Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Biochem Biophys Res Commun ; 471(4): 409-15, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26902118

ABSTRACT

The emergence of resistant Mycobacterium tuberculosis (Mtb) infection and the dearth of drugs against tuberculosis have made it imperative to identify and validate novel targets and classes of drugs for treatment. The pyrimidine operon regulatory protein (PyrR), a regulator of de novo pyrimidine synthesis, is an essential enzyme and a probable 5-fluorouracil (5-FU) target in Mtb, with mutations in PyrR attributable to 5-FU resistance. Here we report, for the first time, the co-crystal structure of the PyrR-5-FU complex along with mapping of spontaneous mutational sites of PyrR. A cluster of mutations in the presence of the drug usually indicates a plausible region of drug-target interaction. Notably, we observed that three of the mutated PyrR residues lie in close proximity to the 5-FU binding site, including the amino acid Val178, which is involved in water mediated hydrogen bonding contact with 5-FU. Computational modeling of the PyrR-5'-phosphoribosyl-α-1'-pyrophosphate (PRPP) complex revealed the location of several other mutations at the PRPP binding site of PyrR, indicating their probable role in resistance. Indeed, 5-FU-resistant strains harboring these mutations exhibited decreased susceptibility to 5-FU. Considering that pyrimidine analogs are predominantly regarded to inhibit PyrR, the present studies will be beneficial for the screening of appropriate inhibitors of PyrR and help provide insight into future TB drug design and development.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Fluorouracil/chemistry , Mutation , Pentosyltransferases/chemistry , Pentosyltransferases/genetics , Repressor Proteins/chemistry , Repressor Proteins/genetics , Amino Acid Sequence , Antitubercular Agents/pharmacology , Bacterial Proteins/metabolism , Binding Sites , Crystallography, X-Ray , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Fluorouracil/pharmacology , Microbial Sensitivity Tests , Models, Molecular , Molecular Sequence Data , Mycobacterium tuberculosis/enzymology , Pentosyltransferases/metabolism , Polymorphism, Genetic , Protein Conformation , Repressor Proteins/metabolism
2.
J Biol Chem ; 289(37): 25611-23, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25074933

ABSTRACT

Hakai, an E3 ubiquitin ligase, disrupts cell-cell contacts in epithelial cells and is up-regulated in human colon and gastric adenocarcinomas. Hakai acts through its phosphotyrosine-binding (HYB) domain, which bears a dimeric fold that recognizes the phosphotyrosine motifs of E-cadherin, cortactin, DOK1, and other Src substrates. Unlike the monomeric nature of the SH2 and phosphotyrosine-binding domains, the architecture of the HYB domain consists of an atypical, zinc-coordinated tight homodimer. Here, we report a C-terminal truncation mutant of the HYB domain (HYB(ΔC)), comprising amino acids 106-194, which exists as a monomer in solution. The NMR structure revealed that this deletion mutant undergoes a dramatic structural change caused by a rearrangement of the atypical zinc-coordinated unit in the C terminus of the HYB domain to a C2H2-like zinc finger in HYB(ΔC). Moreover, using isothermal titration calorimetry, we show that dimerization of HYB(ΔC) can be induced using a phosphotyrosine substrate peptide. This ligand-induced dimerization of HYB(ΔC) is further validated using analytical ultracentrifugation, size-exclusion chromatography, NMR relaxation studies, dynamic light scattering, and circular dichroism experiments. Overall, these observations suggest that the dimeric architecture of the HYB domain is essential for the phosphotyrosine-binding property of Hakai.


Subject(s)
Cadherins/chemistry , Epithelial Cells/chemistry , Ubiquitin-Protein Ligases/chemistry , src Homology Domains , Binding Sites , Cadherins/metabolism , Crystallography, X-Ray , Epithelial Cells/metabolism , Humans , Ligands , Magnetic Resonance Spectroscopy , Phosphotyrosine/chemistry , Protein Binding , Protein Conformation , Solutions/chemistry , Ubiquitin-Protein Ligases/metabolism
3.
Biochem Biophys Res Commun ; 467(3): 577-82, 2015 Nov 20.
Article in English | MEDLINE | ID: mdl-26456658

ABSTRACT

Tuberculosis (TB) remains the second leading cause of death from an infectious disease globally, despite the incessant efforts to control it. Research and development into new TB medicines is imperative for effective TB control; however, new strategies for the rational use of existing drugs, such as through the identification of new drug targets, could also significantly enhance this process. Key enzymes involved in the essential metabolic and regulatory pathways are usually sought in the pursuit of potential drug targets. Uracil phosphoribosyltransferase (UPRT) is a key salvage pathway enzyme in the synthesis of uridine 5'-monophosphate (UMP) and a probable target of 5-fluorouracil (5-FU) in Mycobacterium tuberculosis (Mtb). To date, there is no structure available for UPRT from Mtb (MtUPRT) that would assist in the identification of appropriate inhibitors for the enzyme. Here we report the structure of MtUPRT along with its spontaneous mutational studies in the presence of 5-FU. We further mapped these four single nucleotide polymorphisms (SNPs) onto the MtUPRT structure, with two residues found to be conserved among the MtUPRT homologs. Notably, none of these SNPs are located in the 5-FU binding pocket. However, the mutants harboring these mutations showed increased MICs (minimum inhibitory concentration) as compared to wild type strains. The present study will aid in the screening of inhibitors of MtUPRT and thus assist in TB drug design and development.


Subject(s)
Fluorouracil/pharmacology , Mutation , Mycobacterium tuberculosis/enzymology , Pentosyltransferases/metabolism , Amino Acid Sequence , Molecular Sequence Data , Mycobacterium tuberculosis/genetics , Pentosyltransferases/chemistry , Pentosyltransferases/genetics , Sequence Homology, Amino Acid
4.
FEBS J ; 290(9): 2437-2448, 2023 05.
Article in English | MEDLINE | ID: mdl-36259273

ABSTRACT

Glutaminase catalyses the metabolic process called glutaminolysis. Cancer cells harness glutaminolysis to increase energy reserves under stressful conditions for rapid proliferation. Glutaminases are upregulated in many tumours. In humans, the kidney-type glutaminase (KGA) isoform is highly expressed in the kidney, brain, intestine, foetal liver, lymphocytes and in many tumours. Glutaminase inhibition is shown to be effective in controlling cancers. Previously, we and others reported the inhibition mechanism of KGA using various inhibitors that target the active and allosteric sites of the enzyme. Here, we report the identification of a novel allosteric site in KGA using the compound DDP through its complex crystal structure combined with mutational and hydrogen-deuterium exchange mass spectrometry studies. This allosteric site is located at the dimer interface, situated ~ 31 Å away from the previously identified allosteric site and ~ 32 Å away from the active site. Remarkably, the mechanism of inhibition is conserved, irrespective of which allosteric pocket is targeted, causing the same conformational changes in the key loop near the active site (Glu312-Pro329) and subsequent enzyme inactivation. Contrary to the previously identified allosteric site, the identified new allosteric site is primarily hydrophilic. This site could be effectively targeted for the synthesis of specific and potent water-soluble inhibitors of glutaminase, which will lead to the development of anticancer drugs.


Subject(s)
Antineoplastic Agents , Glutaminase , Humans , Allosteric Site , Glutaminase/genetics , Glutaminase/metabolism , Kidney/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Mutation
5.
Methods Mol Biol ; 2706: 149-165, 2023.
Article in English | MEDLINE | ID: mdl-37558947

ABSTRACT

Cellular thermal shift assay (CETSA) is based on the thermal stabilization of the protein target by a compound binding. Thus, CETSA can be used to measure a compound's cellular target engagement and permeability. HiBiT CETSA method is quantitative and has higher throughput compared to the traditional Western-based CETSA. Here, we describe the protocol for a HiBiT CETSA, which utilizes a HiBiT tag derived from the NanoLuciferase (NanoLuc) that upon complementation by LgBiT NanoLuc tag produces a bright signal enabling tracking of the effects of increasing temperature on the stability of a protein-of-interest in the presence/absence of various compounds. Exposure of a HiBiT-tagged protein to increasing temperatures induces protein denaturation and thus decreased LgBiT complementation and NanoLuc signal. As the stability of proteins at higher temperatures can be influenced by the compound binding, this method enables screening for target engagement in living or permeabilized cells.


Subject(s)
Hot Temperature , Proteins , Temperature
6.
Nat Commun ; 14(1): 6345, 2023 10 10.
Article in English | MEDLINE | ID: mdl-37816714

ABSTRACT

The Src homology 2 (SH2) domain recognizes phosphotyrosine (pY) post translational modifications in partner proteins to trigger downstream signaling. Drug discovery efforts targeting the SH2 domains have long been stymied by the poor drug-like properties of phosphate and its mimetics. Here, we use structure-based design to target the SH2 domain of the E3 ligase suppressor of cytokine signaling 2 (SOCS2). Starting from the highly ligand-efficient pY amino acid, a fragment growing approach reveals covalent modification of Cys111 in a co-crystal structure, which we leverage to rationally design a cysteine-directed electrophilic covalent inhibitor MN551. We report the prodrug MN714 containing a pivaloyloxymethyl (POM) protecting group and evidence its cell permeability and capping group unmasking using cellular target engagement and in-cell 19F NMR spectroscopy. Covalent engagement at Cys111 competitively blocks recruitment of cellular SOCS2 protein to its native substrate. The qualified inhibitors of SOCS2 could find attractive applications as chemical probes to understand the biology of SOCS2 and its CRL5 complex, and as E3 ligase handles in proteolysis targeting chimera (PROTACs) to induce targeted protein degradation.


Subject(s)
Proteins , Ubiquitin-Protein Ligases , Ubiquitin-Protein Ligases/metabolism , Phosphotyrosine , Ligands , src Homology Domains
7.
Curr Opin Struct Biol ; 67: 110-119, 2021 04.
Article in English | MEDLINE | ID: mdl-33271439

ABSTRACT

E3 ubiquitin ligase machineries are emerging as attractive therapeutic targets because they confer specificity to substrate ubiquitination and can be hijacked for targeted protein degradation. In this review, we bring to focus our current structural understanding of E3 ligase complexes, in particular the multi-subunit cullin RING ligases, and modulation thereof by small-molecule glues and PROTAC degraders. We highlight recent advances in elucidating the modular assembly of E3 ligase machineries, their diverse substrate and degron recognition mechanisms, and how these structural features impact on ligase function. We then outline the emergence of structures of E3 ligases bound to neo-substrates and degrader molecules, and highlight the importance of studying such ternary complexes for structure-based degrader design.


Subject(s)
Ubiquitin-Protein Ligases , Proteolysis , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
8.
Nat Commun ; 10(1): 2534, 2019 06 10.
Article in English | MEDLINE | ID: mdl-31182716

ABSTRACT

The suppressor of cytokine signaling 2 (SOCS2) acts as substrate recognition subunit of a Cullin5 E3 ubiquitin ligase complex. SOCS2 binds to phosphotyrosine-modified epitopes as degrons for ubiquitination and proteasomal degradation, yet the molecular basis of substrate recognition has remained elusive. Here, we report co-crystal structures of SOCS2-ElonginB-ElonginC in complex with phosphorylated peptides from substrates growth hormone receptor (GHR-pY595) and erythropoietin receptor (EpoR-pY426) at 1.98 Å and 2.69 Å, respectively. Both peptides bind in an extended conformation recapitulating the canonical SH2 domain-pY pose, but capture different conformations of the EF loop via specific hydrophobic interactions. The flexible BG loop is fully defined in the electron density, and does not contact the substrate degron directly. Cancer-associated SNPs located around the pY pocket weaken substrate-binding affinity in biophysical assays. Our findings reveal insights into substrate recognition and specificity by SOCS2, and provide a blueprint for small molecule ligand design.


Subject(s)
Suppressor of Cytokine Signaling Proteins/chemistry , Ubiquitin-Protein Ligases/chemistry , Crystallography, X-Ray , Humans , Phosphotyrosine/chemistry , Polymorphism, Single Nucleotide , Protein Conformation , Receptors, Erythropoietin/chemistry , Receptors, Somatotropin/chemistry , Sequence Alignment , Substrate Specificity , Suppressor of Cytokine Signaling Proteins/genetics , Ubiquitination
9.
Oncotarget ; 7(36): 57943-57954, 2016 Sep 06.
Article in English | MEDLINE | ID: mdl-27462863

ABSTRACT

Cancer cells employ glutaminolysis to provide a source of intermediates for their upregulated biosynthetic needs. Glutaminase, which catalyzes the conversion of glutamine to glutamate, is gaining increasing attention as a potential drug target. Small-molecule inhibitors such as BPTES and CB-839, which target the allosteric site of glutaminase with high specificity, demonstrate immense promise as anti-tumor drugs. Here, we report the study of a new BPTES analog, N,N'-(5,5'-(trans-cyclohexane-1,3-diyl)bis(1,3,4-tiadiazole-5,2-diyl))bis(2-phenylacetamide) (trans-CBTBP), and compared its inhibitory effect against that of CB-839 and BPTES. We show that CB-839 has a 30- and 50-fold lower IC50 than trans-CBTBP and BPTES, respectively. To explore the structural basis for the differences in their inhibitory efficacy, we solved the complex structures of cKGA with 1S, 3S-CBTBP and CB-839. We found that CB-839 produces a greater degree of interaction with cKGA than 1S, 3S-CBTBP or BPTES. The results of this study will facilitate the rational design of new KGA inhibitors to better treat glutamine-addicted cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Glutaminase/antagonists & inhibitors , Glutaminase/chemistry , Kidney Neoplasms/enzymology , Kidney/enzymology , Sulfides/chemistry , Thiadiazoles/chemistry , Allosteric Site , Antineoplastic Agents/chemistry , Cell Proliferation , HEK293 Cells , Humans , Inhibitory Concentration 50 , Molecular Conformation , Protein Binding , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL