubiquitous constitutive proteasomes and lymphoid tissue-specific immunoproteasomes; immunoproteasome expression is increased in resistant patients. Immunoproteasomes contain 3 distinct pairs of active sites, ß5i, ß1i, and ß2i, which are different from their constitutive ß5c, ß1c, and ß2c counterparts. Bortezomib and carfilzomib block ß5c and ß5i sites. We report here that pharmacologically relevant concentrations of ß5i-specific inhibitor ONX-0914 show cytotoxicity in MM cell lines similar to that of carfilzomib and bortezomib. In addition, increasing immunoproteasome expression by interferon-γ increases sensitivity to ONX-0914 but not to carfilzomib. LU-102, an inhibitor of ß2 sites, dramatically sensitizes MM cell lines and primary cells to ONX-0914. ONX-0914 synergizes with all FDA-approved proteasome inhibitors in MM in vitro and in vivo. Thus, immunoproteasome inhibitors, currently in clinical trials for the treatment of autoimmune diseases, should also be considered for the treatment of MM.


Subject(s)
Antineoplastic Agents/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Animals , Biomarkers , Bortezomib/pharmacology , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Humans , Male , Mice , Multiple Myeloma/metabolism , Oligopeptides/pharmacology
20.
Cell Chem Biol ; 24(2): 218-230, 2017 Feb 16.
Article in English | MEDLINE | ID: mdl-28132893

ABSTRACT

The proteasome inhibitors carfilzomib (Cfz) and bortezomib (Btz) are used successfully to treat multiple myeloma, but have not shown clinical efficacy in solid tumors. Here we show that clinically achievable inhibition of the ß5 site of the proteasome by Cfz and Btz does not result in loss of viability of triple-negative breast cancer cell lines. We use site-specific inhibitors and CRISPR-mediated genetic inactivation of ß1 and ß2 to demonstrate that inhibiting a second site of the proteasome, particularly the ß2 site, sensitizes cell lines to Btz and Cfz in vitro and in vivo. Inhibiting both ß5 and ß2 suppresses production of the soluble, active form of the transcription factor Nrf1 and prevents the recovery of proteasome activity through induction of new proteasomes. These findings provide a strong rationale for the development of dual ß5 and ß2 inhibitors for the treatment of solid tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Nuclear Respiratory Factor 1/antagonists & inhibitors , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Nuclear Respiratory Factor 1/metabolism , Proteasome Inhibitors/chemical synthesis , Proteasome Inhibitors/chemistry , Structure-Activity Relationship , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL