Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
1.
Cell ; 178(4): 949-963.e18, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31353221

ABSTRACT

Estrogen receptor-positive (ER+) breast cancers frequently remain dependent on ER signaling even after acquiring resistance to endocrine agents, prompting the development of optimized ER antagonists. Fulvestrant is unique among approved ER therapeutics due to its capacity for full ER antagonism, thought to be achieved through ER degradation. The clinical potential of fulvestrant is limited by poor physicochemical features, spurring attempts to generate ER degraders with improved drug-like properties. We show that optimization of ER degradation does not guarantee full ER antagonism in breast cancer cells; ER "degraders" exhibit a spectrum of transcriptional activities and anti-proliferative potential. Mechanistically, we find that fulvestrant-like antagonists suppress ER transcriptional activity not by ER elimination, but by markedly slowing the intra-nuclear mobility of ER. Increased ER turnover occurs as a consequence of ER immobilization. These findings provide proof-of-concept that small molecule perturbation of transcription factor mobility may enable therapeutic targeting of this challenging target class.


Subject(s)
Breast Neoplasms/metabolism , Estrogen Receptor Antagonists/pharmacology , Fulvestrant/pharmacology , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cinnamates/pharmacology , Drug Resistance, Neoplasm , Estrogen Receptor Antagonists/therapeutic use , Female , Fulvestrant/therapeutic use , HEK293 Cells , Heterografts , Humans , Indazoles/pharmacology , Ligands , MCF-7 Cells , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Polymorphism, Single Nucleotide , Proteolysis/drug effects , Signal Transduction/drug effects , Transcription, Genetic/drug effects
2.
Mol Cell ; 77(5): 1092-1106.e9, 2020 03 05.
Article in English | MEDLINE | ID: mdl-31973889

ABSTRACT

Co-opting Cullin4 RING ubiquitin ligases (CRL4s) to inducibly degrade pathogenic proteins is emerging as a promising therapeutic strategy. Despite intense efforts to rationally design degrader molecules that co-opt CRL4s, much about the organization and regulation of these ligases remains elusive. Here, we establish protein interaction kinetics and estimation of stoichiometries (PIKES) analysis, a systematic proteomic profiling platform that integrates cellular engineering, affinity purification, chemical stabilization, and quantitative mass spectrometry to investigate the dynamics of interchangeable multiprotein complexes. Using PIKES, we show that ligase assemblies of Cullin4 with individual substrate receptors differ in abundance by up to 200-fold and that Cand1/2 act as substrate receptor exchange factors. Furthermore, degrader molecules can induce the assembly of their cognate CRL4, and higher expression of the associated substrate receptor enhances degrader potency. Beyond the CRL4 network, we show how PIKES can reveal systems level biochemistry for cellular protein networks important to drug development.


Subject(s)
Chromatography, High Pressure Liquid , Proteomics/methods , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry , Ubiquitin-Protein Ligases/metabolism , Cullin Proteins/genetics , Cullin Proteins/metabolism , HEK293 Cells , Humans , Kinetics , Muscle Proteins/genetics , Muscle Proteins/metabolism , NEDD8 Protein/genetics , NEDD8 Protein/metabolism , Protein Interaction Maps , Proteolysis , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics
3.
Nat Chem Biol ; 19(1): 55-63, 2023 01.
Article in English | MEDLINE | ID: mdl-36577875

ABSTRACT

Engineered destruction of target proteins by recruitment to the cell's degradation machinery has emerged as a promising strategy in drug discovery. The majority of molecules that facilitate targeted degradation do so via a select number of ubiquitin ligases, restricting this therapeutic approach to tissue types that express the requisite ligase. Here, we describe a new strategy of targeted protein degradation through direct substrate recruitment to the 26S proteasome. The proteolytic complex is essential and abundantly expressed in all cells; however, proteasomal ligands remain scarce. We identify potent peptidic macrocycles that bind directly to the 26S proteasome subunit PSMD2, with a 2.5-Å-resolution cryo-electron microscopy complex structure revealing a binding site near the 26S pore. Conjugation of this macrocycle to a potent BRD4 ligand enabled generation of chimeric molecules that effectively degrade BRD4 in cells, thus demonstrating that degradation via direct proteasomal recruitment is a viable strategy for targeted protein degradation.


Subject(s)
Nuclear Proteins , Transcription Factors , Nuclear Proteins/metabolism , Cryoelectron Microscopy , Transcription Factors/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Ligases/metabolism , Ubiquitin-Protein Ligases/metabolism
4.
J Am Chem Soc ; 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38848252

ABSTRACT

ß-catenin (CTNNB1) is an oncogenic transcription factor that is important in cell-cell adhesion and transcription of cell proliferation and survival genes that drive the pathogenesis of many different types of cancers. However, direct pharmacological targeting of CTNNB1 has remained challenging. Here, we have performed a screen with a library of cysteine-reactive covalent ligands to identify the monovalent degrader EN83 that depletes CTNNB1 in a ubiquitin-proteasome-dependent manner. We show that EN83 directly and covalently targets CTNNB1 three cysteines C466, C520, and C619, leading to destabilization and degradation of CTNNB1. Through structural optimization, we generate a highly potent and relatively selective destabilizing degrader that acts through the targeting of only C619 on CTNNB1. Our results show that chemoproteomic approaches can be used to covalently target and degrade challenging transcription factors like CTNNB1 through destabilization-mediated degradation.

5.
Mol Cell ; 63(1): 4-6, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27392144

ABSTRACT

Abdul Rehman and colleagues identify a sixth family of deubiquitinase enzymes that are highly conserved throughout eukaryotes and show a remarkable selectivity for cleaving extended Lys-48-linked polyubiquitin chains.


Subject(s)
Eukaryota , Polyubiquitin , Deubiquitinating Enzymes , Humans
6.
Am J Hematol ; 98(3): 449-463, 2023 03.
Article in English | MEDLINE | ID: mdl-36594167

ABSTRACT

The treatment of patients with relapsed or refractory lymphoid neoplasms represents a significant clinical challenge. Here, we identify the pro-survival BCL-2 protein family member MCL-1 as a resistance factor for the BCL-2 inhibitor venetoclax in non-Hodgkin lymphoma (NHL) cell lines and primary NHL samples. Mechanistically, we show that the antibody-drug conjugate polatuzumab vedotin promotes MCL-1 degradation via the ubiquitin/proteasome system. This targeted MCL-1 antagonism, when combined with venetoclax and the anti-CD20 antibodies obinutuzumab or rituximab, results in tumor regressions in preclinical NHL models, which are sustained even off-treatment. In a Phase Ib clinical trial (NCT02611323) of heavily pre-treated patients with relapsed or refractory NHL, 25/33 (76%) patients with follicular lymphoma and 5/17 (29%) patients with diffuse large B-cell lymphoma achieved complete or partial responses with an acceptable safety profile when treated with the recommended Phase II dose of polatuzumab vedotin in combination with venetoclax and an anti-CD20 antibody.


Subject(s)
Immunoconjugates , Lymphoma, Non-Hodgkin , Humans , Myeloid Cell Leukemia Sequence 1 Protein/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/pathology , Rituximab/therapeutic use , Immunoconjugates/therapeutic use
7.
Nat Rev Mol Cell Biol ; 12(7): 439-52, 2011 Jun 23.
Article in English | MEDLINE | ID: mdl-21697901

ABSTRACT

The proper regulation of apoptosis is essential for the survival of multicellular organisms. Furthermore, excessive apoptosis can contribute to neurodegenerative diseases, anaemia and graft rejection, and diminished apoptosis can lead to autoimmune diseases and cancer. It has become clear that the post-translational modification of apoptotic proteins by ubiquitylation regulates key components in cell death signalling cascades. For example, ubiquitin E3 ligases, such as MDM2 (which ubiquitylates p53) and inhibitor of apoptosis (IAP) proteins, and deubiquitinases, such as A20 and ubiquitin-specific protease 9X (USP9X) (which regulate the ubiquitylation and degradation of receptor-interacting protein 1 (RIP1) and myeloid leukaemia cell differentiation 1 (MCL1), respectively), have important roles in apoptosis. Therapeutic agents that target apoptotic regulatory proteins, including those that are part of the ubiquitin-proteasome system, might afford clinical benefits.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Animals , Humans , Models, Biological , Protein Processing, Post-Translational , Signal Transduction , Ubiquitination
8.
Cell ; 134(4): 668-78, 2008 Aug 22.
Article in English | MEDLINE | ID: mdl-18724939

ABSTRACT

Posttranslational modification of proteins with polyubiquitin occurs in diverse signaling pathways and is tightly regulated to ensure cellular homeostasis. Studies employing ubiquitin mutants suggest that the fate of polyubiquitinated proteins is determined by which lysine within ubiquitin is linked to the C terminus of an adjacent ubiquitin. We have developed linkage-specific antibodies that recognize polyubiquitin chains joined through lysine 63 (K63) or 48 (K48). A cocrystal structure of an anti-K63 linkage Fab bound to K63-linked diubiquitin provides insight into the molecular basis for specificity. We use these antibodies to demonstrate that RIP1, which is essential for tumor necrosis factor-induced NF-kappaB activation, and IRAK1, which participates in signaling by interleukin-1beta and Toll-like receptors, both undergo polyubiquitin editing in stimulated cells. Both kinase adaptors initially acquire K63-linked polyubiquitin, while at later times K48-linked polyubiquitin targets them for proteasomal degradation. Polyubiquitin editing may therefore be a general mechanism for attenuating innate immune signaling.


Subject(s)
Antibodies/metabolism , Nuclear Pore Complex Proteins/metabolism , RNA-Binding Proteins/metabolism , Ubiquitin/metabolism , Animals , Cell Line , Humans , Interleukin-1 Receptor-Associated Kinases/metabolism , Mass Spectrometry , Mice , Mice, Inbred C57BL , Nuclear Pore Complex Proteins/chemistry , Peptide Library , RNA-Binding Proteins/chemistry , Saccharomyces cerevisiae , Schizosaccharomyces , Ubiquitin/chemistry , Ubiquitination
9.
Nature ; 550(7677): 534-538, 2017 10 26.
Article in English | MEDLINE | ID: mdl-29045385

ABSTRACT

The ubiquitin system regulates essential cellular processes in eukaryotes. Ubiquitin is ligated to substrate proteins as monomers or chains and the topology of ubiquitin modifications regulates substrate interactions with specific proteins. Thus ubiquitination directs a variety of substrate fates including proteasomal degradation. Deubiquitinase enzymes cleave ubiquitin from substrates and are implicated in disease; for example, ubiquitin-specific protease-7 (USP7) regulates stability of the p53 tumour suppressor and other proteins critical for tumour cell survival. However, developing selective deubiquitinase inhibitors has been challenging and no co-crystal structures have been solved with small-molecule inhibitors. Here, using nuclear magnetic resonance-based screening and structure-based design, we describe the development of selective USP7 inhibitors GNE-6640 and GNE-6776. These compounds induce tumour cell death and enhance cytotoxicity with chemotherapeutic agents and targeted compounds, including PIM kinase inhibitors. Structural studies reveal that GNE-6640 and GNE-6776 non-covalently target USP7 12 Å distant from the catalytic cysteine. The compounds attenuate ubiquitin binding and thus inhibit USP7 deubiquitinase activity. GNE-6640 and GNE-6776 interact with acidic residues that mediate hydrogen-bond interactions with the ubiquitin Lys48 side chain, suggesting that USP7 preferentially interacts with and cleaves ubiquitin moieties that have free Lys48 side chains. We investigated this idea by engineering di-ubiquitin chains containing differential proximal and distal isotopic labels and measuring USP7 binding by nuclear magnetic resonance. This preferential binding protracted the depolymerization kinetics of Lys48-linked ubiquitin chains relative to Lys63-linked chains. In summary, engineering compounds that inhibit USP7 activity by attenuating ubiquitin binding suggests opportunities for developing other deubiquitinase inhibitors and may be a strategy more broadly applicable to inhibiting proteins that require ubiquitin binding for full functional activity.


Subject(s)
Aminopyridines/chemistry , Aminopyridines/pharmacology , Indazoles/chemistry , Indazoles/pharmacology , Phenols/chemistry , Phenols/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors , Ubiquitin/metabolism , Animals , Binding, Competitive , Cell Line, Tumor , Drug Synergism , Female , Humans , Mice , Mice, SCID , Models, Molecular , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/pathology , Protein Binding , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Substrate Specificity , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin/chemistry , Ubiquitin-Specific Peptidase 7/chemistry , Ubiquitin-Specific Peptidase 7/deficiency , Ubiquitin-Specific Peptidase 7/metabolism
10.
J Am Chem Soc ; 143(28): 10571-10575, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34236858

ABSTRACT

We hypothesized that the proximity-driven ubiquitylation of E3-interacting small molecules could affect the degradation of E3 ubiquitin ligases. A series of XIAP BIR2 domain-binding small molecules was modified to append a nucleophilic primary amine. This modification transforms XIAP binders into inducers of XIAP degradation. The degradation of XIAP is E1- and proteasome-dependent, dependent on the ligase function of XIAP, and is rescued by subtle modifications of the small molecule that would obviate ubiquitylation. We demonstrate in vitro ubiquitylation of the small molecule that is dependent on its interaction with XIAP. Taken together, these results demonstrate the designed ubiquitylation of an engineered small molecule and a novel approach for the degradation of E3 ubiquitin ligases.


Subject(s)
Amines/pharmacology , Small Molecule Libraries/pharmacology , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors , Amines/chemistry , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Structure , Small Molecule Libraries/chemistry , X-Linked Inhibitor of Apoptosis Protein/metabolism
11.
Nature ; 528(7582): 370-5, 2015 Dec 17.
Article in English | MEDLINE | ID: mdl-26649818

ABSTRACT

Inactivation of the TNFAIP3 gene, encoding the A20 protein, is associated with critical inflammatory diseases including multiple sclerosis, rheumatoid arthritis and Crohn's disease. However, the role of A20 in attenuating inflammatory signalling is unclear owing to paradoxical in vitro and in vivo findings. Here we utilize genetically engineered mice bearing mutations in the A20 ovarian tumour (OTU)-type deubiquitinase domain or in the zinc finger-4 (ZnF4) ubiquitin-binding motif to investigate these discrepancies. We find that phosphorylation of A20 promotes cleavage of Lys63-linked polyubiquitin chains by the OTU domain and enhances ZnF4-mediated substrate ubiquitination. Additionally, levels of linear ubiquitination dictate whether A20-deficient cells die in response to tumour necrosis factor. Mechanistically, linear ubiquitin chains preserve the architecture of the TNFR1 signalling complex by blocking A20-mediated disassembly of Lys63-linked polyubiquitin scaffolds. Collectively, our studies reveal molecular mechanisms whereby A20 deubiquitinase activity and ubiquitin binding, linear ubiquitination, and cellular kinases cooperate to regulate inflammation and cell death.


Subject(s)
Cysteine Endopeptidases/metabolism , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Ubiquitin/chemistry , Ubiquitin/metabolism , Animals , Cell Death , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/genetics , Female , Inflammation/genetics , Inflammation/pathology , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Lysine/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation , Phosphorylation , Polyubiquitin/chemistry , Polyubiquitin/metabolism , Protein Binding , Protein Kinases/metabolism , Signal Transduction , Tumor Necrosis Factor alpha-Induced Protein 3 , Tumor Necrosis Factor-alpha/metabolism , Ubiquitination
12.
Bioorg Med Chem Lett ; 30(4): 126907, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31902710

ABSTRACT

Chimeric molecules which effect intracellular degradation of target proteins via E3 ligase-mediated ubiquitination (e.g., PROTACs) are currently of high interest in medicinal chemistry. However, these entities are relatively large compounds that often possess molecular characteristics which may compromise oral bioavailability, solubility, and/or in vivo pharmacokinetic properties. Accordingly, we explored whether conjugation of chimeric degraders to monoclonal antibodies using technologies originally developed for cytotoxic payloads might provide alternate delivery options for these novel agents. In this report we describe the construction of several degrader-antibody conjugates comprised of two distinct ERα-targeting degrader entities and three independent ADC linker modalities. We subsequently demonstrate the antigen-dependent delivery to MCF7-neo/HER2 cells of the degrader payloads that are incorporated into these conjugates. We also provide evidence for efficient intracellular degrader release from one of the employed linkers. In addition, preliminary data are described which suggest that reasonably favorable in vivo stability properties are associated with the linkers utilized to construct the degrader conjugates.


Subject(s)
Antibodies, Monoclonal/immunology , Drug Carriers/chemistry , Estrogen Receptor alpha/immunology , Antibodies, Monoclonal/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacology , Drug Design , Estrogen Receptor alpha/metabolism , Humans , Immunoconjugates/chemistry , Immunoconjugates/immunology , Immunoconjugates/pharmacology , MCF-7 Cells , Proteolysis/drug effects , Receptor, ErbB-2/metabolism
13.
Drug Discov Today Technol ; 31: 109-123, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31200854

ABSTRACT

The Ubiquitin/Proteasome System comprises an essential cellular mechanism for regulated protein degradation. Ubiquitination may also promote the assembly of protein complexes that initiate intracellular signaling cascades. Thus, proper regulation of substrate protein ubiquitination is essential for maintaining normal cellular physiology. Deubiquitinases are the class of enzymes responsible for removing ubiquitin modifications from target proteins and have been implicated in regulating human disease. As such, deubiquitinases are now recognized as emerging drug targets. Small molecule deubiquitinase inhibitors have been developed; among those, inhibitors for the deubiquitinases USP7 and USP14 are the best-characterized given that they are structurally validated. In this review we discuss the normal physiological roles of the USP7 and USP14 deubiquitinases as well as the pathological conditions associated with their dysfunction, with a focus on oncology and neurodegenerative diseases. We also review structural biology of USP7 and USP14 enzymes and the characterization of their respective inhibitors, highlighting the various molecular mechanisms by which these deubiquitinases may be functionally inhibited. Finally, we summarize the cellular and in vivo studies performed using the structurally-validated USP7 and USP14 inhibitors.


Subject(s)
Pyrroles/pharmacology , Ubiquitin Thiolesterase/antagonists & inhibitors , Animals , Humans , Molecular Structure , Pyrroles/chemistry , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/metabolism
15.
Chemistry ; 24(19): 4830-4834, 2018 Apr 03.
Article in English | MEDLINE | ID: mdl-29493023

ABSTRACT

A novel strategy to attach indole-containing payloads to antibodies through a carbamate moiety and a self-immolating, disulfide-based linker is described. This new strategy was employed to connect a selective estrogen receptor down-regulator (SERD) to various antibodies in a site-selective manner. The resulting conjugates displayed potent, antigen-dependent down-regulation of estrogen receptor levels in MCF7-neo/HER2 and MCF7-hB7H4 cells. They also exhibited similar antigen-dependent modulation of the estrogen receptor in tumors when administered intravenously to mice bearing MCF7-neo/HER2 tumor xenografts. The indole-carbamate moiety present in the new linker was stable in whole blood from various species and also exhibited good in vivo stability properties in mice.


Subject(s)
Indoles/chemistry , Animals , Antibodies, Monoclonal/chemistry , Cell Line, Tumor , Humans , Immunoconjugates/administration & dosage , MCF-7 Cells , Mice
16.
Mol Cell ; 40(4): 548-57, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-21095585

ABSTRACT

Inactivating mutations in the ubiquitin (Ub) editing protein A20 promote persistent nuclear factor (NF)-κB signaling and are genetically linked to inflammatory diseases and hematologic cancers. A20 tightly regulates NF-κB signaling by acting as an Ub editor, removing K63-linked Ub chains and mediating addition of Ub chains that target substrates for degradation. However, a precise molecular understanding of how A20 modulates this pathway remains elusive. Here, using structural analysis, domain mapping, and functional assays, we show that A20 zinc finger 4 (ZnF4) does not directly interact with E2 enzymes but instead can bind mono-Ub and K63-linked poly-Ub. Mutations to the A20 ZnF4 Ub-binding surface result in decreased A20-mediated ubiquitination and impaired regulation of NF-κB signaling. Collectively, our studies illuminate the mechanistically distinct but biologically interdependent activities of the A20 ZnF and ovarian tumor (OTU) domains that are inherent to the Ub editing process and, ultimately, to regulation of NF-κB signaling.


Subject(s)
NF-kappa B/metabolism , Nuclear Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Ubiquitin/metabolism , Zinc Fingers , Binding Sites , Crystallography, X-Ray , Lysine/metabolism , Models, Molecular , Mutation/genetics , Nuclear Proteins/chemistry , Polyubiquitin/metabolism , Protein Binding , Substrate Specificity , Ubiquitin-Conjugating Enzymes/metabolism
17.
Nature ; 474(7351): 403-6, 2011 May 15.
Article in English | MEDLINE | ID: mdl-21572435

ABSTRACT

The proto-oncogenes ETV1, ETV4 and ETV5 encode transcription factors in the E26 transformation-specific (ETS) family, which includes the most frequently rearranged and overexpressed genes in prostate cancer. Despite being critical regulators of development, little is known about their post-translational regulation. Here we identify the ubiquitin ligase COP1 (also known as RFWD2) as a tumour suppressor that negatively regulates ETV1, ETV4 and ETV5. ETV1, which is mutated in prostate cancer more often, was degraded after being ubiquitinated by COP1. Truncated ETV1 encoded by prostate cancer translocation TMPRSS2:ETV1 lacks the critical COP1 binding motifs and was 50-fold more stable than wild-type ETV1. Almost all patient translocations render ETV1 insensitive to COP1, implying that this confers a selective advantage to prostate epithelial cells. Indeed, COP1 deficiency in mouse prostate elevated ETV1 and produced increased cell proliferation, hyperplasia, and early prostate intraepithelial neoplasia. Combined loss of COP1 and PTEN enhanced the invasiveness of mouse prostate adenocarcinomas. Finally, rare human prostate cancer samples showed hemizygous loss of the COP1 gene, loss of COP1 protein, and elevated ETV1 protein while lacking a translocation event. These findings identify COP1 as a tumour suppressor whose downregulation promotes prostatic epithelial cell proliferation and tumorigenesis.


Subject(s)
Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-ets/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Motifs , Animals , Carrier Proteins/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Male , Mice , Nuclear Proteins/deficiency , PTEN Phosphohydrolase/deficiency , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Binding , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Ubiquitination
18.
Nature ; 471(7336): 110-4, 2011 Mar 03.
Article in English | MEDLINE | ID: mdl-21368834

ABSTRACT

Microtubules have pivotal roles in fundamental cellular processes and are targets of antitubulin chemotherapeutics. Microtubule-targeted agents such as Taxol and vincristine are prescribed widely for various malignancies, including ovarian and breast adenocarcinomas, non-small-cell lung cancer, leukaemias and lymphomas. These agents arrest cells in mitosis and subsequently induce cell death through poorly defined mechanisms. The strategies that resistant tumour cells use to evade death induced by antitubulin agents are also unclear. Here we show that the pro-survival protein MCL1 (ref. 3) is a crucial regulator of apoptosis triggered by antitubulin chemotherapeutics. During mitotic arrest, MCL1 protein levels decline markedly, through a post-translational mechanism, potentiating cell death. Phosphorylation of MCL1 directs its interaction with the tumour-suppressor protein FBW7, which is the substrate-binding component of a ubiquitin ligase complex. The polyubiquitylation of MCL1 then targets it for proteasomal degradation. The degradation of MCL1 was blocked in patient-derived tumour cells that lacked FBW7 or had loss-of-function mutations in FBW7, conferring resistance to antitubulin agents and promoting chemotherapeutic-induced polyploidy. Additionally, primary tumour samples were enriched for FBW7 inactivation and elevated MCL1 levels, underscoring the prominent roles of these proteins in oncogenesis. Our findings suggest that profiling the FBW7 and MCL1 status of tumours, in terms of protein levels, messenger RNA levels and genetic status, could be useful to predict the response of patients to antitubulin chemotherapeutics.


Subject(s)
Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Tubulin Modulators/pharmacology , Tubulin/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Apoptosis/drug effects , Cell Cycle Proteins/genetics , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Drug Resistance, Neoplasm , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Fibroblasts , Humans , Mice , Mitosis/drug effects , Myeloid Cell Leukemia Sequence 1 Protein , Paclitaxel/pharmacology , Pharmacogenetics , Phosphorylation/drug effects , Polyploidy , Proteasome Endopeptidase Complex/metabolism , Protein Binding/drug effects , Proto-Oncogene Proteins c-bcl-2/deficiency , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Vincristine/pharmacology
20.
Biochem J ; 465(1): 1-26, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25631680

ABSTRACT

The post-translational modification of proteins with ubiquitin represents a complex signalling system that co-ordinates essential cellular functions, including proteolysis, DNA repair, receptor signalling and cell communication. DUBs (deubiquitinases), the enzymes that disassemble ubiquitin chains and remove ubiquitin from proteins, are central to this system. Reflecting the complexity and versatility of ubiquitin signalling, DUB activity is controlled in multiple ways. Although several lines of evidence indicate that aberrant DUB function may promote human disease, the underlying molecular mechanisms are often unclear. Notwithstanding, considerable interest in DUBs as potential drug targets has emerged over the past years. The future success of DUB-based therapy development will require connecting the basic science of DUB function and enzymology with drug discovery. In the present review, we discuss new insights into DUB activity regulation and their links to disease, focusing on the role of DUBs as regulators of cell identity and differentiation, and discuss their potential as emerging drug targets.


Subject(s)
Cells/enzymology , Cells/pathology , Disease , Ubiquitin-Specific Proteases/metabolism , Animals , Humans , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL