Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
Acta Crystallogr D Struct Biol ; 79(Pt 8): 735-745, 2023 Aug 01.
Article En | MEDLINE | ID: mdl-37428844

Candida auris has emerged as a global health problem with a dramatic spread by nosocomial transmission and a high mortality rate. Antifungal therapy for C. auris infections is currently limited due to widespread resistance to fluconazole and amphotericin B and increasing resistance to the front-line drug echinocandin. Therefore, new treatments are urgently required to combat this pathogen. Dihydrofolate reductase (DHFR) has been validated as a potential drug target for Candida species, although no structure of the C. auris enzyme (CauDHFR) has been reported. Here, crystal structures of CauDHFR are reported as an apoenzyme, as a holoenzyme and in two ternary complexes with pyrimethamine and cycloguanil, which are common antifolates, at near-atomic resolution. Preliminary biochemical and biophysical assays and antifungal susceptibility testing with a variety of classical antifolates were also performed, highlighting the enzyme-inhibition rates and the inhibition of yeast growth. These structural and functional data might provide the basis for a novel drug-discovery campaign against this global threat.


Candidiasis, Invasive , Folic Acid Antagonists , Humans , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Candida auris , Tetrahydrofolate Dehydrogenase , Microbial Sensitivity Tests , Candidiasis, Invasive/drug therapy , Saccharomyces cerevisiae
2.
J Struct Biol ; 215(2): 107957, 2023 06.
Article En | MEDLINE | ID: mdl-36944394

The cytoplasmatic biosynthesis of the stem peptide from the peptidoglycan in bacteria involves six steps, which have the role of three ATP-dependent Mur ligases that incorporate three consecutive amino acids to a substrate precursor. MurE is the last Mur ligase to incorporate a free amino acid. Although the structure of MurE from Mycobacterium tuberculosis (MtbMurE) was determined at 3.0 Å, the binding mode of meso-Diaminopimelate (m-DAP) and the effect of substrate absence is unknown. Herein, we show the structure of MurE from M. thermoresistibile (MthMurE) in complex with ADP and m-DAP at 1.4 Å resolution. The analysis of the structure indicates key conformational changes that the substrate UDP-MurNAc-L-Ala-D-Glu (UAG) and the free amino acid m-DAP cause on the MthMurE conformation. We observed several movements of domains or loop regions that displace their position in order to perform enzymatic catalysis. Since MthMurE has a high similarity to MtbMurE, this enzyme could also guide strategies for structure-based antimicrobial discovery to fight against tuberculosis or other mycobacterial infections.


Mycobacterium tuberculosis , Peptide Synthases , Peptide Synthases/chemistry , Bacterial Proteins/chemistry , Mycobacterium tuberculosis/metabolism , Amino Acids
3.
Genet Mol Biol ; 46(1 Suppl 2): e20220261, 2023.
Article En | MEDLINE | ID: mdl-36718771

Genetically antimicrobial resistance in Mycobacterium tuberculosis is currently one of the most important aspects of tuberculosis, considering that there are emerging resistant strains for almost every known drug used for its treatment. There are multiple antimicrobials used for tuberculosis treatment, and the most effective ones are the first-line drugs, which include isoniazid, pyrazinamide, rifampicin, and ethambutol. In this context, understanding the mechanisms of action and resistance of these molecules is essential for proposing new therapies and strategies of treatment. Additionally, understanding how and where mutations arise conferring a resistance profile to the bacteria and their effect on bacterial metabolism is an important requisite to be taken in producing safer and less susceptible drugs to the emergence of resistance. In this review, we summarize the most recent literature regarding novel mutations reported between 2017 and 2022 and the advances in the molecular mechanisms of action and resistance against first-line drugs used in tuberculosis treatment, highlighting recent findings in pyrazinamide resistance involving PanD and, additionally, resistance-conferring mutations for novel drugs such as bedaquiline, pretomanid, delamanid and linezolid.

4.
PLoS Negl Trop Dis ; 14(10): e0008091, 2020 10.
Article En | MEDLINE | ID: mdl-33017394

Eukaryotes from the Excavata superphylum have been used as models to study the evolution of cellular molecular processes. Strikingly, human parasites of the Trypanosomatidae family (T. brucei, T. cruzi and L. major) conserve the complex machinery responsible for selenocysteine biosynthesis and incorporation in selenoproteins (SELENOK/SelK, SELENOT/SelT and SELENOTryp/SelTryp), although these proteins do not seem to be essential for parasite viability under laboratory controlled conditions. Selenophosphate synthetase (SEPHS/SPS) plays an indispensable role in selenium metabolism, being responsible for catalyzing the formation of selenophosphate, the biological selenium donor for selenocysteine synthesis. We solved the crystal structure of the L. major selenophosphate synthetase and confirmed that its dimeric organization is functionally important throughout the domains of life. We also demonstrated its interaction with selenocysteine lyase (SCLY) and showed that it is not present in other stable assemblies involved in the selenocysteine pathway, namely the phosphoseryl-tRNASec kinase (PSTK)-Sec-tRNASec synthase (SEPSECS) complex and the tRNASec-specific elongation factor (eEFSec) complex. Endoplasmic reticulum stress with dithiothreitol (DTT) or tunicamycin upon selenophosphate synthetase ablation in procyclic T. brucei cells led to a growth defect. On the other hand, only DTT presented a negative effect in bloodstream T. brucei expressing selenophosphate synthetase-RNAi. Furthermore, selenoprotein T (SELENOT) was dispensable for both forms of the parasite. Together, our data suggest a role for the T. brucei selenophosphate synthetase in the regulation of the parasite's ER stress response.


Lyases/metabolism , Phosphotransferases/metabolism , Selenocysteine/biosynthesis , Selenoproteins/metabolism , Trypanosoma brucei brucei/enzymology , Protein Conformation , Protozoan Proteins/metabolism , Selenium/metabolism
5.
Acta Crystallogr D Struct Biol ; 76(Pt 9): 868-875, 2020 Sep 01.
Article En | MEDLINE | ID: mdl-32876062

The genus Streptomyces is characterized by the production of a wide variety of secondary metabolites with remarkable biological activities and broad antibiotic capabilities. The presence of an unprecedented number of genes encoding hydrolytic enzymes with industrial appeal such as epoxide hydrolases (EHs) reveals its resourceful microscopic machinery. The whole-genome sequence of Streptomyces sp. CBMAI 2042, an endophytic actinobacterium isolated from Citrus sinensis branches, was explored by genome mining, and a putative α/ß-epoxide hydrolase named B1EPH2 and encoded by 344 amino acids was selected for functional and structural studies. The crystal structure of B1EPH2 was obtained at a resolution of 2.2 Šand it was found to have a similar fold to other EHs, despite its hexameric quaternary structure, which contrasts with previously solved dimeric and monomeric EH structures. While B1EPH2 has a high sequence similarity to EHB from Mycobacterium tuberculosis, its cavity is similar to that of human EH. A group of 12 aromatic and aliphatic racemic epoxides were assayed to determine the activity of B1EPH2; remarkably, this enzyme was able to hydrolyse all the epoxides to the respective 1,2-diols, indicating a wide-range substrate scope acceptance. Moreover, the (R)- and (S)-enantiomers of styrene oxide, epichlorohydrin and 1,2-epoxybutane were used to monitor enantiopreference. Taken together, the functional and structural analyses indicate that this enzyme is an attractive biocatalyst for future biotechnological applications.


Bacterial Proteins/chemistry , Epoxide Hydrolases/chemistry , Streptomyces/enzymology , Models, Molecular , Protein Conformation
6.
Bioorg Med Chem ; 28(15): 115600, 2020 08 01.
Article En | MEDLINE | ID: mdl-32631571

The enzyme dihydrofolate reductase from M.tuberculosis (MtDHFR) has a high unexploited potential to be a target for new drugs against tuberculosis (TB), due to its importance for pathogen survival. Preliminary studies have obtained fragment-like molecules with low affinity to MtDHFR which can potentially become lead compounds. Taking this into account, the fragment MB872 was used as a prototype for analogue development by bioisosterism/retro-bioisosterism, which resulted in 20 new substituted 3-benzoic acid derivatives. Compounds were active against MtDHFR, with IC50 values ranging from 7 to 40 µM, where compound 4e not only had the best inhibitory activity (IC50 = 7 µM), but also was 71-fold more active than the original fragment MB872. The 4e inhibition kinetics indicated an uncompetitive mechanism, which was supported by molecular modeling which suggested that the compounds can access an independent backpocket from the substrate and competitive inhibitors. Thus, based on these results, substituted 3-benzoic acid derivatives have strong potential to be developed as novel MtDHFR inhibitors and also anti-TB agents.


Antitubercular Agents/pharmacology , Bacterial Proteins/metabolism , Benzoates/pharmacology , Folic Acid Antagonists/pharmacology , Mycobacterium tuberculosis/drug effects , Tetrahydrofolate Dehydrogenase/metabolism , Antitubercular Agents/chemical synthesis , Antitubercular Agents/metabolism , Bacterial Proteins/chemistry , Benzoates/chemical synthesis , Benzoates/metabolism , Catalytic Domain , Drug Design , Folic Acid Antagonists/chemical synthesis , Folic Acid Antagonists/metabolism , Kinetics , Molecular Dynamics Simulation , Molecular Structure , Protein Binding , Structure-Activity Relationship , Tetrahydrofolate Dehydrogenase/chemistry
7.
Acta Crystallogr D Struct Biol ; 75(Pt 7): 682-693, 2019 Jul 01.
Article En | MEDLINE | ID: mdl-31282477

Tuberculosis is a disease caused by Mycobacterium tuberculosis and is the leading cause of death from a single infectious pathogen, with a high prevalence in developing countries in Africa and Asia. There still is a need for the development or repurposing of novel therapies to combat this disease owing to the long-term nature of current therapies and because of the number of reported resistant strains. Here, structures of dihydrofolate reductase from M. tuberculosis (MtDHFR), which is a key target of the folate pathway, are reported in complex with four antifolates, pyrimethamine, cycloguanil, diaverdine and pemetrexed, and its substrate dihydrofolate in order to understand their binding modes. The structures of all of these complexes were obtained in the closed-conformation state of the enzyme and a fine structural analysis indicated motion in key regions of the substrate-binding site and different binding modes of the ligands. In addition, the affinities, through Kd measurement, of diaverdine and methotrexate have been determined; MtDHFR has a lower affinity (highest Kd) for diaverdine than pyrimethamine and trimethoprim, and a very high affinity for methotrexate, as expected. The structural comparisons and analysis described in this work provide new information about the plasticity of MtDHFR and the binding effects of different antifolates.


Folic Acid Antagonists/chemistry , Folic Acid/analogs & derivatives , Mycobacterium tuberculosis/enzymology , Tetrahydrofolate Dehydrogenase/chemistry , Binding Sites , Crystallization/methods , Crystallography, X-Ray/methods , Escherichia coli/genetics , Folic Acid/chemistry , Humans , Ligands , Models, Molecular , Protein Conformation , Tuberculosis/microbiology
8.
ACS Chem Biol ; 14(5): 925-933, 2019 05 17.
Article En | MEDLINE | ID: mdl-30995396

Gentamicins are clinically relevant aminoglycoside antibiotics produced by several Micromonospora species. Gentamicins are highly methylated and functionalized molecules, and their biosynthesis include glycosyltransferases, dehydratase/oxidoreductases, aminotransferases, and methyltransferases. The biosynthesis of gentamicin A from gentamicin A2 involves three enzymatic steps that modify the hydroxyl group at position 3″ of the unusual garosamine sugar to provide its substitution for an amino group, followed by an N-methylation. The first of these reactions is catalyzed by GenD2, an oxidoreductase from the Gfo/Idh/MocA protein family, which reduces the hydroxyl at the C3″ of gentamicin A to produce 3''-dehydro-3''-oxo-gentamicin A2 (DOA2). In this work, we solved the structure of GenD2 in complex with NAD+. Although the structure of GenD2 has a similar fold to other members of the Gfo/Idh/MocA family, this enzyme has several new features, including a 3D-domain swapping of two ß-strands that are involved in a novel oligomerization interface for this protein family. In addition, the active site of this enzyme also has several specialties which are possibly involved in the substrate specificity, including a number of aromatic residues and a negatively charged region, which is complementary to the polycationic aminoglycoside-substrate. Therefore, docking simulations provided insights into the recognition of gentamicin A2 and into the catalytic mechanism of GenD2. This is the first report describing the structure of an oxidoreductase involved in aminoglycoside biosynthesis and could open perspectives into producing new aminoglycoside derivatives by protein engineering.


Gentamicins/biosynthesis , NAD/metabolism , Oxidoreductases/metabolism , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Crystallography, X-Ray , Methylation , Molecular Docking Simulation , Oxidoreductases/chemistry , Protein Conformation , Sequence Homology, Amino Acid , Substrate Specificity
9.
ACS Infect Dis ; 5(2): 260-271, 2019 02 08.
Article En | MEDLINE | ID: mdl-30556998

Targeting Mycobacterium tuberculosis peptidoglycans with ß-lactam antibiotics represents a strategy to address increasing resistance to antitubercular drugs. ß-Lactams inhibit peptidoglycan synthases such as l,d-transpeptidases, a group of carbapenem-sensitive enzymes that stabilize peptidoglycans through 3 → 3 cross-links. M. tuberculosis encodes five l,d-transpeptidases (LdtMt1-5), of which LdtMt3 is one of the less understood. Herein, we structurally characterized the apo and faropenem-acylated forms of LdtMt3 at 1.3 and 1.8 Å resolution, respectively. These structures revealed a fold and catalytic diad similar to those of other LdtsMt enzymes, supporting its involvement in transpeptidation reactions despite divergences in active site size and charges. The LdtMt3-faropenem structure indicated that faropenem is degraded after Cys-246 acylation, and possibly only a ß-OH-butyrate or an acetyl group (C2H3O) covalently attached to the enzyme remains, an observation that strongly supports the notion that LdtMt3 is inactivated by ß-lactams. Docking simulations with intact ß-lactams predicted key LdtMt3 residues that interact with these antibiotics. We also characterized the heat of acylation involved in the binding and reaction of LdtMt3 for ten ß-lactams belonging to four different classes, and imipenem had the highest inactivation constant. This work provides key insights into the structure, binding mechanisms, and degradation of ß-lactams by LdtMt3, which may be useful for the development of additional ß-lactams with potential antitubercular activity.


Antitubercular Agents/metabolism , Mycobacterium tuberculosis/enzymology , Peptidyl Transferases/metabolism , beta-Lactams/metabolism , Antitubercular Agents/pharmacology , Catalytic Domain , Molecular Docking Simulation , Mycobacterium tuberculosis/drug effects , beta-Lactams/pharmacology
10.
ACS Chem Biol ; 12(11): 2779-2787, 2017 11 17.
Article En | MEDLINE | ID: mdl-28876898

Gentamicins are heavily methylated, clinically valuable pseudotrisaccharide antibiotics produced by Micromonospora echinospora. GenN has been characterized as an S-adenosyl-l-methionine-dependent methyltransferase with low sequence similarity to other enzymes. It is responsible for the 3″-N-methylation of 3″-dehydro-3″-amino-gentamicin A2, an essential modification of ring III in the biosynthetic pathway to the gentamicin C complex. Purified recombinant GenN also efficiently catalyzes 3″-N-methylation of related aminoglycosides kanamycin B and tobramycin, which both contain an additional hydroxymethyl group at the C5″ position in ring III. We have obtained eight cocrystal structures of GenN, at a resolution of 2.2 Šor better, including the binary complex of GenN and S-adenosyl-l-homocysteine (SAH) and the ternary complexes of GenN, SAH, and several aminoglycosides. The GenN structure reveals several features not observed in any other N-methyltransferase that fit it for its role in gentamicin biosynthesis. These include a novel N-terminal domain that might be involved in protein:protein interaction with upstream enzymes of the gentamicin X2 biosynthesis and two long loops that are involved in aminoglycoside substrate recognition. In addition, the analysis of structures of GenN in complex with different ligands, supported by the results of active site mutagenesis, has allowed us to propose a catalytic mechanism and has revealed the structural basis for the surprising ability of native GenN to act on these alternative substrates.


Aminoglycosides/metabolism , Anti-Bacterial Agents/metabolism , Bacterial Proteins/metabolism , Gentamicins/metabolism , Methyltransferases/metabolism , Micromonospora/enzymology , Bacterial Proteins/chemistry , Crystallography, X-Ray , Kanamycin/analogs & derivatives , Kanamycin/metabolism , Methyltransferases/chemistry , Micromonospora/chemistry , Micromonospora/metabolism , Models, Molecular , Protein Conformation , Substrate Specificity , Tobramycin/metabolism
11.
Structure ; 22(1): 94-103, 2014 Jan 07.
Article En | MEDLINE | ID: mdl-24210757

Inhibition of the biosynthesis of tetrahydrofolate (THF) has long been a focus in the treatment of both cancer and infectious diseases. Dihydrofolate reductase (DHFR), which catalyzes the last step, is one of the most thoroughly explored targets of this pathway, but there are no DHFR inhibitors used for tuberculosis treatment. Here, we report a structural, site-directed mutagenesis and calorimetric analysis of Mycobacterium tuberculosis DHFR (MtDHFR) in complex with classical DHFR inhibitors. Our study provides insights into the weak inhibition of MtDHFR by trimethoprim and other antifolate drugs, such as pyrimethamine and cycloguanil. The construction of the mutant Y100F, together with calorimetric studies, gives insights into low affinity of MtDHFR for classical DHFR inhibitors. Finally, the structures of MtDHFR in complex with pyrimethamine and cycloguanil define important interactions in the active site and provide clues to the more effective design of antibiotics targeted against MtDHFR.


Bacterial Proteins/chemistry , Enzyme Inhibitors/chemistry , Folic Acid Antagonists/chemistry , Mycobacterium tuberculosis/chemistry , Tetrahydrofolate Dehydrogenase/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Calorimetry , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Ligands , Molecular Docking Simulation , Mutagenesis, Site-Directed , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/genetics , Proguanil/chemistry , Protein Conformation , Pyrimethamine/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Structure-Activity Relationship , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/metabolism , Thermodynamics , Triazines/chemistry , Trimethoprim/chemistry
12.
J Struct Biol ; 159(3): 369-80, 2007 Sep.
Article En | MEDLINE | ID: mdl-17588773

The resumption of tuberculosis led to an increased need to understand the molecular mechanisms of drug action and drug resistance, which should provide significant insight into the development of newer compounds. Isoniazid (INH), the most prescribed drug to treat TB, inhibits an NADH-dependent enoyl-acyl carrier protein reductase (InhA) that provides precursors of mycolic acids, which are components of the mycobacterial cell wall. InhA is the major target of the mode of action of isoniazid. INH is a pro-drug that needs activation to form the inhibitory INH-NAD adduct. Missense mutations in the inhA structural gene have been identified in clinical isolates of Mycobacterium tuberculosis resistant to INH. To understand the mechanism of resistance to INH, we have solved the structure of two InhA mutants (I21V and S94A), identified in INH-resistant clinical isolates, and compare them to INH-sensitive WT InhA structure in complex with the INH-NAD adduct. We also solved the structure of unliganded INH-resistant S94A protein, which is the first report on apo form of InhA. The salient features of these structures are discussed and should provide structural information to improve our understanding of the mechanism of action of, and resistance to, INH in M. tuberculosis. The unliganded structure of InhA allows identification of conformational changes upon ligand binding and should help structure-based drug design of more potent antimycobacterial agents.


Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Drug Resistance, Bacterial/genetics , Mycobacterium tuberculosis/enzymology , NADH, NADPH Oxidoreductases/chemistry , NADH, NADPH Oxidoreductases/genetics , Oxidoreductases/chemistry , Oxidoreductases/genetics , Crystallography, X-Ray , Isoniazid/analogs & derivatives , Isoniazid/chemistry , Isoniazid/pharmacology , Mutation, Missense , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , NAD/analogs & derivatives , NAD/chemistry , Oxidoreductases Acting on CH-CH Group Donors , Protein Conformation
13.
Article En | MEDLINE | ID: mdl-17183161

Bacteria, fungi and plants can convert carbohydrate and phosphoenolpyruvate into chorismate, which is the precursor of various aromatic compounds. The seven enzymes of the shikimate pathway are responsible for this conversion. Shikimate kinase (SK) is the fifth enzyme in this pathway and converts shikimate to shikimate-3-phosphate. In this work, the conformational changes that occur on binding of shikimate, magnesium and chloride ions to SK from Mycobacterium tuberculosis (MtSK) are described. It was observed that both ions and shikimate influence the conformation of residues of the active site of MtSK. Magnesium influences the conformation of the shikimate hydroxyl groups and the position of the side chains of some of the residues of the active site. Chloride seems to influence the affinity of ADP and its position in the active site and the opening length of the LID domain. Shikimate binding causes a closing of the LID domain and also seems to influence the crystallographic packing of SK. The results shown here could be useful for understanding the catalytic mechanism of SK and the role of ions in the activity of this protein.


Chlorides/metabolism , Magnesium/metabolism , Mycobacterium tuberculosis/enzymology , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Shikimic Acid/metabolism , Binding Sites , Chlorides/chemistry , Crystallography, X-Ray , Magnesium/chemistry , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Structure, Secondary , Shikimic Acid/chemistry
14.
Cell Biochem Biophys ; 44(3): 375-84, 2006.
Article En | MEDLINE | ID: mdl-16679524

The development of new therapies against infectious diseases is vital in developing countries. Among infectious diseases, tuberculosis is considered the leading cause of death. A target for development of new drugs is the tryptophan pathway. The last enzyme of this pathway, tryptophan synthase (TRPS), is responsible for conversion of the indole 3-glycerol phosphate into indol and the condensation of this molecule with serine-producing tryptophan. The present work describes the molecular models of TRPS from Mycobacterium tuberculosis (MtTRPS) complexed with six inhibitors, the indole 3-propanol phosphate and five arylthioalkyl-phosphonated analogs of substrate of the alpha-subunit. The molecular models of MtTRPS present good stereochemistry, and the binding of the inhibitors is favorable. Thus, the generated models can be used in the design of more specific drugs against tuberculosis and other infectious diseases.


Models, Molecular , Mycobacterium tuberculosis/enzymology , Tryptophan Synthase/antagonists & inhibitors , Tryptophan Synthase/chemistry , Binding Sites , Computer Simulation , Drug Design , Fagaceae/genetics , Hydrogen Bonding , Ligands , Molecular Structure , Plant Proteins/chemistry , Plant Proteins/classification , Protein Binding/physiology , Protein Conformation , Rosaceae/genetics , Sequence Alignment , Substrate Specificity
15.
Biochem Biophys Res Commun ; 326(2): 335-8, 2005 Jan 14.
Article En | MEDLINE | ID: mdl-15582582

Purine nucleoside phosphorylase (PNP) is a ubiquitous enzyme, which plays a key role in the purine salvage pathway, and PNP deficiency in humans leads to an impairment of T-cell function, usually with no apparent effects on B-cell function. Human PNP has been submitted to intensive structure-based design of inhibitors, most of them using low-resolution structures of human PNP. Here we report the crystal structure of human PNP in complex with hypoxanthine, refined to 2.6A resolution. The intermolecular interaction between ligand and PNP is discussed.


Hypoxanthine/metabolism , Purine-Nucleoside Phosphorylase/chemistry , Purine-Nucleoside Phosphorylase/metabolism , Sulfates/metabolism , Crystallography, X-Ray , Humans , Hypoxanthine/chemistry , Ions/chemistry , Ions/metabolism , Ligands , Models, Molecular , Molecular Structure , Protein Conformation , Sulfates/chemistry
16.
Biochem Biophys Res Commun ; 325(1): 10-7, 2004 Dec 03.
Article En | MEDLINE | ID: mdl-15522194

The crystal structure of shikimate kinase from Mycobacterium tuberculosis (MtSK) complexed with MgADP and shikimic acid (shikimate) has been determined at 2.3A resolution, clearly revealing the amino acid residues involved in shikimate binding. In MtSK, the Glu61 strictly conserved in SK forms a hydrogen bond and salt-bridge with Arg58 and assists in positioning the guanidinium group of Arg58 for shikimate binding. The carboxyl group of shikimate interacts with Arg58, Gly81, and Arg136, and hydroxyl groups with Asp34 and Gly80. The crystal structure of MtSK-MgADP-shikimate will provide crucial information for elucidation of the mechanism of SK-catalyzed reaction and for the development of a new generation of drugs against tuberculosis.


Bacterial Proteins , Mycobacterium tuberculosis/enzymology , Phosphotransferases (Alcohol Group Acceptor) , Shikimic Acid , Adenosine Diphosphate/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Crystallography, X-Ray , Hydrogen Bonding , Models, Molecular , Molecular Sequence Data , Molecular Structure , Mycobacterium tuberculosis/chemistry , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Binding , Protein Structure, Tertiary , Shikimic Acid/chemistry , Shikimic Acid/metabolism
17.
Biochem Biophys Res Commun ; 309(4): 917-22, 2003 Oct 03.
Article En | MEDLINE | ID: mdl-13679061

Purine nucleoside phosphorylase (PNP) catalyzes the phosphorolysis of the N-ribosidic bonds of purine nucleosides and deoxynucleosides. PNP is a target for inhibitor development aiming at T-cell immune response modulation. This work reports on the crystallographic study of the complex of human PNP-immucillin-H (HsPNP-ImmH) solved at 2.6A resolution using synchrotron radiation. Immucillin-H (ImmH) inhibits the growth of malignant T-cell lines in the presence of deoxyguanosine without affecting non-T-cell tumor lines. ImmH inhibits activated normal human T cells after antigenic stimulation in vitro. These biological effects of ImmH suggest that this agent may have utility in the treatment of certain human diseases characterized by abnormal T-cell growth or activation. This is the first structural report of human PNP complexed with immucillin-H. The comparison of the complex HsPNP-ImmH with recent crystallographic structures of human PNP explains the high specificity of immucillin-H for human PNP.


Enzyme Inhibitors/pharmacology , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , Pyrimidinones/pharmacology , Pyrroles/pharmacology , Crystallography, X-Ray , Humans , Ligands , Models, Molecular , Protein Conformation , Purine Nucleosides , Purine-Nucleoside Phosphorylase/chemistry
...