Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Cell ; 145(7): 1075-87, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21683433

ABSTRACT

In the ubiquitin-proteasome system (UPS), E2 enzymes mediate the conjugation of ubiquitin to substrates and thereby control protein stability and interactions. The E2 enzyme hCdc34 catalyzes the ubiquitination of hundreds of proteins in conjunction with the cullin-RING (CRL) superfamily of E3 enzymes. We identified a small molecule termed CC0651 that selectively inhibits hCdc34. Structure determination revealed that CC0651 inserts into a cryptic binding pocket on hCdc34 distant from the catalytic site, causing subtle but wholesale displacement of E2 secondary structural elements. CC0651 analogs inhibited proliferation of human cancer cell lines and caused accumulation of the SCF(Skp2) substrate p27(Kip1). CC0651 does not affect hCdc34 interactions with E1 or E3 enzymes or the formation of the ubiquitin thioester but instead interferes with the discharge of ubiquitin to acceptor lysine residues. E2 enzymes are thus susceptible to noncatalytic site inhibition and may represent a viable class of drug target in the UPS.


Subject(s)
Amino Acids/pharmacology , Biphenyl Compounds/pharmacology , Ubiquitin-Protein Ligase Complexes/antagonists & inhibitors , Allosteric Site , Amino Acid Sequence , Anaphase-Promoting Complex-Cyclosome , DNA Mutational Analysis , Humans , Models, Molecular , Molecular Sequence Data , Sequence Alignment , Ubiquitin-Conjugating Enzymes , Ubiquitin-Protein Ligase Complexes/chemistry , Ubiquitin-Protein Ligase Complexes/genetics
2.
Mol Cell ; 59(6): 970-83, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26344097

ABSTRACT

BRCC36 is a Zn(2+)-dependent deubiquitinating enzyme (DUB) that hydrolyzes lysine-63-linked ubiquitin chains as part of distinct macromolecular complexes that participate in either interferon signaling or DNA-damage recognition. The MPN(+) domain protein BRCC36 associates with pseudo DUB MPN(-) proteins KIAA0157 or Abraxas, which are essential for BRCC36 enzymatic activity. To understand the basis for BRCC36 regulation, we have solved the structure of an active BRCC36-KIAA0157 heterodimer and an inactive BRCC36 homodimer. Structural and functional characterizations show how BRCC36 is switched to an active conformation by contacts with KIAA0157. Higher-order association of BRCC36 and KIAA0157 into a dimer of heterodimers (super dimers) was required for DUB activity and interaction with targeting proteins SHMT2 and RAP80. These data provide an explanation of how an inactive pseudo DUB allosterically activates a cognate DUB partner and implicates super dimerization as a new regulatory mechanism underlying BRCC36 DUB activity, subcellular localization, and biological function.


Subject(s)
Ants/enzymology , Insect Proteins/chemistry , Nuclear Matrix-Associated Proteins/chemistry , Ubiquitin-Specific Proteases/chemistry , Animals , Catalytic Domain , Crystallography, X-Ray , Deubiquitinating Enzymes , HEK293 Cells , HeLa Cells , Humans , Insect Proteins/physiology , Kinetics , Membrane Proteins/chemistry , Models, Molecular , Nuclear Matrix-Associated Proteins/physiology , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Secondary , Ubiquitin-Specific Proteases/physiology
3.
Nat Chem Biol ; 16(11): 1170-1178, 2020 11.
Article in English | MEDLINE | ID: mdl-32778845

ABSTRACT

The RAF family kinases function in the RAS-ERK pathway to transmit signals from activated RAS to the downstream kinases MEK and ERK. This pathway regulates cell proliferation, differentiation and survival, enabling mutations in RAS and RAF to act as potent drivers of human cancers. Drugs targeting the prevalent oncogenic mutant BRAF(V600E) have shown great efficacy in the clinic, but long-term effectiveness is limited by resistance mechanisms that often exploit the dimerization-dependent process by which RAF kinases are activated. Here, we investigated a proteolysis-targeting chimera (PROTAC) approach to BRAF inhibition. The most effective PROTAC, termed P4B, displayed superior specificity and inhibitory properties relative to non-PROTAC controls in BRAF(V600E) cell lines. In addition, P4B displayed utility in cell lines harboring alternative BRAF mutations that impart resistance to conventional BRAF inhibitors. This work provides a proof of concept for a substitute to conventional chemical inhibition to therapeutically constrain oncogenic BRAF.


Subject(s)
Antineoplastic Agents , Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Thalidomide , Ubiquitin , Animals , Female , Humans , Mice , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Drug Resistance, Neoplasm , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Kinases/metabolism , Models, Molecular , Molecular Structure , Molecular Targeted Therapy , Mutation , Phosphorylation/drug effects , Protein Binding , Protein Kinase Inhibitors/pharmacology , Proteolysis , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Signal Transduction , Structure-Activity Relationship , Thalidomide/analogs & derivatives , Thalidomide/chemistry , Ubiquitin/chemistry
4.
Mol Cell ; 53(2): 221-34, 2014 Jan 23.
Article in English | MEDLINE | ID: mdl-24462203

ABSTRACT

RNase L is an ankyrin repeat domain-containing dual endoribonuclease-pseudokinase that is activated by unusual 2,'5'-oligoadenylate (2-5A) second messengers and which impedes viral infections in higher vertebrates. Despite its importance in interferon-regulated antiviral innate immunity, relatively little is known about its precise mechanism of action. Here we present a functional characterization of 2.5 Å and 3.25 Å X-ray crystal and small-angle X-ray scattering structures of RNase L bound to a natural 2-5A activator with and without ADP or the nonhydrolysable ATP mimetic AMP-PNP. These studies reveal how recognition of 2-5A through interactions with the ankyrin repeat domain and the pseudokinase domain, together with nucleotide binding, imposes a rigid intertwined dimer configuration that is essential for RNase catalytic and antiviral functions. The involvement of the pseudokinase domain of RNase L in 2-5A sensing, nucleotide binding, dimerization, and ribonuclease functions highlights the evolutionary adaptability of the eukaryotic protein kinase fold.


Subject(s)
Adenine Nucleotides/chemistry , Endoribonucleases/chemistry , Oligoribonucleotides/chemistry , Adenosine Diphosphate/chemistry , Adenylyl Imidodiphosphate/chemistry , Animals , Ankyrin Repeat , Binding Sites , Crystallography, X-Ray , Dimerization , Encephalomyocarditis virus , Endoribonucleases/genetics , Endoribonucleases/physiology , HeLa Cells , Humans , Models, Molecular , Mutagenesis, Site-Directed , Picornaviridae , Protein Structure, Tertiary , Scattering, Radiation , Structure-Activity Relationship , Sus scrofa
5.
Mol Cell ; 45(3): 384-97, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-22325355

ABSTRACT

Ubiquitylation entails the concerted action of E1, E2, and E3 enzymes. We recently reported that OTUB1, a deubiquitylase, inhibits the DNA damage response independently of its isopeptidase activity. OTUB1 does so by blocking ubiquitin transfer by UBC13, the cognate E2 enzyme for RNF168. OTUB1 also inhibits E2s of the UBE2D and UBE2E families. Here we elucidate the structural mechanism by which OTUB1 binds E2s to inhibit ubiquitin transfer. OTUB1 recognizes ubiquitin-charged E2s through contacts with both donor ubiquitin and the E2 enzyme. Surprisingly, free ubiquitin associates with the canonical distal ubiquitin-binding site on OTUB1 to promote formation of the inhibited E2 complex. Lys48 of donor ubiquitin lies near the OTUB1 catalytic site and the C terminus of free ubiquitin, a configuration that mimics the products of Lys48-linked ubiquitin chain cleavage. OTUB1 therefore co-opts Lys48-linked ubiquitin chain recognition to suppress ubiquitin conjugation and the DNA damage response.


Subject(s)
Cysteine Endopeptidases/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitinated Proteins/metabolism , Amino Acid Substitution , Cell Line , Crystallography, X-Ray , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/genetics , Deubiquitinating Enzymes , Humans , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Organisms, Genetically Modified , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Ubiquitin/chemistry , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitination , Yeasts/genetics , Yeasts/growth & development
6.
Nature ; 498(7454): 318-24, 2013 Jun 20.
Article in English | MEDLINE | ID: mdl-23708998

ABSTRACT

A complex interaction of signalling events, including the Wnt pathway, regulates sprouting of blood vessels from pre-existing vasculature during angiogenesis. Here we show that two distinct mutations in the (uro)chordate-specific gumby (also called Fam105b) gene cause an embryonic angiogenic phenotype in gumby mice. Gumby interacts with disheveled 2 (DVL2), is expressed in canonical Wnt-responsive endothelial cells and encodes an ovarian tumour domain class of deubiquitinase that specifically cleaves linear ubiquitin linkages. A crystal structure of gumby in complex with linear diubiquitin reveals how the identified mutations adversely affect substrate binding and catalytic function in line with the severity of their angiogenic phenotypes. Gumby interacts with HOIP (also called RNF31), a key component of the linear ubiquitin assembly complex, and decreases linear ubiquitination and activation of NF-κB-dependent transcription. This work provides support for the biological importance of linear (de)ubiquitination in angiogenesis, craniofacial and neural development and in modulating Wnt signalling.


Subject(s)
Endopeptidases/chemistry , Endopeptidases/metabolism , Neovascularization, Physiologic , Ubiquitin/chemistry , Ubiquitin/metabolism , Ubiquitination , Adaptor Proteins, Signal Transducing/metabolism , Alleles , Amino Acid Sequence , Animals , Base Sequence , Crystallography, X-Ray , Dishevelled Proteins , Embryo, Mammalian/blood supply , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Endopeptidases/deficiency , Endopeptidases/genetics , Female , Gene Expression Profiling , HEK293 Cells , Humans , Mice , Models, Molecular , Molecular Sequence Data , Neovascularization, Physiologic/genetics , Phenotype , Phosphoproteins/metabolism , Protein Conformation , Ubiquitin-Protein Ligases/metabolism , Wnt Signaling Pathway
7.
BMC Biol ; 16(1): 88, 2018 08 10.
Article in English | MEDLINE | ID: mdl-30097011

ABSTRACT

BACKGROUND: The ubiquitin-proteasome system (UPS) controls the stability, localization and/or activity of the proteome. However, the identification and characterization of complex individual ubiquitination cascades and their modulators remains a challenge. Here, we report a broadly applicable, multiplexed, miniaturized on-bead technique for real-time monitoring of various ubiquitination-related enzymatic activities. The assay, termed UPS-confocal fluorescence nanoscanning (UPS-CONA), employs a substrate of interest immobilized on a micro-bead and a fluorescently labeled ubiquitin which, upon enzymatic conjugation to the substrate, is quantitatively detected on the bead periphery by confocal imaging. RESULTS: UPS-CONA is suitable for studying individual enzymatic activities, including various E1, E2, and HECT-type E3 enzymes, and for monitoring multi-step reactions within ubiquitination cascades in a single experimental compartment. We demonstrate the power of the UPS-CONA technique by simultaneously following ubiquitin transfer from Ube1 through Ube2L3 to E6AP. We applied this multi-step setup to investigate the selectivity of five ubiquitination inhibitors reportedly targeting different classes of ubiquitination enzymes. Using UPS-CONA, we have identified a new activity of a small molecule E2 inhibitor, BAY 11-7082, and of a HECT E3 inhibitor, heclin, towards the Ube1 enzyme. CONCLUSIONS: As a sensitive, quantitative, flexible, and reagent-efficient method with a straightforward protocol, UPS-CONA constitutes a powerful tool for interrogation of ubiquitination-related enzymatic pathways and their chemical modulators, and is readily scalable for large experiments.


Subject(s)
Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Proteasome Endopeptidase Complex/chemistry , Ubiquitination , Humans , Microscopy, Confocal/instrumentation , Microscopy, Fluorescence/instrumentation
8.
Nat Chem Biol ; 10(2): 156-163, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24316736

ABSTRACT

Weak protein interactions between ubiquitin and the ubiquitin-proteasome system (UPS) enzymes that mediate its covalent attachment to substrates serve to position ubiquitin for optimal catalytic transfer. We show that a small-molecule inhibitor of the E2 ubiquitin-conjugating enzyme Cdc34A, called CC0651, acts by trapping a weak interaction between ubiquitin and the E2 donor ubiquitin-binding site. A structure of the ternary CC0651-Cdc34A-ubiquitin complex reveals that the inhibitor engages a composite binding pocket formed from Cdc34A and ubiquitin. CC0651 also suppresses the spontaneous hydrolysis rate of the Cdc34A-ubiquitin thioester without decreasing the interaction between Cdc34A and the RING domain subunit of the E3 enzyme. Stabilization of the numerous other weak interactions between ubiquitin and UPS enzymes by small molecules may be a feasible strategy to selectively inhibit different UPS activities.


Subject(s)
Amino Acids/chemistry , Biphenyl Compounds/chemistry , Ubiquitin-Conjugating Enzymes/antagonists & inhibitors , Ubiquitin/chemistry , Amino Acids/pharmacology , Binding Sites , Biphenyl Compounds/pharmacology , Coordination Complexes/chemistry , Crystallography, X-Ray , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Inhibitory Concentration 50 , Models, Molecular , Protein Binding , Protein Stability/drug effects , Small Molecule Libraries/pharmacology
9.
Mol Cell ; 29(5): 535-7, 2008 Mar 14.
Article in English | MEDLINE | ID: mdl-18342599

ABSTRACT

In this issue of Molecular Cell, Ngo et al. (2008) describe the crystal structure of the SRPK1 protein kinase in complex with its substrate, the spliceosome factor ASF/SF2, providing an unprecedented view of multiple targeting mechanisms in action on a single substrate.


Subject(s)
Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , Animals , Humans , Mice , Nuclear Proteins/genetics , Phosphorylation , Protein Serine-Threonine Kinases/genetics , RNA-Binding Proteins , Serine-Arginine Splicing Factors , Substrate Specificity
10.
Mol Cell ; 32(4): 564-75, 2008 Nov 21.
Article in English | MEDLINE | ID: mdl-19026786

ABSTRACT

PH domains, by binding to phosphoinositides, often serve as membrane-targeting modules. Using crystallographic, biochemical, and cell biological approaches, we have uncovered a mechanism that the integrin-signaling adaptor Skap-hom uses to mediate cytoskeletal interactions. Skap-hom is a homodimer containing an N-terminal four-helix bundle dimerization domain, against which its two PH domains pack in a conformation incompatible with phosphoinositide binding. The isolated PH domains bind PI[3,4,5]P(3), and mutations targeting the dimerization domain or the PH domain's PI[3,4,5]P(3)-binding pocket prevent Skap-hom localization to ruffles. Targeting is retained when the PH domain is deleted or by combined mutation of the PI[3,4,5]P(3)-binding pocket and the PH/dimerization domain interface. Thus, the dimerization and PH domain form a PI[3,4,5]P(3)-responsive molecular switch that controls Skap-hom function.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Phosphatidylinositols/chemistry , Phosphatidylinositols/metabolism , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Animals , Dimerization , Green Fluorescent Proteins/metabolism , Mice , Mice, Knockout , Models, Chemical , Models, Molecular , Molecular Sequence Data , Phosphatidylinositols/genetics , Point Mutation , Protein Binding , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Retroviridae/genetics , Sequence Homology, Amino Acid , Transduction, Genetic
11.
Mol Cell ; 32(2): 259-75, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18951093

ABSTRACT

Kae1 is a universally conserved ATPase and part of the essential gene set in bacteria. In archaea and eukaryotes, Kae1 is embedded within the protein kinase-containing KEOPS complex. Mutation of KEOPS subunits in yeast leads to striking telomere and transcription defects, but the exact biochemical function of KEOPS is not known. As a first step to elucidating its function, we solved the atomic structure of archaea-derived KEOPS complexes involving Kae1, Bud32, Pcc1, and Cgi121 subunits. Our studies suggest that Kae1 is regulated at two levels by the primordial protein kinase Bud32, which is itself regulated by Cgi121. Moreover, Pcc1 appears to function as a dimerization module, perhaps suggesting that KEOPS may be a processive molecular machine. Lastly, as Bud32 lacks the conventional substrate-recognition infrastructure of eukaryotic protein kinases including an activation segment, Bud32 may provide a glimpse of the evolutionary history of the protein kinase family.


Subject(s)
Archaeal Proteins/chemistry , Multiprotein Complexes/chemistry , Protein Kinases/chemistry , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Carrier Proteins/chemistry , Crystallography, X-Ray , Escherichia coli/genetics , Humans , Intracellular Signaling Peptides and Proteins , Methanococcus/genetics , Methanococcus/metabolism , Models, Molecular , Multiprotein Complexes/physiology , Nuclear Magnetic Resonance, Biomolecular , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Structure, Tertiary , Protein Subunits/chemistry , Sequence Homology, Amino Acid , Telomere/metabolism , Thermoplasma/genetics , Thermoplasma/metabolism , Transcription, Genetic
12.
J Biol Chem ; 287(35): 29285-9, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22782892

ABSTRACT

The TrkA receptor tyrosine kinase induces death in medulloblastoma cells via an interaction with the cerebral cavernous malformation 2 (CCM2) protein. We used affinity proteomics to identify the germinal center kinase class III (GCKIII) kinases STK24 and STK25 as novel CCM2 interactors. Down-modulation of STK25, but not STK24, rescued medulloblastoma cells from NGF-induced TrkA-dependent cell death, suggesting that STK25 is part of the death-signaling pathway initiated by TrkA and CCM2. CCM2 can be phosphorylated by STK25, and the kinase activity of STK25 is required for death signaling. Finally, STK25 expression in tumors is correlated with positive prognosis in neuroblastoma patients. These findings delineate a death-signaling pathway downstream of neurotrophic receptor tyrosine kinases that may provide targets for therapeutic intervention in pediatric tumors of neural origin.


Subject(s)
Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Medulloblastoma/metabolism , Microfilament Proteins/metabolism , Neoplasm Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor, trkA/metabolism , Signal Transduction , Adolescent , Animals , Carrier Proteins/genetics , Cell Death , Cell Line, Tumor , Child , Child, Preschool , Female , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Medulloblastoma/genetics , Medulloblastoma/pathology , Mice , Microfilament Proteins/genetics , Neoplasm Proteins/genetics , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proteomics , Receptor, trkA/genetics
13.
J Biol Chem ; 286(28): 25065-75, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21561862

ABSTRACT

Cerebral cavernous malformations (CCMs) are alterations in brain capillary architecture that can result in neurological deficits, seizures, or stroke. We recently demonstrated that CCM3, a protein mutated in familial CCMs, resides predominantly within the STRIPAK complex (striatin interacting phosphatase and kinase). Along with CCM3, STRIPAK contains the Ser/Thr phosphatase PP2A. The PP2A holoenzyme consists of a core catalytic subunit along with variable scaffolding and regulatory subunits. Within STRIPAK, striatin family members act as PP2A regulatory subunits. STRIPAK also contains all three members of a subfamily of Sterile 20 kinases called the GCKIII proteins (MST4, STK24, and STK25). Here, we report that striatins and CCM3 bridge the phosphatase and kinase components of STRIPAK and map the interacting regions on each protein. We show that striatins and CCM3 regulate the Golgi localization of MST4 in an opposite manner. Consistent with a previously described function for MST4 and CCM3 in Golgi positioning, depletion of CCM3 or striatins affects Golgi polarization, also in an opposite manner. We propose that STRIPAK regulates the balance between MST4 localization at the Golgi and in the cytosol to control Golgi positioning.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Golgi Apparatus/metabolism , Membrane Proteins/metabolism , Multiprotein Complexes/metabolism , Protein Phosphatase 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Calmodulin-Binding Proteins/chemistry , Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/metabolism , Germinal Center Kinases , Golgi Apparatus/chemistry , Golgi Apparatus/genetics , HEK293 Cells , HeLa Cells , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Phosphatase 2/chemistry , Protein Phosphatase 2/genetics , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Structure-Activity Relationship
14.
J Biol Chem ; 286(28): 25056-64, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21561863

ABSTRACT

CCM3 mutations give rise to cerebral cavernous malformations (CCMs) of the vasculature through a mechanism that remains unclear. Interaction of CCM3 with the germinal center kinase III (GCKIII) subfamily of Sterile 20 protein kinases, MST4, STK24, and STK25, has been implicated in cardiovascular development in the zebrafish, raising the possibility that dysregulated GCKIII function may contribute to the etiology of CCM disease. Here, we show that the amino-terminal region of CCM3 is necessary and sufficient to bind directly to the C-terminal tail region of GCKIII proteins. This same region of CCM3 was shown previously to mediate homodimerization through the formation of an interdigitated α-helical domain. Sequence conservation and binding studies suggest that CCM3 may preferentially heterodimerize with GCKIII proteins through a manner structurally analogous to that employed for CCM3 homodimerization.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Membrane Proteins/metabolism , Protein Multimerization/physiology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Cardiovascular System/embryology , Germinal Center Kinases , HEK293 Cells , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Organogenesis/physiology , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Protein Structure, Secondary , Protein Structure, Tertiary , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Zebrafish/genetics , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
15.
J Med Chem ; 65(19): 12725-12746, 2022 10 13.
Article in English | MEDLINE | ID: mdl-36117290

ABSTRACT

Targeted protein degradation (TPD) strategies exploit bivalent small molecules to bridge substrate proteins to an E3 ubiquitin ligase to induce substrate degradation. Few E3s have been explored as degradation effectors due to a dearth of E3-binding small molecules. We show that genetically induced recruitment to the GID4 subunit of the CTLH E3 complex induces protein degradation. An NMR-based fragment screen followed by structure-guided analog elaboration identified two binders of GID4, 16 and 67, with Kd values of 110 and 17 µM in vitro. A parallel DNA-encoded library (DEL) screen identified five binders of GID4, the best of which, 88, had a Kd of 5.6 µM in vitro and an EC50 of 558 nM in cells with strong selectivity for GID4. X-ray co-structure determination revealed the basis for GID4-small molecule interactions. These results position GID4-CTLH as an E3 for TPD and provide candidate scaffolds for high-affinity moieties that bind GID4.


Subject(s)
DNA , Ubiquitin-Protein Ligases , DNA/metabolism , Humans , Proteolysis , Ubiquitin-Protein Ligases/metabolism
16.
Structure ; 29(9): 975-988.e5, 2021 09 02.
Article in English | MEDLINE | ID: mdl-33989513

ABSTRACT

Skp2 and cyclin A are cell-cycle regulators that control the activity of CDK2. Cyclin A acts as an activator and substrate recruitment factor of CDK2, while Skp2 mediates the ubiquitination and subsequent destruction of the CDK inhibitor protein p27. The N terminus of Skp2 can interact directly with cyclin A but is not required for p27 ubiquitination. To gain insight into this poorly understood interaction, we have solved the 3.2 Å X-ray crystal structure of the N terminus of Skp2 bound to cyclin A. The structure reveals a bipartite mode of interaction with two motifs in Skp2 recognizing two discrete surfaces on cyclin A. The uncovered binding mechanism allows for a rationalization of the inhibitory effect of Skp2 on CDK2-cyclin A kinase activity toward the RxL motif containing substrates and raises the possibility that other intermolecular regulators and substrates may use similar non-canonical modes of interaction for cyclin targeting.


Subject(s)
Cyclin A/metabolism , S-Phase Kinase-Associated Proteins/chemistry , Binding Sites , Cyclin A/chemistry , Humans , Molecular Docking Simulation , Protein Binding , S-Phase Kinase-Associated Proteins/metabolism
17.
Sci Adv ; 7(44): eabi5797, 2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34705497

ABSTRACT

Pharmacological control of the ubiquitin-proteasome system (UPS) is of intense interest in drug discovery. Here, we report the development of chemical inhibitors of the ubiquitin-conjugating (E2) enzyme CDC34A (also known as UBE2R1), which donates activated ubiquitin to the cullin-RING ligase (CRL) family of ubiquitin ligase (E3) enzymes. A FRET-based interaction assay was used to screen for novel compounds that stabilize the noncovalent complex between CDC34A and ubiquitin, and thereby inhibit the CDC34A catalytic cycle. An isonipecotamide hit compound was elaborated into analogs with ~1000-fold increased potency in stabilizing the CDC34A-ubiquitin complex. These analogs specifically inhibited CDC34A-dependent ubiquitination in vitro and stabilized an E2~ubiquitin thioester reaction intermediate in cells. The x-ray crystal structure of a CDC34A-ubiquitin-inhibitor complex uncovered the basis for analog structure-activity relationships. The development of chemical stabilizers of the CDC34A-ubiquitin complex illustrates a general strategy for de novo discovery of molecular glue compounds that stabilize weak protein interactions.

18.
J Mol Biol ; 432(4): 952-966, 2020 02 14.
Article in English | MEDLINE | ID: mdl-31634471

ABSTRACT

Ubiquitin-conjugating E2 enzymes are central to the ubiquitination cascade and have been implicated in cancer and other diseases. Despite strong interest in developing specific E2 inhibitors, the shallow and exposed active site has proven recalcitrant to targeting with reversible small-molecule inhibitors. Here, we used phage display to generate highly potent and selective ubiquitin variants (UbVs) that target the E2 backside, which is located opposite to the active site. A UbV targeting Ube2D1 did not affect charging but greatly attenuated chain elongation. Likewise, a UbV targeting the E2 variant Ube2V1 did not interfere with the charging of its partner E2 enzyme but inhibited formation of diubiquitin. In contrast, a UbV that bound to the backside of Ube2G1 impeded the generation of thioester-linked ubiquitin to the active site cysteine of Ube2G1 by the E1 enzyme. Crystal structures of UbVs in complex with three E2 proteins revealed distinctive molecular interactions in each case, but they also highlighted a common backside pocket that the UbVs used for enhanced affinity and specificity. These findings validate the E2 backside as a target for inhibition and provide structural insights to aid inhibitor design and screening efforts.


Subject(s)
Enzyme Inhibitors/pharmacology , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin/metabolism , Amino Acid Sequence , Enzyme Inhibitors/chemistry , Humans , Peptide Library , Protein Binding , Protein Engineering/methods , Protein Structure, Secondary , Ubiquitin-Conjugating Enzymes/antagonists & inhibitors , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitination
19.
Nat Commun ; 11(1): 6233, 2020 12 04.
Article in English | MEDLINE | ID: mdl-33277478

ABSTRACT

The KEOPS complex, which is conserved across archaea and eukaryotes, is composed of four core subunits; Pcc1, Kae1, Bud32 and Cgi121. KEOPS is crucial for the fitness of all organisms examined. In humans, pathogenic mutations in KEOPS genes lead to Galloway-Mowat syndrome, an autosomal-recessive disease causing childhood lethality. Kae1 catalyzes the universal and essential tRNA modification N6-threonylcarbamoyl adenosine, but the precise roles of all other KEOPS subunits remain an enigma. Here we show using structure-guided studies that Cgi121 recruits tRNA to KEOPS by binding to its 3' CCA tail. A composite model of KEOPS bound to tRNA reveals that all KEOPS subunits form an extended tRNA-binding surface that we have validated in vitro and in vivo to mediate the interaction with the tRNA substrate and its modification. These findings provide a framework for understanding the inner workings of KEOPS and delineate why all KEOPS subunits are essential.


Subject(s)
Archaeal Proteins/chemistry , Methanocaldococcus/metabolism , Multiprotein Complexes/chemistry , RNA, Transfer/chemistry , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Crystallography, X-Ray , Methanocaldococcus/genetics , Models, Molecular , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Nucleic Acid Conformation , Protein Binding , Protein Domains , RNA, Transfer/genetics , RNA, Transfer/metabolism , RNA, Transfer, Lys/chemistry , RNA, Transfer, Lys/genetics , RNA, Transfer, Lys/metabolism
20.
Structure ; 27(6): 1000-1012.e6, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31056421

ABSTRACT

Pseudoenzymes have been identified across a diverse range of enzyme classes and fulfill important cellular functions. Examples of pseudoenzymes exist within ubiquitin conjugating and deubiquitinase (DUB) protein families. Here we characterize FAM105A/OTULINL, the only putative pseudodeubiquitinase of the ovarian tumor protease (OTU domain) family in humans. The crystal structure of FAM105A revealed that the OTU domain possesses structural deficiencies in both active site and substrate-binding infrastructure predicted to impair normal DUB function. We confirmed the absence of catalytic function against all ubiquitin linkages and an inability of FAM105A to bind ubiquitin compared with catalytically active FAM105B/OTULIN. FAM105A co-localized with KDEL markers and Lamin B1 at the endoplasmic reticulum (ER) and nuclear envelope, respectively. Accordingly, the FAM105A interactome exhibited significant enrichment in proteins localized to the ER/outer nuclear, Golgi and vesicular membranes. In light of undetectable deubiquitinase activity, we posit that FAM105A/OTULINL functions through its ability to mediate protein-protein interactions.


Subject(s)
Deubiquitinating Enzymes/chemistry , Endopeptidases/chemistry , Endoplasmic Reticulum/metabolism , Intracellular Membranes/metabolism , Amino Acid Sequence , Animals , Catalytic Domain , Cell Line, Tumor , Crystallography, X-Ray , Deubiquitinating Enzymes/genetics , Deubiquitinating Enzymes/metabolism , Endopeptidases/genetics , Endopeptidases/metabolism , HEK293 Cells , Humans , Mice , Models, Molecular , Protein Binding , Protein Domains , Sequence Homology, Amino Acid , Ubiquitin/chemistry , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL