Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 299(12): 105482, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37992806

ABSTRACT

Bromodomains (BDs) regulate gene expression by recognizing protein motifs containing acetyllysine. Although originally characterized as histone-binding proteins, it has since become clear that these domains interact with other acetylated proteins, perhaps most prominently transcription factors. The likely transient nature and low stoichiometry of such modifications, however, has made it challenging to fully define the interactome of any given BD. To begin to address this knowledge gap in an unbiased manner, we carried out mRNA display screens against a BD-the N-terminal BD of BRD3-using peptide libraries that contained either one or two acetyllysine residues. We discovered peptides with very strong consensus sequences and with affinities that are significantly higher than typical BD-peptide interactions. X-ray crystal structures also revealed modes of binding that have not been seen with natural ligands. Intriguingly, however, our sequences are not found in the human proteome, perhaps suggesting that strong binders to BDs might have been selected against during evolution.


Subject(s)
Proteome , Transcription Factors , Humans , Proteome/metabolism , Transcription Factors/metabolism , Protein Domains , Amino Acid Motifs , Peptides/metabolism , Protein Binding , Acetylation
2.
J Med Chem ; 66(10): 6652-6681, 2023 05 25.
Article in English | MEDLINE | ID: mdl-37134237

ABSTRACT

Purine nucleoside phosphorylase (PNP) is a well-known molecular target with potential therapeutic applications in the treatment of T-cell malignancies and/or bacterial/parasitic infections. Here, we report the design, development of synthetic methodology, and biological evaluation of a series of 30 novel PNP inhibitors based on acyclic nucleoside phosphonates bearing a 9-deazahypoxanthine nucleobase. The strongest inhibitors exhibited IC50 values as low as 19 nM (human PNP) and 4 nM (Mycobacterium tuberculosis (Mt) PNP) and highly selective cytotoxicity toward various T-lymphoblastic cell lines with CC50 values as low as 9 nM. No cytotoxic effect was observed on other cancer cell lines (HeLa S3, HL60, HepG2) or primary PBMCs for up to 10 µM. We report the first example of the PNP inhibitor exhibiting over 60-fold selectivity for the pathogenic enzyme (MtPNP) over hPNP. The results are supported by a crystallographic study of eight enzyme-inhibitor complexes and by ADMET profiling in vitro and in vivo.


Subject(s)
Enzyme Inhibitors , Purine-Nucleoside Phosphorylase , Humans , Purine-Nucleoside Phosphorylase/metabolism , Enzyme Inhibitors/chemistry , Crystallography
3.
J Biol Chem ; 299(3): 102970, 2023 03.
Article in English | MEDLINE | ID: mdl-36736427

ABSTRACT

Fasciolosis is a worldwide parasitic disease of ruminants and an emerging human disease caused by the liver fluke Fasciola hepatica. The cystatin superfamily of cysteine protease inhibitors is composed of distinct families of intracellular stefins and secreted true cystatins. FhCyLS-2 from F. hepatica is an unusual member of the superfamily, where our sequence and 3D structure analyses in this study revealed that it combines characteristics of both families. The protein architecture demonstrates its relationship to stefins, but FhCyLS-2 also contains the secretion signal peptide and disulfide bridges typical of true cystatins. The secretion status was confirmed by detecting the presence of FhCyLS-2 in excretory/secretory products, supported by immunolocalization. Our high-resolution crystal structure of FhCyLS-2 showed a distinct disulfide bridging pattern and functional reactive center. We determined that FhCyLS-2 is a broad specificity inhibitor of cysteine cathepsins from both the host and F. hepatica, suggesting a dual role in the regulation of exogenous and endogenous proteolysis. Based on phylogenetic analysis that identified several FhCyLS-2 homologues in liver/intestinal foodborne flukes, we propose a new group within the cystatin superfamily called cystatin-like stefins.


Subject(s)
Cystatins , Fasciola hepatica , Animals , Amino Acid Sequence , Cystatins/genetics , Cystatins/chemistry , Disulfides , Fasciola hepatica/genetics , Phylogeny , Helminth Proteins/chemistry , Helminth Proteins/genetics
4.
Eur J Med Chem ; 225: 113798, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34482272

ABSTRACT

Some pathogens, including parasites of the genus Trypanosoma causing Human and Animal African Trypanosomiases, cannot synthesize purines de novo and they entirely rely on the purine salvage pathway (PSP) for their nucleotide generation. Thus, their PSP enzymes are considered as promising drug targets, sparsely explored so far. Recently, a significant role of acyclic nucleoside phosphonates (ANPs) as inhibitors of key enzymes of PSP, namely of 6-oxopurine phosphoribosyltransferases (PRTs), has been discovered. Herein, we designed and synthesized two series of new ANPs branched at the C1' position as mimics of adenosine monophosphate. The novel ANPs efficaciously inhibited Trypanosoma brucei adenine PRT (TbrAPRT1) activity in vitro and it was shown that the configuration on the C1' chiral centre strongly influenced their activity: the (R)-enantiomers proved to be more potent compared to the (S)-enantiomers. Two ANPs, with Ki values of 0.39 µM and 0.57 µM, represent the most potent TbrAPRT1 inhibitors reported to date and they are an important tool to further study purine metabolism in various parasites.


Subject(s)
Adenine Phosphoribosyltransferase/antagonists & inhibitors , Adenosine Monophosphate/pharmacology , Antiprotozoal Agents/pharmacology , Enzyme Inhibitors/pharmacology , Nucleosides/pharmacology , Trypanosoma brucei brucei/drug effects , Adenine Phosphoribosyltransferase/metabolism , Adenosine Monophosphate/chemical synthesis , Adenosine Monophosphate/chemistry , Antiprotozoal Agents/chemical synthesis , Antiprotozoal Agents/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Molecular Structure , Nucleosides/chemical synthesis , Nucleosides/chemistry , Parasitic Sensitivity Tests , Structure-Activity Relationship , Trypanosoma brucei brucei/enzymology
5.
J Biol Chem ; 296: 100797, 2021.
Article in English | MEDLINE | ID: mdl-34019879

ABSTRACT

Bacterial methionine biosynthesis can take place by either the trans-sulfurylation route or direct sulfurylation. The enzymes responsible for trans-sulfurylation have been characterized extensively because they occur in model organisms such as Escherichia coli. However, direct sulfurylation is actually the predominant route for methionine biosynthesis across the phylogenetic tree. In this pathway, most bacteria use an O-acetylhomoserine aminocarboxypropyltransferase (MetY) to catalyze the formation of homocysteine from O-acetylhomoserine and bisulfide. Despite the widespread distribution of MetY, this pyridoxal 5'-phosphate-dependent enzyme remains comparatively understudied. To address this knowledge gap, we have characterized the MetY from Thermotoga maritima (TmMetY). At its optimal temperature of 70 °C, TmMetY has a turnover number (apparent kcat = 900 s-1) that is 10- to 700-fold higher than the three other MetY enzymes for which data are available. We also present crystal structures of TmMetY in the internal aldimine form and, fortuitously, with a ß,γ-unsaturated ketimine reaction intermediate. This intermediate is identical to that found in the catalytic cycle of cystathionine γ-synthase (MetB), which is a homologous enzyme from the trans-sulfurylation pathway. By comparing the TmMetY and MetB structures, we have identified Arg270 as a critical determinant of specificity. It helps to wall off the active site of TmMetY, disfavoring the binding of the first MetB substrate, O-succinylhomoserine. It also ensures a strict specificity for bisulfide as the second substrate of MetY by occluding the larger MetB substrate, cysteine. Overall, this work illuminates the subtle structural mechanisms by which homologous pyridoxal 5'-phosphate-dependent enzymes can effect different catalytic, and therefore metabolic, outcomes.


Subject(s)
Bacterial Proteins/metabolism , Methionine/metabolism , Thermotoga maritima/metabolism , Bacterial Proteins/chemistry , Biosynthetic Pathways , Crystallography, X-Ray , Kinetics , Models, Molecular , Thermotoga maritima/chemistry
6.
Eur J Med Chem ; 216: 113309, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33711765

ABSTRACT

Pharmacological inhibition of cyclin-dependent kinases has emerged as a possible treatment option for various cancer types. We recently identified substituted imidazo[1,2-c]pyrimidin-5(6H)-ones as inhibitors of cyclin-dependent kinase 2 (CDK2). Here, we report the synthesis of derivatives modified at positions 2, 3, 6 or 8 prepared using Suzuki-Miyaura cross-coupling, halogenation, Dimroth-type rearrangement and alkylation as the main synthetic methods. The compounds displayed micro- to submicromolar inhibition of CDK2/cyclin E activity. Binding of the most potent compound 3b to CDK2 was determined using isothermal titration calorimetry. The co-crystal structure of 3b in complex with fully active CDK2 was solved, revealing the binding mode of 3b in the ATP pocket and a hydrogen bonding interaction with hinge region residue Leu83. Evaluation against leukaemia cell lines revealed low cytotoxicity, which is in line with the high selectivity towards CDK2. This study demonstrates that substituted imidazo[1,2-c]pyrimidines can be exploited for future kinase inhibitor development.


Subject(s)
Cyclin-Dependent Kinase 2/antagonists & inhibitors , Imidazoles/chemistry , Protein Kinase Inhibitors/chemical synthesis , Pyrimidines/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Binding Sites , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Cyclin-Dependent Kinase 2/metabolism , Humans , Hydrogen Bonding , Imidazoles/metabolism , Imidazoles/pharmacology , Molecular Dynamics Simulation , Protein Binding , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines/metabolism , Pyrimidines/pharmacology , Structure-Activity Relationship
7.
Antiviral Res ; 185: 104971, 2021 01.
Article in English | MEDLINE | ID: mdl-33166574

ABSTRACT

Influenza viruses can cause severe respiratory infections in humans, leading to nearly half a million deaths worldwide each year. Improved antiviral drugs are needed to address the threat of development of novel pandemic strains. Current therapeutic interventions target three key proteins in the viral life cycle: neuraminidase, the M2 channel and RNA-dependent-RNA polymerase. Protein-protein interactions between influenza polymerase subunits are potential new targets for drug development. Using a newly developed assay based on AlphaScreen technology, we screened a peptide panel for protein-protein interaction inhibitors to identify a minimal PB1 subunit-derived peptide that retains high inhibition potential and can be further modified. Here, we present an X-ray structure of the resulting decapeptide bound to the C-terminal domain of PA polymerase subunit from pandemic isolate A/California/07/2009 H1N1 at 1.6 Å resolution and discuss its implications for the design of specific, potent influenza polymerase inhibitors.


Subject(s)
Influenza A Virus, H1N1 Subtype/enzymology , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Antiviral Agents/pharmacology , Crystallization , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/metabolism , Protein Binding , Protein Interaction Domains and Motifs/drug effects , Protein Interaction Domains and Motifs/physiology , Viral Proteins/antagonists & inhibitors
8.
ACS Infect Dis ; 7(1): 189-201, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33301315

ABSTRACT

Azapeptide nitriles are postulated to reversibly covalently react with the active-site cysteine residue of cysteine proteases and form isothiosemicarbazide adducts. We investigated the interaction of azadipeptide nitriles with the cathepsin B1 drug target (SmCB1) from Schistosoma mansoni, a pathogen that causes the global neglected disease schistosomiasis. Azadipeptide nitriles were superior inhibitors of SmCB1 over their parent carba analogs. We determined the crystal structure of SmCB1 in complex with an azadipeptide nitrile and analyzed the reaction mechanism using quantum chemical calculations. The data demonstrate that azadipeptide nitriles, in contrast to their carba counterparts, undergo a change from E- to Z-configuration upon binding, which gives rise to a highly favorable energy profile of noncovalent and covalent complex formation. Finally, azadipeptide nitriles were considerably more lethal than their carba analogs against the schistosome pathogen in culture, supporting the further development of this chemotype as a treatment for schistosomiasis.


Subject(s)
Peptide Hydrolases , Schistosoma mansoni , Animals , Cathepsin B
9.
FEBS J ; 287(15): 3315-3327, 2020 08.
Article in English | MEDLINE | ID: mdl-31943739

ABSTRACT

Rutinosidases (α-l-rhamnosyl-ß-d-glucosidases) catalyze the cleavage of the glycosidic bond between the aglycone and the disaccharide rutinose (α-l-rhamnopyranosyl-(1→6)-ß-d-glucopyranose) of specific flavonoid glycosides such as rutin (quercetin 3-O-rutinoside). Microbial rutinosidases are part of the rutin catabolic pathway, enabling the microorganism to utilize rutin and related plant phenolic glycosides. Here, we report the first three-dimensional structure of a rutinosidase determined at 1.27-Å resolution. The rutinosidase from Aspergillus niger K2 (AnRut), a member of glycoside hydrolase family GH-5, subfamily 23, was heterologously produced in Pichia pastoris. The X-ray structure of AnRut is represented by a distorted (ß/α)8 barrel fold with its closest structural homologue being an exo-ß-(1,3)-glucanase from Candida albicans (CaExg). The catalytic site is located in a deep pocket with a striking structural similarity to CaExg. However, the entrance to the active site of AnRut has been found to be different from that of CaExg - a mostly unstructured section of ~ 40 residues present in CaExg is missing in AnRut, whereas an additional loop of 13 amino acids partially covers the active site of AnRut. NMR analysis of reaction products provided clear evidence for a retaining reaction mechanism of AnRut. Unexpectedly, quercetin 3-O-glucoside was found to be a better substrate than rutin, and thus, AnRut cannot be considered a typical diglycosidase. Mutational analysis of conserved active site residues in combination with in silico modeling allowed identification of essential interactions for enzyme activity and helped to reveal further details of substrate binding. The protein sequence of AnRut has been revised. DATABASES: The nucleotide sequence of the rutinosidase-encoding gene is available in the GenBank database under the accession number MN393234. Structural data are available in the PDB database under the accession number 6I1A. ENZYME: α-l-Rhamnosyl-ß-d-glucosidase (EC 3.2.1.168).


Subject(s)
Aspergillus niger/enzymology , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/metabolism , Rutin/metabolism , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Fungal Proteins/genetics , Glycoside Hydrolases/genetics , Models, Molecular , Mutation , Oxidation-Reduction , Protein Conformation , Rutin/chemistry , Sequence Homology
10.
J Med Chem ; 62(21): 9560-9575, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31568723

ABSTRACT

Carbonic anhydrase IX (CAIX) is a transmembrane enzyme that regulates pH in hypoxic tumors and promotes tumor cell survival. Its expression is associated with the occurrence of metastases and poor prognosis. Here, we present nine derivatives of the cobalt bis(dicarbollide)(1-) anion substituted at the boron or carbon sites by alkysulfamide group(s) as highly specific and selective inhibitors of CAIX. Interactions of these compounds with the active site of CAIX were explored on the atomic level using protein crystallography. Two selected derivatives display subnanomolar or picomolar inhibition constants and high selectivity for the tumor-specific CAIX over cytosolic isoform CAII. Both derivatives had a time-dependent effect on the growth of multicellular spheroids of HT-29 and HCT116 colorectal cancer cells, facilitated penetration and/or accumulation of doxorubicin into spheroids, and displayed low toxicity and showed promising pharmacokinetics and a significant inhibitory effect on tumor growth in syngenic breast 4T1 and colorectal HT-29 cancer xenotransplants.


Subject(s)
Amides/chemistry , Boranes/chemistry , Boranes/pharmacology , Carbonic Anhydrase IX/metabolism , Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrase Inhibitors/pharmacology , Animals , Biological Transport/drug effects , Carbonic Anhydrase IX/chemistry , Catalytic Domain , Cell Line, Tumor , Doxorubicin/metabolism , Drug Design , Drug Synergism , Humans , Mice , Models, Molecular , Xenograft Model Antitumor Assays
11.
Acta Crystallogr D Struct Biol ; 75(Pt 8): 743-752, 2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31373573

ABSTRACT

The haloacid dehalogenase (HAD) superfamily is one of the largest known groups of enzymes and the majority of its members catalyze the hydrolysis of phosphoric acid monoesters into a phosphate ion and an alcohol. Despite the fact that sequence similarity between HAD phosphatases is generally very low, the members of the family possess some characteristic features, such as a Rossmann-like fold, HAD signature motifs or the requirement for Mg2+ ion as an obligatory cofactor. This study focuses on a new hypothetical HAD phosphatase from Thermococcus thioreducens. The protein crystallized in space group P21212, with unit-cell parameters a = 66.3, b = 117.0, c = 33.8 Å, and the crystals contained one molecule in the asymmetric unit. The protein structure was determined by X-ray crystallography and was refined to 1.75 Šresolution. The structure revealed a putative active site common to all HAD members. Computational docking into the crystal structure was used to propose substrates of the enzyme. The activity of this thermophilic enzyme towards several of the selected substrates was confirmed at temperatures of 37°C as well as 60°C.


Subject(s)
Hydrolases/chemistry , Phosphoric Monoester Hydrolases/chemistry , Thermococcus/enzymology , Binding Sites , Catalytic Domain , Crystallography, X-Ray/methods , Kinetics , Models, Molecular , Substrate Specificity
12.
Bioorg Med Chem ; 27(13): 2935-2947, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31128993

ABSTRACT

This study focuses on design, synthesis and in vitro evaluation of inhibitory potency of two series of sialylmimetic that target an exosite ("150-cavity") adjacent to the active site of influenza neuraminidases from A/California/07/2009 (H1N1) pandemic strain and A/chicken/Nakorn-Patom/Thailand/CU-K2-2004 (H5N1). The structure-activity analysis as well as 3-D structure of the complex of parental compound with the pandemic neuraminidase p09N1 revealed high flexibility of the 150-cavity towards various modification of the neuraminidase inhibitors. Furthermore, our comparison of two methods for inhibition constant determination performed at slightly different pH values suggest that the experimental conditions of the measurement could dramatically influence the outcome of the analysis in the compound-dependent manner. Therefore, previously reported Ki values determined at non-physiological pH should be carefully scrutinized.


Subject(s)
Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/pathogenicity , Neuraminidase/therapeutic use , Oseltamivir/therapeutic use , Humans , Neuraminidase/pharmacology , Oseltamivir/pharmacology
13.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 5): 324-331, 2019 May 01.
Article in English | MEDLINE | ID: mdl-31045561

ABSTRACT

Haloalkane dehalogenases (HLDs) convert halogenated aliphatic pollutants to less toxic compounds by a hydrolytic mechanism. Owing to their broad substrate specificity and high enantioselectivity, haloalkane dehalogenases can function as biosensors to detect toxic compounds in the environment or can be used for the production of optically pure compounds. Here, the structural analysis of the haloalkane dehalogenase DpcA isolated from the psychrophilic bacterium Psychrobacter cryohalolentis K5 is presented at the atomic resolution of 1.05 Å. This enzyme exhibits a low temperature optimum, making it attractive for environmental applications such as biosensing at the subsurface environment, where the temperature typically does not exceed 25°C. The structure revealed that DpcA possesses the shortest access tunnel and one of the most widely open main tunnels among structural homologs of the HLD-I subfamily. Comparative analysis revealed major differences in the region of the α4 helix of the cap domain, which is one of the key determinants of the anatomy of the tunnels. The crystal structure of DpcA will contribute to better understanding of the structure-function relationships of cold-adapted enzymes.


Subject(s)
Bacterial Proteins/chemistry , Hydrocarbons, Halogenated/chemistry , Hydrolases/chemistry , Psychrobacter/enzymology , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Cloning, Molecular , Cold Temperature , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Hydrocarbons, Halogenated/metabolism , Hydrolases/genetics , Hydrolases/metabolism , Molecular Docking Simulation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Psychrobacter/chemistry , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Structural Homology, Protein , Substrate Specificity , Thermodynamics
14.
J Med Chem ; 62(9): 4606-4623, 2019 05 09.
Article in English | MEDLINE | ID: mdl-30943029

ABSTRACT

Cyclin-dependent kinases are therapeutic targets frequently deregulated in various cancers. By convenient alkylation of the 5-sulfanyl group, we synthesized 3-isopropyl-7-[4-(2-pyridyl)benzyl]amino-1(2) H-pyrazolo[4,3- d]pyrimidines with various substitutions at position 5 with potent antiproliferative activity in non-Hodgkin lymphoma cell lines. The most potent derivative 4.35 also displayed activities across more than 60 cancer cell lines. The kinase profiling confirmed high selectivity of 4.35 toward cyclin-dependent kinases (CDKs) 2, 5, and 9, and the cocrystal with CDK2/cyclin A2 revealed its binding in the active site. Cultured lymphoma cell lines treated with 4.35 showed dephosphorylation of CDK substrates, cleavage of PARP-1, downregulation of XIAP and MCL-1, and activation of caspases, which collectively confirmed ongoing apoptosis. Moreover, 4.35 demonstrated significant activity in various cell line xenograft and patient-derived xenograft mouse models in vivo both as a monotherapy and as a combination therapy with the BCL2-targeting venetoclax. These findings support further studies of combinatorial treatment based on CDK inhibitors.


Subject(s)
Antineoplastic Agents/therapeutic use , Cyclin-Dependent Kinases/antagonists & inhibitors , Lymphoma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Female , Humans , Mice , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Pyrazoles/chemical synthesis , Pyrazoles/pharmacology , Pyrimidines/chemical synthesis , Pyrimidines/pharmacology , Structure-Activity Relationship , Sulfonamides/pharmacology , Xenograft Model Antitumor Assays
15.
Biochem J ; 475(23): 3847-3860, 2018 12 10.
Article in English | MEDLINE | ID: mdl-30404922

ABSTRACT

Influenza neuraminidase is responsible for the escape of new viral particles from the infected cell surface. Several neuraminidase inhibitors are used clinically to treat patients or stockpiled for emergencies. However, the increasing development of viral resistance against approved inhibitors has underscored the need for the development of new antivirals effective against resistant influenza strains. A facile, sensitive, and inexpensive screening method would help achieve this goal. Recently, we described a multiwell plate-based DNA-linked inhibitor antibody assay (DIANA). This highly sensitive method can quantify femtomolar concentrations of enzymes. DIANA also has been applied to high-throughput enzyme inhibitor screening, allowing the evaluation of inhibition constants from a single inhibitor concentration. Here, we report the design, synthesis, and structural characterization of a tamiphosphor derivative linked to a reporter DNA oligonucleotide for the development of a DIANA-type assay to screen potential influenza neuraminidase inhibitors. The neuraminidase is first captured by an immobilized antibody, and the test compound competes for binding to the enzyme with the oligo-linked detection probe, which is then quantified by qPCR. We validated this novel assay by comparing it with the standard fluorometric assay and demonstrated its usefulness for sensitive neuraminidase detection as well as high-throughput screening of potential new neuraminidase inhibitors.


Subject(s)
DNA/chemistry , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Influenza A virus/drug effects , Oseltamivir/analogs & derivatives , Phosphorous Acids/chemistry , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Enzyme Inhibitors/chemistry , Humans , Influenza A virus/enzymology , Influenza A virus/physiology , Influenza, Human/drug therapy , Influenza, Human/enzymology , Influenza, Human/virology , Neuraminidase/antagonists & inhibitors , Neuraminidase/metabolism , Oseltamivir/chemistry , Reproducibility of Results , Viral Proteins/antagonists & inhibitors , Viral Proteins/metabolism
16.
Acta Crystallogr D Struct Biol ; 74(Pt 11): 1078-1084, 2018 Nov 01.
Article in English | MEDLINE | ID: mdl-30387766

ABSTRACT

α-L-Rhamnosidases cleave terminal nonreducing α-L-rhamnosyl residues from many natural rhamnoglycosides. This makes them catalysts of interest for various biotechnological applications. The X-ray structure of the GH78 family α-L-rhamnosidase from Aspergillus terreus has been determined at 1.38 Šresolution using the sulfur single-wavelength anomalous dispersion phasing method. The protein was isolated from its natural source in the native glycosylated form, and the active site contained a glucose molecule, probably from the growth medium. In addition to its catalytic domain, the α-L-rhamnosidase from A. terreus contains four accessory domains of unknown function. The structural data suggest that two of these accessory domains, E and F, might play a role in stabilizing the aglycon portion of the bound substrate.


Subject(s)
Aspergillus/enzymology , Glycoside Hydrolases/chemistry , Protein Conformation , Catalytic Domain , Crystallography, X-Ray , Glycoside Hydrolases/metabolism , Glycosylation , Models, Molecular
17.
Viruses ; 10(7)2018 06 21.
Article in English | MEDLINE | ID: mdl-29933553

ABSTRACT

Neuraminidase is the main target for current influenza drugs. Reduced susceptibility to oseltamivir, the most widely prescribed neuraminidase inhibitor, has been repeatedly reported. The resistance substitutions I223V and S247N, alone or in combination with the major oseltamivir-resistance mutation H275Y, have been observed in 2009 pandemic H1N1 viruses. We overexpressed and purified the ectodomain of wild-type neuraminidase from the A/California/07/2009 (H1N1) influenza virus, as well as variants containing H275Y, I223V, and S247N single mutations and H275Y/I223V and H275Y/S247N double mutations. We performed enzymological and thermodynamic analyses and structurally examined the resistance mechanism. Our results reveal that the I223V or S247N substitution alone confers only a moderate reduction in oseltamivir affinity. In contrast, the major oseltamivir resistance mutation H275Y causes a significant decrease in the enzyme's ability to bind this drug. Combination of H275Y with an I223V or S247N mutation results in extreme impairment of oseltamivir's inhibition potency. Our structural analyses revealed that the H275Y substitution has a major effect on the oseltamivir binding pose within the active site while the influence of other studied mutations is much less prominent. Our crystal structures also helped explain the augmenting effect on resistance of combining H275Y with both substitutions.


Subject(s)
Drug Resistance, Viral/genetics , Influenza A Virus, H1N1 Subtype/genetics , Neuraminidase/chemistry , Neuraminidase/genetics , Amino Acid Substitution , Antiviral Agents/pharmacology , Calorimetry , Crystallization , Enzyme Inhibitors/pharmacology , Humans , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/enzymology , Influenza, Human/virology , Kinetics , Mutation, Missense , Oseltamivir/pharmacology , Thermodynamics , Viral Proteins/chemistry , Viral Proteins/genetics , Virus Replication
18.
Acta Crystallogr F Struct Biol Commun ; 74(Pt 5): 300-306, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29717998

ABSTRACT

Human aldo-keto reductase 1C3 (AKR1C3) stereospecifically reduces steroids and prostaglandins and is involved in the biotransformation of xenobiotics. Its role in various cancers makes it a potential therapeutic target for the development of inhibitors. Recombinant AKR1C3 with a thrombin-cleavable N-terminal His6 tag was expressed from a pET-28(+) vector for structural studies of enzyme-inhibitor complexes. A modified in situ proteolysis approach was applied to specifically remove the His tag by thrombin cleavage during crystallization screening trials. This improved the morphology and diffraction quality of the crystals and allowed the acquisition of high-resolution diffraction data and structure solution. This approach may be generally applicable to other proteins expressed using the pET-28(+) vector.


Subject(s)
Aldo-Keto Reductase Family 1 Member C3/chemistry , Aldo-Keto Reductase Family 1 Member C3/metabolism , Histidine , Thrombin/metabolism , Aldo-Keto Reductase Family 1 Member C3/genetics , Amino Acid Sequence , Crystallization/methods , Crystallography, X-Ray/methods , Histidine/genetics , Humans , Proteolysis , X-Ray Diffraction/methods
19.
Leukemia ; 32(6): 1393-1403, 2018 06.
Article in English | MEDLINE | ID: mdl-29535428

ABSTRACT

Activating mutations in NT5C2, a gene encoding cytosolic purine 5'-nucleotidase (cN-II), confer chemoresistance in relapsed acute lymphoblastic leukemia. Here we show that all mutants became independent of allosteric effects of ATP and thus constitutively active. Structural mapping of mutations described in patients demonstrates that 90% of leukemia-specific allelles directly affect two regulatory hotspots within the cN-II molecule-the helix A region: residues 355-365, and the intersubunit interface: helix B (232-242) and flexible interhelical loop L (400-418). Furthermore, analysis of hetero-oligomeric complexes combining wild-type (WT) and mutant subunits showed that the activation is transmitted from the mutated to the WT subunit. This intersubunit interaction forms structural basis of hyperactive NT5C2 in drug-resistant leukemia in which heterozygous NT5C2 mutation gave rise to hetero-tetramer mutant and WT proteins. This enabled us to define criteria to aid the prediction of NT5C2 drug resistance mutations in leukemia.


Subject(s)
5'-Nucleotidase/genetics , Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , 5'-Nucleotidase/chemistry , Drug Resistance, Neoplasm , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Protein Conformation , Protein Subunits/chemistry , Recurrence
20.
FEBS J ; 285(3): 580-598, 2018 02.
Article in English | MEDLINE | ID: mdl-29239122

ABSTRACT

ß-N-acetylhexosaminidase from the fungus Aspergillus oryzae is a secreted extracellular enzyme that cleaves chitobiose into constituent monosaccharides. It belongs to the GH 20 glycoside hydrolase family and consists of two N-glycosylated catalytic cores noncovalently associated with two 10-kDa O-glycosylated propeptides. We used X-ray diffraction and mass spectrometry to determine the structure of A. oryzae ß-N-acetylhexosaminidase isolated from its natural source. The three-dimensional structure determined and refined to a resolution of 2.3 Å revealed that this enzyme is active as a uniquely tight dimeric assembly further stabilized by N- and O-glycosylation. The propeptide from one subunit forms extensive noncovalent interactions with the catalytic core of the second subunit in the dimer, and this chain swap suggests the distinctive structural mechanism of the enzyme's activation. Unique structural features of ß-N-acetylhexosaminidase from A. oryzae define a very stable and robust framework suitable for biotechnological applications. The crystal structure reported here provides structural insights into the enzyme architecture as well as the detailed configuration of the active site. These insights can be applied to rational enzyme engineering. DATABASE: Structural data are available in the PDB database under the accession number 5OAR. ENZYME: ß-N-acetylhexosaminidase (EC 3.2.1.52).


Subject(s)
Aspergillus oryzae/enzymology , Fungal Proteins/metabolism , G(M2) Activator Protein/metabolism , G(M2) Ganglioside/metabolism , Models, Molecular , beta-N-Acetylhexosaminidases/metabolism , Amino Acid Sequence , Binding Sites , Catalytic Domain , Conserved Sequence , Crystallography, X-Ray , Dimerization , Enzyme Precursors/chemistry , Enzyme Precursors/metabolism , Fungal Proteins/chemistry , G(M2) Activator Protein/chemistry , G(M2) Ganglioside/chemistry , Glycosylation , Ligands , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational , Protein Stability , Sequence Alignment , Structural Homology, Protein , Substrate Specificity , beta-N-Acetylhexosaminidases/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...