Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Commun Biol ; 7(1): 497, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38658677

ABSTRACT

Most lung cancer patients with metastatic cancer eventually relapse with drug-resistant disease following treatment and EGFR mutant lung cancer is no exception. Genome-wide CRISPR screens, to either knock out or overexpress all protein-coding genes in cancer cell lines, revealed the landscape of pathways that cause resistance to the EGFR inhibitors osimertinib or gefitinib in EGFR mutant lung cancer. Among the most recurrent resistance genes were those that regulate the Hippo pathway. Following osimertinib treatment a subpopulation of cancer cells are able to survive and over time develop stable resistance. These 'persister' cells can exploit non-genetic (transcriptional) programs that enable cancer cells to survive drug treatment. Using genetic and pharmacologic tools we identified Hippo signalling as an important non-genetic mechanism of cell survival following osimertinib treatment. Further, we show that combinatorial targeting of the Hippo pathway and EGFR is highly effective in EGFR mutant lung cancer cells and patient-derived organoids, suggesting a new therapeutic strategy for EGFR mutant lung cancer patients.


Subject(s)
Acrylamides , Drug Resistance, Neoplasm , ErbB Receptors , Indoles , Lung Neoplasms , Mutation , Pyrimidines , Transcription Factors , Humans , Lung Neoplasms/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Drug Resistance, Neoplasm/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Cell Line, Tumor , Acrylamides/pharmacology , Acrylamides/therapeutic use , YAP-Signaling Proteins/metabolism , YAP-Signaling Proteins/genetics , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Gefitinib/pharmacology , Hippo Signaling Pathway , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Signal Transduction , TEA Domain Transcription Factors , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Clustered Regularly Interspaced Short Palindromic Repeats , CRISPR-Cas Systems
2.
Trends Pharmacol Sci ; 42(8): 688-699, 2021 08.
Article in English | MEDLINE | ID: mdl-34074527

ABSTRACT

KMT2 methyltransferases are important regulators of gene transcription through the methylation of histone H3 lysine 4 at promoter and enhancer regions. They reside in large, multisubunit protein complexes, which not only regulate their catalytic activities but also mediate their interactions with chromatin. The KMT2 family was initially associated with cancer due to the discovery of KMT2A translocations in mixed-lineage leukemia (MLL). However, emerging evidences suggest that the methyltransferase activity of KMT2 enzymes can also be important in cancer, raising the prospect of targeting the catalytic domain of KMT2 as a therapeutic strategy. In this review, we summarize recent advances in our understanding of KMT2 enzyme mechanisms and their regulation on nucleosomes, which will provide mechanistic insights into therapeutic discoveries targeting their methyltransferase activities.


Subject(s)
Myeloid-Lymphoid Leukemia Protein , Neoplasms , DNA-Binding Proteins/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Methylation , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Neoplasms/drug therapy , Neoplasms/genetics
3.
Biochim Biophys Acta Gene Regul Mech ; 1864(2): 194605, 2021 02.
Article in English | MEDLINE | ID: mdl-32711094

ABSTRACT

Eukaryotic genomes are maintained within DNA-protein complexes called chromatin. Post-translational modification of chromatin proteins, and especially acetylation of the core histone amino-terminal tails, has long been associated with chromatin assembly and the regulation of gene expression. It is now well accepted that an elaborate array of enzymes are responsible for posttranslational chromatin marks including acetylation and methylation among others and that together they have profound effects on gene regulation. However, this was not always the case. Here we describe the events surrounding the initial identification of GCN5 as a histone acetyltransferase from Tetrahymena thermophila and the discovery that it is an ortholog of a transcription co-activator complex in yeast. This discovery was the first to directly link a well-described transcription factor and histone modifying activity.


Subject(s)
Chromatin/metabolism , Epigenesis, Genetic/physiology , Histone Acetyltransferases/metabolism , Histones/metabolism , Protein Processing, Post-Translational/physiology , Acetylation , Enzyme Assays , Histone Acetyltransferases/genetics , Protozoan Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Homology, Amino Acid , Tetrahymena thermophila/enzymology , Tetrahymena thermophila/genetics , Transcription, Genetic/physiology
4.
Bioorg Med Chem ; 28(19): 115681, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32912429

ABSTRACT

Autophagy is postulated to be required by cancer cells to survive periods of metabolic and/or hypoxic stress. ATG7 is the E1 enzyme that is required for activation of Ubl conjugation pathways involved in autophagosome formation. This article describes the design and optimization of pyrazolopyrimidine sulfamate compounds as potent and selective inhibitors of ATG7. Cellular levels of the autophagy markers, LC3B and NBR1, are regulated following treatment with these compounds.


Subject(s)
Autophagy-Related Protein 7/antagonists & inhibitors , Drug Discovery , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Sulfonic Acids/pharmacology , Autophagy/drug effects , Autophagy-Related Protein 7/metabolism , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Molecular Structure , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Structure-Activity Relationship , Sulfonic Acids/chemical synthesis , Sulfonic Acids/chemistry
5.
Nat Chem Biol ; 13(11): 1164-1171, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28892090

ABSTRACT

Small ubiquitin-like modifier (SUMO) family proteins regulate target-protein functions by post-translational modification. However, a potent and selective inhibitor targeting the SUMO pathway has been lacking. Here we describe ML-792, a mechanism-based SUMO-activating enzyme (SAE) inhibitor with nanomolar potency in cellular assays. ML-792 selectively blocks SAE enzyme activity and total SUMOylation, thus decreasing cancer cell proliferation. Moreover, we found that induction of the MYC oncogene increased the ML-792-mediated viability effect in cancer cells, thus indicating a potential application of SAE inhibitors in treating MYC-amplified tumors. Using ML-792, we further explored the critical roles of SUMOylation in mitotic progression and chromosome segregation. Furthermore, expression of an SAE catalytic-subunit (UBA2) S95N M97T mutant rescued SUMOylation loss and the mitotic defect induced by ML-792, thus confirming the selectivity of ML-792. As a potent and selective SAE inhibitor, ML-792 provides rapid loss of endogenously SUMOylated proteins, thereby facilitating novel insights into SUMO biology.


Subject(s)
Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Neoplasms/metabolism , Small Ubiquitin-Related Modifier Proteins/antagonists & inhibitors , Sumoylation , Cell Proliferation/drug effects , Chromosome Segregation/drug effects , DNA Damage/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genes, myc , Humans , Mitosis/drug effects , Neoplasms/genetics , Neoplasms/pathology , Protein Processing, Post-Translational , Tumor Cells, Cultured
6.
J Biomark ; 2016: 1274603, 2016.
Article in English | MEDLINE | ID: mdl-27247826

ABSTRACT

Objective. Use of tyramide signal amplification (TSA) to detect autophagy biomarkers in formalin fixed and paraffin embedded (FFPE) xenograft tissue. Materials and Methods. Autophagy marker regulation was studied in xenograft tissues using Amp HQ IHC and standard IHC methods. Results. The data demonstrate the feasibility of using high sensitivity TSA IHC assays to measure low abundant autophagy markers in FFPE xenograft tissue.

7.
Autophagy ; : 0, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-26090719

ABSTRACT

Autophagy is a major cellular process for bulk degradation of proteins and organelles in order to maintain metabolic homeostasis, and it represents an emerging target area for cancer. Initially proposed to be a cancer-restricting process for tumor initiation, recent studies suggest that autophagy can also promote cell survival in established tumors. ATG7 is an essential autophagy gene that encodes the E1 enzyme necessary for the lipidation of the LC3 family of ubiquitin-like proteins and autophagosome formation. In this study we identified a rare case of a cancer cell line, H1650 lung adenocarcinoma, which has lost ATG7 expression due to a focal biallelic deletion within the ATG7 locus. These cells displayed no evidence of ATG7 pathway activity; however, reconstituting the cells with wild-type ATG7 restored both LC3 lipidation and downstream autophagic consumption of autophagy substrates such as the SQSTM1/p62 protein. We characterized several phenotypes reported to be influenced by autophagy, and observed an ATG7-dependent increase in cell growth and clearance of proteasome-inhibitor induced protein aggregates. Cellular changes in mitochondrial metabolism or response to nutrient starvation were unaffected by ATG7 expression. In addition, parental H1650 cells that lacked ATG7 were still able to consume autophagy substrates SQSTM1, NBR1 and TAX1BP1 via a bafilomycin A1-sensitive pathway, suggesting that these proteins were not exclusively degraded by autophagy. Overall, these findings highlight a unique outlier instance of complete loss of ATG7-dependent autophagy in a cancer cell line. The H1650 cell line may be a useful system for future studies to further understand the role of autophagy in tumorigenesis and potential redundant pathways that allow cells to circumvent the loss of ATG7-dependent autophagy in cancer.

8.
PLoS One ; 10(4): e0123882, 2015.
Article in English | MEDLINE | ID: mdl-25860128

ABSTRACT

SUMOylation is a post-translational ubiquitin-like protein modification pathway that regulates important cellular processes including chromosome structure, kinetochore function, chromosome segregation, nuclear and sub-nuclear organization, transcription and DNA damage repair. There is increasing evidence that the SUMO pathway is dysregulated in cancer, raising the possibility that modulation of this pathway may have therapeutic potential. To investigate the importance of the SUMO pathway in the context of cancer cell proliferation and tumor growth, we applied lentivirus-based short hairpin RNAs (shRNA) to knockdown SUMO pathway genes in human cancer cells. shRNAs for SAE2 and UBC9 reduced SUMO conjugation activity and inhibited proliferation of human cancer cells. To expand upon these observations, we generated doxycycline inducible conditional shRNA cell lines for SAE2 to achieve acute and reversible SAE2 knockdown. Conditional SAE2 knockdown in U2OS and HCT116 cells slowed cell growth in vitro, and SAE2 knockdown induced multiple terminal outcomes including apoptosis, endoreduplication and senescence. Multinucleated cells became senescent and stained positive for the senescence marker, SA-ß Gal, and displayed elevated levels of p53 and p21. In an attempt to explain these phenotypes, we confirmed that loss of SUMO pathway activity leads to a loss of SUMOylated Topoisomerase IIα and the appearance of chromatin bridges which can impair proper cytokinesis and lead to multinucleation. Furthermore, knockdown of SAE2 induces disruption of PML nuclear bodies which may further promote apoptosis or senescence. In an in vivo HCT116 xenograft tumor model, conditional SAE2 knockdown strongly impaired tumor growth. These data demonstrate that the SUMO pathway is required for cancer cell proliferation in vitro and tumor growth in vivo, implicating the SUMO pathway as a potential cancer therapeutic target.


Subject(s)
Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction , Small Ubiquitin-Related Modifier Proteins/metabolism , Animals , Apoptosis/genetics , Cell Cycle/genetics , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Proliferation , Cellular Senescence/genetics , Disease Models, Animal , Gene Knockdown Techniques , Heterografts , Humans , Neoplasms/genetics , Phenotype , RNA Interference , Small Ubiquitin-Related Modifier Proteins/genetics , Sumoylation , Tumor Burden/genetics
9.
Cell Biochem Biophys ; 67(1): 139-47, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23754621

ABSTRACT

Ubiquitin (Ub) and ubiquitin-like (Ubl) proteins regulate a variety of important cellular processes by forming covalent conjugates with target proteins or lipids. Ubl conjugation is catalyzed by a cascade of proteins including activating enzymes (E1), conjugating enzymes (E2), and in many cases ligation enzymes (E3). The discovery of MLN4924 (Brownell et al., Mol Cell 37: 102-111, 1), an investigational small molecule that is a mechanism-based inhibitor of NEDD8-activating enzyme (NAE), reveals a promising strategy of targeting E1/Ubl pathway for therapeutic purposes. In order to better understand, the biochemical dynamics of Ubl conjugation in cells and tissues, we have developed a mass spectrometry-based method to quantify E1 and Ubls using isotope-labeled proteins as internal standards. Furthermore, we have used the described method to quantify levels of the covalent Nedd8-inhibitor adduct formed in MLN4924 treated cells and tissues. The Nedd8-MLN4924 adduct is a tight-binding inhibitor of NAE, and its cellular concentration represents an indirect pharmacodynamic readout of NAE/Nedd8 pathway inhibition.


Subject(s)
Cyclopentanes/chemistry , Pyrimidines/chemistry , Ubiquitin-Activating Enzymes/chemistry , Ubiquitins/chemistry , Animals , Cell Line , Chromatography, High Pressure Liquid , Female , HCT116 Cells , HeLa Cells , Humans , Isotope Labeling , NEDD8 Protein , Nanotechnology , Peptides/analysis , Rats , Rats, Nude , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Tandem Mass Spectrometry , Ubiquitin/chemistry , Ubiquitin/metabolism , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism , Ubiquitins/metabolism
10.
Cancer Cell ; 21(3): 388-401, 2012 Mar 20.
Article in English | MEDLINE | ID: mdl-22439935

ABSTRACT

MLN4924 is an investigational small-molecule inhibitor of NEDD8-activating enzyme (NAE) in clinical trials for the treatment of cancer. MLN4924 is a mechanism-based inhibitor, with enzyme inhibition occurring through the formation of a tight-binding NEDD8-MLN4924 adduct. In cell and xenograft models of cancer, we identified treatment-emergent heterozygous mutations in the adenosine triphosphate binding pocket and NEDD8-binding cleft of NAEß as the primary mechanism of resistance to MLN4924. Biochemical analyses of NAEß mutants revealed slower rates of adduct formation and reduced adduct affinity for the mutant enzymes. A compound with tighter binding properties was able to potently inhibit mutant enzymes in cells. These data provide rationales for patient selection and the development of next-generation NAE inhibitors designed to overcome treatment-emergent NAEß mutations.


Subject(s)
Cyclopentanes/pharmacology , Enzyme Inhibitors/pharmacology , Mutation , Pyrimidines/pharmacology , Ubiquitin-Activating Enzymes/genetics , Animals , Binding Sites , Cell Line, Tumor , Clinical Trials as Topic , Drug Resistance, Neoplasm/genetics , Female , Humans , Mice , Mice, Nude , Rats , Rats, Nude , Tumor Cells, Cultured , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Activating Enzymes/physiology , Xenograft Model Antitumor Assays
11.
J Biol Chem ; 287(19): 15512-22, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22427669

ABSTRACT

Uba6 is a homolog of the ubiquitin-activating enzyme, Uba1, and activates two ubiquitin-like proteins (UBLs), ubiquitin and FAT10. In this study, biochemical and biophysical experiments were performed to understand the mechanisms of how Uba6 recognizes two distinct UBLs and catalyzes their activation and transfer. Uba6 is shown to undergo a three-step activation process and form a ternary complex with both UBLs, similar to what has been observed for Uba1. The catalytic mechanism of Uba6 is further supported by inhibition studies using a mechanism-based E1 inhibitor, Compound 1, which forms covalent adducts with both ubiquitin and FAT10. In addition, pre-steady state kinetic analysis revealed that the rates of UBL-adenylate (step 1) and thioester (step 2) formation are similar between ubiquitin and FAT10. However, distinct kinetic behaviors were also observed for ubiquitin and FAT10. FAT10 binds Uba6 with much higher affinity than ubiquitin while demonstrating lower catalytic activity in both ATP-PP(i) exchange and E1-E2 transthiolation assays. Also, Compound 1 is less potent with FAT10 as the UBL compared with ubiquitin in ATP-PP(i) exchange assays, and both a slow rate of covalent adduct formation and weak adduct binding to Uba6 contribute to the diminished potency observed for FAT10. Together with expression level analysis in IM-9 cells, this study sheds light on the potential role of cytokine-induced FAT10 expression in regulating Uba6 pathways.


Subject(s)
Adenosine Triphosphate/metabolism , Diphosphates/metabolism , Ubiquitin-Activating Enzymes/metabolism , Ubiquitins/metabolism , Adenosine Monophosphate/chemistry , Adenosine Monophosphate/metabolism , Adenosine Monophosphate/pharmacology , Amino Acid Sequence , Animals , Blotting, Western , Cell Line , Enzyme Activation/drug effects , Humans , Interferon-gamma/pharmacology , Kinetics , Mass Spectrometry , Molecular Sequence Data , Molecular Structure , Protein Binding/drug effects , Spodoptera , Substrate Specificity , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/metabolism , Surface Plasmon Resonance , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitin-Activating Enzymes/chemistry , Ubiquitin-Activating Enzymes/genetics , Ubiquitins/chemistry , Ubiquitins/genetics
12.
Methods Mol Biol ; 832: 577-88, 2012.
Article in English | MEDLINE | ID: mdl-22350913

ABSTRACT

The NEDD8 conjugation pathway is initiated by the NEDD8 E1, also known as NEDD8 activating enzyme (NAE) or APPBP1/UBA3 (Gong, Yeh. J Biol Chem 274:12063-12042, 1999). The best described biological role for NEDD8 conjugation is to regulate the activity of the cullin RING ligase (CRL) family of ubiquitin E3 ligases (Gong, Yeh. J Biol Chem 274:12063-12042, 1999). In this way, the NEDD8 pathway regulates the turnover of a subset of ubiquitin proteasome system (UPS) substrates that are essential for cancer cell growth and survival (Soucy, Smith, Milhollen. Nature 458:732-737, 2009). We recently initiated clinical trials with a first-in-class small molecule inhibitor of NAE for the treatment of cancer (Soucy, Smith, Milhollen. Nature 458:732-737, 2009). Here we describe a biochemical and cell-based assay used to identify NAE inhibitors and monitor inhibition of the NEDD8 conjugation pathway.


Subject(s)
Ubiquitin-Activating Enzymes/antagonists & inhibitors , Ubiquitins/metabolism , Cell Line, Tumor , Cullin Proteins/metabolism , Enzyme Activation , Humans , NEDD8 Protein , Ubiquitins/analysis
13.
J Biol Chem ; 286(47): 40867-77, 2011 Nov 25.
Article in English | MEDLINE | ID: mdl-21969368

ABSTRACT

Ubiquitin-activating enzyme (UAE or E1) activates ubiquitin via an adenylate intermediate and catalyzes its transfer to a ubiquitin-conjugating enzyme (E2). MLN4924 is an adenosine sulfamate analogue that was identified as a selective, mechanism-based inhibitor of NEDD8-activating enzyme (NAE), another E1 enzyme, by forming a NEDD8-MLN4924 adduct that tightly binds at the active site of NAE, a novel mechanism termed substrate-assisted inhibition (Brownell, J. E., Sintchak, M. D., Gavin, J. M., Liao, H., Bruzzese, F. J., Bump, N. J., Soucy, T. A., Milhollen, M. A., Yang, X., Burkhardt, A. L., Ma, J., Loke, H. K., Lingaraj, T., Wu, D., Hamman, K. B., Spelman, J. J., Cullis, C. A., Langston, S. P., Vyskocil, S., Sells, T. B., Mallender, W. D., Visiers, I., Li, P., Claiborne, C. F., Rolfe, M., Bolen, J. B., and Dick, L. R. (2010) Mol. Cell 37, 102-111). In the present study, substrate-assisted inhibition of human UAE (Ube1) by another adenosine sulfamate analogue, 5'-O-sulfamoyl-N(6)-[(1S)-2,3-dihydro-1H-inden-1-yl]-adenosine (Compound I), a nonselective E1 inhibitor, was characterized. Compound I inhibited UAE-dependent ATP-PP(i) exchange activity, caused loss of UAE thioester, and inhibited E1-E2 transthiolation in a dose-dependent manner. Mechanistic studies on Compound I and its purified ubiquitin adduct demonstrate that the proposed substrate-assisted inhibition via covalent adduct formation is entirely consistent with the three-step ubiquitin activation process and that the adduct is formed via nucleophilic attack of UAE thioester by the sulfamate group of Compound I after completion of step 2. Kinetic and affinity analysis of Compound I, MLN4924, and their purified ubiquitin adducts suggest that both the rate of adduct formation and the affinity between the adduct and E1 contribute to the overall potency. Because all E1s are thought to use a similar mechanism to activate their cognate ubiquitin-like proteins, the substrate-assisted inhibition by adenosine sulfamate analogues represents a promising strategy to develop potent and selective E1 inhibitors that can modulate diverse biological pathways.


Subject(s)
Enzyme Inhibitors/pharmacology , Sulfonic Acids/pharmacology , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Animals , Cattle , Diphosphates/metabolism , Humans , Hydrolysis/drug effects , Kinetics , Sulfhydryl Compounds/metabolism , Ubiquitin-Activating Enzymes/metabolism
14.
Nat Rev Drug Discov ; 10(1): 29-46, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21151032

ABSTRACT

The ubiquitin-proteasome system (UPS) and ubiquitin-like protein (UBL) conjugation pathways are integral to cellular protein homeostasis. The growing recognition of the fundamental importance of these pathways to normal cell function and in disease has prompted an in-depth search for small-molecule inhibitors that selectively block the function of these pathways. However, our limited understanding of the molecular mechanisms and biological consequences of UBL conjugation is a significant hurdle to identifying drug-like inhibitors of enzyme targets within these pathways. Here, we highlight recent advances in understanding the role of some of these enzymes and how these new insights may be the key to developing novel therapeutics for diseases including immuno-inflammatory disorders, cancer, infectious diseases, cardiovascular disease and neurodegenerative disorders.


Subject(s)
Drug Delivery Systems , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Ubiquitins/metabolism , Animals , Drug Design , Enzyme Inhibitors/pharmacology , Humans
15.
Mol Cell ; 37(1): 102-11, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20129059

ABSTRACT

The NEDD8-activating enzyme (NAE) initiates a protein homeostatic pathway essential for cancer cell growth and survival. MLN4924 is a selective inhibitor of NAE currently in clinical trials for the treatment of cancer. Here, we show that MLN4924 is a mechanism-based inhibitor of NAE and creates a covalent NEDD8-MLN4924 adduct catalyzed by the enzyme. The NEDD8-MLN4924 adduct resembles NEDD8 adenylate, the first intermediate in the NAE reaction cycle, but cannot be further utilized in subsequent intraenzyme reactions. The stability of the NEDD8-MLN4924 adduct within the NAE active site blocks enzyme activity, thereby accounting for the potent inhibition of the NEDD8 pathway by MLN4924. Importantly, we have determined that compounds resembling MLN4924 demonstrate the ability to form analogous adducts with other ubiquitin-like proteins (UBLs) catalyzed by their cognate-activating enzymes. These findings reveal insights into the mechanism of E1s and suggest a general strategy for selective inhibition of UBL conjugation pathways.


Subject(s)
Adenosine Monophosphate/metabolism , Cyclopentanes/metabolism , Enzyme Inhibitors/metabolism , Pyrimidines/metabolism , Ubiquitins/metabolism , Adenosine Monophosphate/chemistry , Binding Sites , Binding, Competitive , Cell Line, Tumor , Crystallography, X-Ray , Cyclopentanes/chemistry , Cyclopentanes/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , NEDD8 Protein , Protein Structure, Tertiary , Pyrimidines/chemistry , Pyrimidines/pharmacology , Ubiquitins/chemistry
16.
Genes Cancer ; 1(7): 708-16, 2010 Jul.
Article in English | MEDLINE | ID: mdl-21779466

ABSTRACT

Cancer cells depend on signals that promote cell cycle progression and prevent programmed cell death that would otherwise result from cumulative, aberrant stress. These activities require the temporally controlled destruction of specific intracellular proteins by the ubiquitin-proteasome system (UPS). To a large extent, the control points in this process include a family of E3 ubiquitin ligases called cullin-RING ligases (CRLs). The ligase activity of these multicomponent complexes requires modification of the cullin protein situated at their core with a ubiquitin-like protein called NEDD8. Neddylation results in conformational rearrangements within the CRL, which are necessary for ubiquitin transfer to a substrate. The NEDD8 pathway thus has a critical role in mediating the ubiquitination of numerous CRL substrate proteins involved in cell cycle progression and survival including the DNA replication licensing factor Cdt-1, the NF-κB transcription factor inhibitor pIκBα, and the cell cycle regulators cyclin E and p27. The initial step required for attachment of NEDD8 to a cullin is catalyzed by the E1, NEDD8-activating enzyme (NAE). The first-in-class inhibitor of NAE, MLN4924, has been shown to block the activity of NAE and prevent the subsequent neddylation of cullins. Preclinical studies have demonstrated antitumor activity in various solid tumors and hematological malignancies, and preliminary clinical data have shown the anticipated pharmacodynamic effects in humans. Here, we review the NEDD8 pathway, its importance in cancer, and the therapeutic potential of NAE inhibition.

17.
Nature ; 458(7239): 732-6, 2009 Apr 09.
Article in English | MEDLINE | ID: mdl-19360080

ABSTRACT

The clinical development of an inhibitor of cellular proteasome function suggests that compounds targeting other components of the ubiquitin-proteasome system might prove useful for the treatment of human malignancies. NEDD8-activating enzyme (NAE) is an essential component of the NEDD8 conjugation pathway that controls the activity of the cullin-RING subtype of ubiquitin ligases, thereby regulating the turnover of a subset of proteins upstream of the proteasome. Substrates of cullin-RING ligases have important roles in cellular processes associated with cancer cell growth and survival pathways. Here we describe MLN4924, a potent and selective inhibitor of NAE. MLN4924 disrupts cullin-RING ligase-mediated protein turnover leading to apoptotic death in human tumour cells by a new mechanism of action, the deregulation of S-phase DNA synthesis. MLN4924 suppressed the growth of human tumour xenografts in mice at compound exposures that were well tolerated. Our data suggest that NAE inhibitors may hold promise for the treatment of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclopentanes/pharmacology , Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Pyrimidines/pharmacology , Ubiquitin-Activating Enzymes/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Cullin Proteins/metabolism , Female , Humans , Mice , NEDD8 Protein , Proteasome Inhibitors , Transplantation, Heterologous , Ubiquitins/metabolism
18.
Phys Rev Lett ; 96(20): 200402, 2006 May 26.
Article in English | MEDLINE | ID: mdl-16803156

ABSTRACT

We propose an experiment for generating and detecting vacuum-induced dissipative motion. A high frequency mechanical resonator driven in resonance is expected to dissipate mechanical energy in quantum vacuum via photon emission. The photons are stored in a high quality electromagnetic cavity and detected through their interaction with ultracold alkali-metal atoms prepared in an inverted population of hyperfine states. Superradiant amplification of the generated photons results in a detectable radio-frequency signal temporally distinguishable from the expected background.

19.
J Biol Chem ; 279(35): 36440-4, 2004 Aug 27.
Article in English | MEDLINE | ID: mdl-15247280

ABSTRACT

The human topoisomerase I- and p53-binding protein topors contains a highly conserved, N-terminal C3HC4-type RING domain that is homologous to the RING domains of known E3 ubiquitin ligases. We demonstrate that topors functions in vitro as a RING-dependent E3 ubiquitin ligase with the E2 enzymes UbcH5a, UbcH5c, and UbcH6 but not with UbcH7, CDC34, or UbcH2b. Additional studies indicate that a conserved tryptophan within the topors RING domain is required for ubiquitination activity. Furthermore, both in vitro and cellular studies implicate p53 as a ubiquitination substrate for topors. Similar to MDM2, overexpression of topors results in a proteasome-dependent decrease in p53 protein expression in a human osteosarcoma cell line. These results are similar to the recent finding that a Drosophila topors orthologue ubiquitinates the Hairy transcriptional repressor and suggest that topors functions as a ubiquitin ligase for multiple transcription factors.


Subject(s)
Carrier Proteins/physiology , DNA-Binding Proteins/physiology , Neoplasm Proteins , Nuclear Proteins/physiology , Transcription Factors/physiology , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/physiology , Ubiquitin/metabolism , Amino Acid Sequence , Anaphase-Promoting Complex-Cyclosome , Animals , Carrier Proteins/metabolism , Cell Line, Tumor , Cysteine Endopeptidases/metabolism , DNA-Binding Proteins/metabolism , Drosophila , Electrophoresis, Polyacrylamide Gel , Glutathione Transferase/metabolism , Green Fluorescent Proteins , Humans , Immediate-Early Proteins/metabolism , Immunoblotting , Iron-Binding Proteins/chemistry , Luminescent Proteins/metabolism , Mass Spectrometry , Molecular Sequence Data , Multienzyme Complexes/metabolism , Nuclear Proteins/metabolism , Plasmids/metabolism , Proteasome Endopeptidase Complex , Protein Structure, Tertiary , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-mdm2 , Recombinant Proteins/chemistry , Sequence Homology, Amino Acid , Silver Staining , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transcription Factors/metabolism , Transcription, Genetic , Transfection , Tryptophan/chemistry , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Protein Ligase Complexes/biosynthesis , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...