Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Nat Commun ; 15(1): 489, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38216578

ABSTRACT

Although stapled α-helical peptides can address challenging targets, their advancement is impeded by poor understandings for making them cell permeable while avoiding off-target toxicities. By synthesizing >350 molecules, we present workflows for identifying stapled peptides against Mdm2(X) with in vivo activity and no off-target effects. Key insights include a clear correlation between lipophilicity and permeability, removal of positive charge to avoid off-target toxicities, judicious anionic residue placement to enhance solubility/behavior, optimization of C-terminal length/helicity to enhance potency, and optimization of staple type/number to avoid polypharmacology. Workflow application gives peptides with >292x improved cell proliferation potencies and no off-target cell proliferation effects ( > 3800x on-target index). Application of these 'design rules' to a distinct Mdm2(X) peptide series improves ( > 150x) cellular potencies and removes off-target toxicities. The outlined workflow should facilitate therapeutic impacts, especially for those targets such as Mdm2(X) that have hydrophobic interfaces and are targetable with a helical motif.


Subject(s)
Peptides , Proto-Oncogene Proteins c-mdm2 , Peptides/pharmacology , Peptides/chemistry
2.
ACS Cent Sci ; 7(2): 274-291, 2021 Feb 24.
Article in English | MEDLINE | ID: mdl-33655066

ABSTRACT

Mutations to RAS proteins (H-, N-, and K-RAS) are among the most common oncogenic drivers, and tumors harboring these lesions are some of the most difficult to treat. Although covalent small molecules against KRASG12C have shown promising efficacy against lung cancers, traditional barriers remain for drugging the more prevalent KRASG12D and KRASG12V mutants. Targeted degradation has emerged as an attractive alternative approach, but for KRAS, identification of the required high-affinity ligands continues to be a challenge. Another significant hurdle is the discovery of a hybrid molecule that appends an E3 ligase-recruiting moiety in a manner that satisfies the precise geometries required for productive polyubiquitin transfer while maintaining favorable druglike properties. To gain insights into the advantages and feasibility of KRAS targeted degradation, we applied a protein-based degrader (biodegrader) approach. This workflow centers on the intracellular expression of a chimeric protein consisting of a high-affinity target-binding domain fused to an engineered E3 ligase adapter. A series of anti-RAS biodegraders spanning different RAS isoform/nucleotide-state specificities and leveraging different E3 ligases provided definitive evidence for RAS degradability. Further, these established that the functional consequences of KRAS degradation are context dependent. Of broader significance, using the exquisite degradation specificity that biodegraders can possess, we demonstrated how this technology can be applied to answer questions that other approaches cannot. Specifically, application of the GDP-state specific degrader uncovered the relative prevalence of the "off-state" of WT and various KRAS mutants in the cellular context. Finally, if delivery challenges can be addressed, anti-RAS biodegraders will be exciting candidates for clinical development.

3.
Chem Sci ; 12(48): 15975-15987, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-35024121

ABSTRACT

Macrocyclic peptides have the potential to address intracellular protein-protein interactions (PPIs) of high value therapeutic targets that have proven largely intractable to small molecules. Here, we report broadly applicable lessons for applying this modality to intracellular targets and specifically for advancing chemical matter to address KRAS, a protein that represents the most common oncogene in human lung, colorectal and pancreatic cancers yet is one of the most challenging targets in human disease. Specifically, we focused on KRpep-2d, an arginine-rich KRAS-binding peptide with a disulfide-mediated macrocyclic linkage and a protease-sensitive backbone. These latter redox and proteolytic labilities obviated cellular activity. Extensive structure-activity relationship studies involving macrocyclic linker replacement, stereochemical inversion, and backbone α-methylation, gave a peptide with on-target cellular activity. However, we uncovered an important generic insight - the arginine-dependent cell entry mechanism limited its therapeutic potential. In particular, we observed a strong correlation between net positive charge and histamine release in an ex vivo assay, thus making this series unsuitable for advancement due to the potentially fatal consequences of mast cell degranulation. This observation should signal to researchers that cationic-mediated cell entry - an approach that has yet to succeed in the clinic despite a long history of attempts - carries significant therapy-limiting safety liabilities. Nonetheless, the cell-active molecules identified here validate a unique inhibitory epitope on KRAS and thus provide valuable molecular templates for the development of therapeutics that are desperately needed to address KRAS-driven cancers - some of the most treatment-resistant human malignancies.

4.
ACS Med Chem Lett ; 11(10): 1993-2001, 2020 Oct 08.
Article in English | MEDLINE | ID: mdl-33062184

ABSTRACT

Nonspecific promiscuous compounds can mislead researchers and waste significant resources. This phenomenon, though well-documented for small molecules, has not been widely explored for the peptide modality. Here we demonstrate that two purported peptide-based KRas inhibitors, SAH-SOS1 A and cyclorasin 9A5, exemplify false-positive molecules-in terms of both their binding affinities and cellular activities. Through multiple gold-standard biophysical techniques, we unambiguously show that both peptides lack specific binding to KRas and instead induce protein unfolding. Although these peptides inhibited cellular proliferation, the activities appeared to be off-target on the basis of a counterscreen with KRas-independent cell lines. We further demonstrate that their cellular activities are derived from membrane disruption. Accordingly, we propose that to de-risk false-positive molecules, orthogonal binding assays and cellular counterscreens are indispensable.

5.
Proc Natl Acad Sci U S A ; 116(31): 15514-15523, 2019 07 30.
Article in English | MEDLINE | ID: mdl-31296562

ABSTRACT

The most frequent extracellular signal-regulated kinase 2 (ERK2) mutation occurring in cancers is E322K (E-K). ERK2 E-K reverses a buried charge in the ERK2 common docking (CD) site, a region that binds activators, inhibitors, and substrates. Little is known about the cellular consequences associated with this mutation, other than apparent increases in tumor resistance to pathway inhibitors. ERK2 E-K, like the mutation of the preceding aspartate (ERK2 D321N [D-N]) known as the sevenmaker mutation, causes increased activity in cells and evades inactivation by dual-specificity phosphatases. As opposed to findings in cancer cells, in developmental assays in Drosophila, only ERK2 D-N displays a significant gain of function, revealing mutation-specific phenotypes. The crystal structure of ERK2 D-N is indistinguishable from that of wild-type protein, yet this mutant displays increased thermal stability. In contrast, the crystal structure of ERK2 E-K reveals profound structural changes, including disorder in the CD site and exposure of the activation loop phosphorylation sites, which likely account for the decreased thermal stability of the protein. These contiguous mutations in the CD site of ERK2 are both required for docking interactions but lead to unpredictably different functional outcomes. Our results suggest that the CD site is in an energetically strained configuration, and this helps drive conformational changes at distal sites on ERK2 during docking interactions.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Mutation/genetics , Animals , Animals, Genetically Modified , Crystallography, X-Ray , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Enzyme Activation , Enzyme Stability , Extracellular Signal-Regulated MAP Kinases/chemistry , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Models, Molecular , Mutant Proteins/metabolism
6.
Molecules ; 24(12)2019 Jun 20.
Article in English | MEDLINE | ID: mdl-31226791

ABSTRACT

Stapled α-helical peptides represent an emerging superclass of macrocyclic molecules with drug-like properties, including high-affinity target binding, protease resistance, and membrane permeability. As a model system for probing the chemical space available for optimizing these properties, we focused on dual Mdm2/MdmX antagonist stapled peptides related to the p53 N-terminus. Specifically, we first generated a library of ATSP-7041 (Chang et al., 2013) analogs iteratively modified by L-Ala and D-amino acids. Single L-Ala substitutions beyond the Mdm2/(X) binding interfacial residues (i.e., Phe3, Trp7, and Cba10) had minimal effects on target binding, α-helical content, and cellular activity. Similar binding affinities and cellular activities were noted at non-interfacial positions when the template residues were substituted with their d-amino acid counterparts, despite the fact that d-amino acid residues typically 'break' right-handed α-helices. d-amino acid substitutions at the interfacial residues Phe3 and Cba10 resulted in the expected decreases in binding affinity and cellular activity. Surprisingly, substitution at the remaining interfacial position with its d-amino acid equivalent (i.e., Trp7 to d-Trp7) was fully tolerated, both in terms of its binding affinity and cellular activity. An X-ray structure of the d-Trp7-modified peptide was determined and revealed that the indole side chain was able to interact optimally with its Mdm2 binding site by a slight global re-orientation of the stapled peptide. To further investigate the comparative effects of d-amino acid substitutions we used linear analogs of ATSP-7041, where we replaced the stapling amino acids by Aib (i.e., R84 to Aib4 and S511 to Aib11) to retain the helix-inducing properties of α-methylation. The resultant analog sequence Ac-Leu-Thr-Phe-Aib-Glu-Tyr-Trp-Gln-Leu-Cba-Aib-Ser-Ala-Ala-NH2 exhibited high-affinity target binding (Mdm2 Kd = 43 nM) and significant α-helicity in circular dichroism studies. Relative to this linear ATSP-7041 analog, several d-amino acid substitutions at Mdm2(X) non-binding residues (e.g., d-Glu5, d-Gln8, and d-Leu9) demonstrated decreased binding and α-helicity. Importantly, circular dichroism (CD) spectroscopy showed that although helicity was indeed disrupted by d-amino acids in linear versions of our template sequence, stapled molecules tolerated these residues well. Further studies on stapled peptides incorporating N-methylated amino acids, l-Pro, or Gly substitutions showed that despite some positional dependence, these helix-breaking residues were also generally tolerated in terms of secondary structure, binding affinity, and cellular activity. Overall, macrocyclization by hydrocarbon stapling appears to overcome the destabilization of α-helicity by helix breaking residues and, in the specific case of d-Trp7-modification, a highly potent ATSP-7041 analog (Mdm2 Kd = 30 nM; cellular EC50 = 600 nM) was identified. Our findings provide incentive for future studies to expand the chemical diversity of macrocyclic α-helical peptides (e.g., d-amino acid modifications) to explore their biophysical properties and cellular permeability. Indeed, using the library of 50 peptides generated in this study, a good correlation between cellular permeability and lipophilicity was observed.


Subject(s)
Amino Acids/chemistry , Cell-Penetrating Peptides/chemistry , Peptide Fragments/chemistry , Protein Conformation , Amino Acid Sequence/genetics , Amino Acid Substitution/genetics , Amino Acids/chemical synthesis , Cell-Penetrating Peptides/chemical synthesis , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/pharmacology , Circular Dichroism , Dipeptides/chemistry , Humans , Oligopeptides/chemistry , Peptides, Cyclic/pharmacology , Permeability/drug effects , Protein Structure, Secondary , Proto-Oncogene Proteins c-mdm2/chemistry , Proto-Oncogene Proteins c-mdm2/genetics
7.
Structure ; 26(8): 1101-1115.e6, 2018 08 07.
Article in English | MEDLINE | ID: mdl-29983373

ABSTRACT

The human NDR family kinases control diverse aspects of cell growth, and are regulated through phosphorylation and association with scaffolds such as MOB1. Here, we report the crystal structure of the human NDR1 kinase domain in its non-phosphorylated state, revealing a fully resolved atypically long activation segment that blocks substrate binding and stabilizes a non-productive position of helix αC. Consistent with an auto-inhibitory function, mutations within the activation segment of NDR1 dramatically enhance in vitro kinase activity. Interestingly, NDR1 catalytic activity is further potentiated by MOB1 binding, suggesting that regulation through modulation of the activation segment and by MOB1 binding are mechanistically distinct. Lastly, deleting the auto-inhibitory activation segment of NDR1 causes a marked increase in the association with upstream Hippo pathway components and the Furry scaffold. These findings provide a point of departure for future efforts to explore the cellular functions and the mechanism of NDR1.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Epithelial Cells/enzymology , Hepatocyte Growth Factor/chemistry , Microtubule-Associated Proteins/chemistry , Protein Serine-Threonine Kinases/chemistry , Proto-Oncogene Proteins/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Binding Sites , Cell Cycle Proteins , Cell Line, Tumor , Cloning, Molecular , Crystallography, X-Ray , Epithelial Cells/cytology , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Humans , Kinetics , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Models, Molecular , Mutation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Serine-Threonine Kinase 3 , Signal Transduction , Substrate Specificity
8.
SLAS Discov ; 23(9): 974-981, 2018 10.
Article in English | MEDLINE | ID: mdl-29884090

ABSTRACT

Histidine decarboxylase (HDC) is the primary enzyme that catalyzes the conversion of histidine to histamine. HDC contributes to many physiological responses as histamine plays important roles in allergic reaction, neurological response, gastric acid secretion, and cell proliferation and differentiation. Small-molecule modulation of HDC represents a potential therapeutic strategy for a range of histamine-associated diseases, including inflammatory disease, neurological disorders, gastric ulcers, and select cancers. High-throughput screening (HTS) methods for measuring HDC activity are currently limited. Here, we report the development of a time-resolved fluorescence resonance energy transfer (TR-FRET) assay for monitoring HDC activity. The assay is based on competition between HDC-generated histamine and fluorophore-labeled histamine for binding to a Europium cryptate (EuK)-labeled anti-histamine antibody. We demonstrated that the assay is highly sensitive and simple to develop. Assay validation experiments were performed using low-volume 384-well plates and resulted in good statistical parameters. A pilot HTS screen gave a Z' score > 0.5 and a hit rate of 1.1%, and led to the identification of a validated hit series. Overall, the presented assay should facilitate the discovery of therapeutic HDC inhibitors by acting as a novel tool suitable for large-scale HTS and subsequent interrogation of compound structure-activity relationships.


Subject(s)
High-Throughput Screening Assays , Histamine Antagonists/pharmacology , Histamine/metabolism , Histidine Decarboxylase/metabolism , Dose-Response Relationship, Drug , Enzyme Assays , Fluorescence Resonance Energy Transfer/methods , Histamine Antagonists/chemistry , Humans , Reproducibility of Results , Structure-Activity Relationship
9.
Bioorg Med Chem ; 26(10): 2807-2815, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29598901

ABSTRACT

Macrocyclic α-helical peptides have emerged as a compelling new therapeutic modality to tackle targets confined to the intracellular compartment. Within the scope of hydrocarbon-stapling there has been significant progress to date, including the first stapled α-helical peptide to enter into clinical trials. The principal design concept of stapled α-helical peptides is to mimic a cognate (protein) ligand relative to binding its target via an α-helical interface. However, it was the proclivity of such stapled α-helical peptides to exhibit cell permeability and proteolytic stability that underscored their promise as unique macrocyclic peptide drugs for intracellular targets. This perspective highlights key learnings as well as challenges in basic research with respect to structure-based design, innovative chemistry, cell permeability and proteolytic stability that are essential to fulfill the promise of stapled α-helical peptide drug development.


Subject(s)
Drug Discovery/methods , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacology , Peptides/chemistry , Peptides/pharmacology , Animals , Humans , Macrocyclic Compounds/pharmacokinetics , Models, Molecular , Peptides/pharmacokinetics , Protein Conformation, alpha-Helical
10.
Nat Biotechnol ; 36(1): 95-102, 2018 01.
Article in English | MEDLINE | ID: mdl-29176614

ABSTRACT

Programmable nucleases, such as Cas9, are used for precise genome editing by homology-dependent repair (HDR). However, HDR efficiency is constrained by competition from other double-strand break (DSB) repair pathways, including non-homologous end-joining (NHEJ). We report the discovery of a genetically encoded inhibitor of 53BP1 that increases the efficiency of HDR-dependent genome editing in human and mouse cells. 53BP1 is a key regulator of DSB repair pathway choice in eukaryotic cells and functions to favor NHEJ over HDR by suppressing end resection, which is the rate-limiting step in the initiation of HDR. We screened an existing combinatorial library of engineered ubiquitin variants for inhibitors of 53BP1. Expression of one variant, named i53 (inhibitor of 53BP1), in human and mouse cells, blocked accumulation of 53BP1 at sites of DNA damage and improved gene targeting and chromosomal gene conversion with either double-stranded DNA or single-stranded oligonucleotide donors by up to 5.6-fold. Inhibition of 53BP1 is a robust method to increase efficiency of HDR-based precise genome editing.


Subject(s)
CRISPR-Cas Systems/genetics , Gene Editing , Tumor Suppressor p53-Binding Protein 1/genetics , Animals , DNA Damage/genetics , DNA End-Joining Repair/genetics , DNA Repair/genetics , Gene Expression Regulation/genetics , Humans , Mice , Recombinational DNA Repair/genetics , Tumor Suppressor p53-Binding Protein 1/antagonists & inhibitors
11.
Biochemistry ; 54(32): 5063-71, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26208601

ABSTRACT

The related protein kinases SPAK and OSR1 regulate ion homeostasis in part by phosphorylating cation cotransporter family members. The structure of the kinase domain of OSR1 was determined in the unphosphorylated inactive form and, like some other Ste20 kinases, exhibited a domain-swapped activation loop. To further probe the role of domain swapping in SPAK and OSR1, we have determined the crystal structures of SPAK 63-403 at 3.1 Å and SPAK 63-390 T243D at 2.5 Å resolution. These structures encompass the kinase domain and different portions of the C-terminal tail, the longer without and the shorter with an activating T243D point mutation. The structure of the T243D protein reveals significant conformational differences relative to unphosphorylated SPAK and OSR1 but also has some features of an inactive kinase. Both structures are domain-swapped dimers. Sequences involved in domain swapping were identified and mutated to create a SPAK monomeric mutant with kinase activity, indicating that monomeric forms are active. The monomeric mutant is activated by WNK1 but has reduced activity toward its substrate NKCC2, suggesting regulatory roles for domain swapping. The structure of partially active SPAK T243D is consistent with a multistage activation process in which phosphorylation induces a SPAK conformation that requires further remodeling to build the active structure.


Subject(s)
Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Crystallography, X-Ray , Enzyme Activation , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphorylation , Protein Conformation , Protein Interaction Domains and Motifs , Protein Serine-Threonine Kinases/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
12.
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
13.
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
14.
Science ; 339(6119): 590-5, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23287719

ABSTRACT

The ubiquitin system regulates virtually all aspects of cellular function. We report a method to target the myriad enzymes that govern ubiquitination of protein substrates. We used massively diverse combinatorial libraries of ubiquitin variants to develop inhibitors of four deubiquitinases (DUBs) and analyzed the DUB-inhibitor complexes with crystallography. We extended the selection strategy to the ubiquitin conjugating (E2) and ubiquitin ligase (E3) enzymes and found that ubiquitin variants can also enhance enzyme activity. Last, we showed that ubiquitin variants can bind selectively to ubiquitin-binding domains. Ubiquitin variants exhibit selective function in cells and thus enable orthogonal modulation of specific enzymatic steps in the ubiquitin system.


Subject(s)
Combinatorial Chemistry Techniques , Endopeptidases/metabolism , Protease Inhibitors/isolation & purification , Ubiquitin Thiolesterase/metabolism , Ubiquitin/metabolism , Ubiquitination/drug effects , Amino Acid Sequence , Conserved Sequence , Drug Design , Endopeptidases/chemistry , HEK293 Cells , Humans , Molecular Sequence Data , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Protein Conformation , Protein Structure, Secondary , Small Molecule Libraries , Ubiquitin/chemistry , Ubiquitin/genetics , Ubiquitin Thiolesterase/chemistry , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
15.
Proc Natl Acad Sci U S A ; 109(42): 16841-6, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23027929

ABSTRACT

We report an action of the protein kinase WNK3 on the neuronal mRNA splicing factor Fox-1. Fox-1 splices mRNAs encoding proteins important in synaptic transmission and membrane excitation. WNK3, implicated in the control of neuronal excitability through actions on ion transport, binds Fox-1 and inhibits its splicing activity in a kinase activity-dependent manner. Phosphorylation of Fox-1 by WNK3 does not change its RNA binding capacity; instead, WNK3 increases the cytoplasmic localization of Fox-1, thereby suppressing Fox-1-dependent splicing. These findings demonstrate a role of WNK3 in RNA processing. Considering the implication of WNK3 and Fox-1 in disorders of neuronal development such as autism, WNK3 may offer a target for treatment of Fox-1-induced disease.


Subject(s)
Brain/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA-Binding Proteins/metabolism , Animals , DNA Primers/genetics , Deuterium , Electrophoretic Mobility Shift Assay , Gene Library , HEK293 Cells , Humans , Immunoprecipitation , Mice , Phosphorus Radioisotopes , RNA Splicing Factors , Reverse Transcriptase Polymerase Chain Reaction , Two-Hybrid System Techniques
16.
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
17.
Nature ; 466(7309): 941-6, 2010 Aug 19.
Article in English | MEDLINE | ID: mdl-20725033

ABSTRACT

DNA double-strand breaks (DSBs) pose a potent threat to genome integrity. These lesions also contribute to the efficacy of radiotherapy and many cancer chemotherapeutics. DSBs elicit a signalling cascade that modifies the chromatin surrounding the break, first by ATM-dependent phosphorylation and then by RNF8-, RNF168- and BRCA1-dependent regulatory ubiquitination. Here we report that OTUB1, a deubiquitinating enzyme, is an inhibitor of DSB-induced chromatin ubiquitination. Surprisingly, we found that OTUB1 suppresses RNF168-dependent poly-ubiquitination independently of its catalytic activity. OTUB1 does so by binding to and inhibiting UBC13 (also known as UBE2N), the cognate E2 enzyme for RNF168. This unusual mode of regulation is unlikely to be limited to UBC13 because analysis of OTUB1-associated proteins revealed that OTUB1 binds to E2s of the UBE2D and UBE2E subfamilies. Finally, OTUB1 depletion mitigates the DSB repair defect associated with defective ATM signalling, indicating that pharmacological targeting of the OTUB1-UBC13 interaction might enhance the DNA damage response.


Subject(s)
Chromatin/metabolism , Cysteine Endopeptidases/metabolism , DNA Breaks, Double-Stranded , Ubiquitination/physiology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Cell Line , Cell Line, Tumor , Chromatin/chemistry , Cysteine Endopeptidases/deficiency , Cysteine Endopeptidases/genetics , DNA Repair/physiology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Deubiquitinating Enzymes , Humans , Protein Binding , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/antagonists & inhibitors , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
18.
J Biol Chem ; 285(33): 25161-7, 2010 Aug 13.
Article in English | MEDLINE | ID: mdl-20525693

ABSTRACT

The four WNK (with no lysine (K)) protein kinases affect ion balance and contain an unusual protein kinase domain due to the unique placement of the active site lysine. Mutations in two WNKs cause a heritable form of ion imbalance culminating in hypertension. WNK1 activates the serum- and glucocorticoid-induced protein kinase SGK1; the mechanism is noncatalytic. SGK1 increases membrane expression of the epithelial sodium channel (ENaC) and sodium reabsorption via phosphorylation and sequestering of the E3 ubiquitin ligase neural precursor cell expressed, developmentally down-regulated 4-2 (Nedd4-2), which otherwise promotes ENaC endocytosis. Questions remain about the intrinsic abilities of WNK family members to regulate this pathway. We find that expression of the N termini of all four WNKs results in modest to strong activation of SGK1. In reconstitution experiments in the same cell line all four WNKs also increase sodium current blocked by the ENaC inhibitor amiloride. The N termini of the WNKs also have the capacity to interact with SGK1. More detailed analysis of activation by WNK4 suggests mechanisms in common with WNK1. Further evidence for the importance of WNK1 in this process comes from the ability of Nedd4-2 to bind to WNK1 and the finding that endogenous SGK1 has reduced activity if WNK1 is knocked down by small interfering RNA.


Subject(s)
Epithelial Sodium Channels/metabolism , Immediate-Early Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , CHO Cells , Cell Line , Cell Line, Tumor , Cricetinae , Cricetulus , Endosomal Sorting Complexes Required for Transport/genetics , Endosomal Sorting Complexes Required for Transport/metabolism , Epithelial Sodium Channels/genetics , HeLa Cells , Humans , Immediate-Early Proteins/genetics , Immunoblotting , Immunoprecipitation , Mice , Minor Histocompatibility Antigens , Nedd4 Ubiquitin Protein Ligases , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Rats , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , WNK Lysine-Deficient Protein Kinase 1
19.
J Biol Chem ; 284(43): 29437-45, 2009 Oct 23.
Article in English | MEDLINE | ID: mdl-19684019

ABSTRACT

Initially identified in Chlamydomonas, RSP3 (radial spoke protein 3) is 1 of more than 20 identified radial spoke structural components of motile cilia and is required for axonemal sliding and flagellar motility. The mammalian orthologs for this and other radial spoke proteins, however, remain to be characterized. We found mammalian RSP3 to bind to the MAPK ERK2 through a yeast two-hybrid screen designed to identify interacting proteins that have a higher affinity for the phosphorylated, active form of the protein kinase. Consistent with the screening result, the human homolog, RSPH3, interacts with and is a substrate for ERK1/2. Moreover, RSPH3 is a protein kinase A-anchoring protein (AKAP) that scaffolds the cAMP-dependent protein kinase holoenzyme. The binding of RSPH3 to the regulatory subunits of cAMP-dependent protein kinase, RIIalpha and RIIbeta, is regulated by ERK1/2 activity and phosphorylation. Here we describe an ERK1/2-interacting AKAP and suggest a mechanism by which cAMP-dependent protein kinase-AKAP binding can be modulated by the activity of other enzymes.


Subject(s)
A Kinase Anchor Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Nerve Tissue Proteins/metabolism , A Kinase Anchor Proteins/genetics , Animals , Cyclic AMP-Dependent Protein Kinase RIIalpha Subunit/genetics , Cyclic AMP-Dependent Protein Kinase RIIalpha Subunit/metabolism , Humans , Mitogen-Activated Protein Kinase 1/genetics , Nerve Tissue Proteins/genetics , Protein Binding , Two-Hybrid System Techniques
20.
J Biol Chem ; 284(6): 3453-60, 2009 Feb 06.
Article in English | MEDLINE | ID: mdl-19068477

ABSTRACT

SGK1 (serum- and glucocorticoid-induced kinase 1) is a member of the AGC branch of the protein kinase family. Among well described functions of SGK1 is the regulation of epithelial transport through phosphorylation of the ubiquitin protein ligase Nedd4-2 (neuronal precursor cell expressed developmentally down-regulated 4-2). The activation of SGK1 has been widely accepted to be dependent on the phosphorylation of Thr256 in the activation loop and Ser422 in the hydrophobic motif near the C terminus. Here, we report the identification of two additional phosphorylation sites, Ser397 and Ser401. Both are required for maximum SGK1 activity induced by extracellular agents or by coexpression with other protein kinases, with the largest loss of activity from mutation of Ser397. Coexpression with active Akt1 increased the phosphorylation of Ser397 and thereby SGK1 kinase activity. SGK1 activation was further augmented by coexpression with the protein kinase WNK1 (with no lysine kinase 1). These findings reveal further complexity underlying the regulation of SGK1 activity.


Subject(s)
Immediate-Early Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Amino Acid Motifs/physiology , Endosomal Sorting Complexes Required for Transport , Enzyme Activation/physiology , HeLa Cells , Humans , Immediate-Early Proteins/genetics , Intracellular Signaling Peptides and Proteins , Minor Histocompatibility Antigens , Mutation , Nedd4 Ubiquitin Protein Ligases , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Protein Transport/physiology , Proto-Oncogene Proteins c-akt/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , WNK Lysine-Deficient Protein Kinase 1
SELECTION OF CITATIONS
SEARCH DETAIL
...