Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
NPJ Vaccines ; 6(1): 7, 2021 Jan 08.
Article in English | MEDLINE | ID: mdl-33420102

ABSTRACT

HCV vaccine development is stymied by the high genetic diversity of the virus and the variability of the envelope glycoproteins. One strategy to overcome this is to identify conserved, functionally important regions-such as the epitopes of broadly neutralizing antibodies (bNAbs)-and use these as a basis for structure-based vaccine design. Here, we report an anti-idiotype approach that has generated an antibody that mimics a highly conserved neutralizing epitope on HCV E2. Crucially, a mutagenesis screen was used to identify the antibody, designated B2.1 A, whose binding characteristics to the bNAb AP33 closely resemble those of the original antigen. Protein crystallography confirmed that B2.1 A is a structural mimic of the AP33 epitope. When used as an immunogen B2.1 A induced antibodies that recognized the same epitope and E2 residues as AP33 and most importantly protected against HCV challenge in a mouse model.

2.
Biochemistry ; 57(26): 3797-3806, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29812914

ABSTRACT

The thermoacidophilic archaea Picrophilus torridus and Sulfolobus solfataricus catabolize glucose via a nonphosphorylative Entner-Doudoroff pathway and a branched Entner-Doudoroff pathway, respectively. Key enzymes for these Entner-Doudoroff pathways are the aldolases, 2-keto-3-deoxygluconate aldolase (KDG-aldolase) and 2-keto-3-deoxy-6-phosphogluconate aldolase [KD(P)G-aldolase]. KDG-aldolase from P. torridus (Pt-KDG-aldolase) is highly specific for the nonphosphorylated substrate, 2-keto-3-deoxygluconate (KDG), whereas KD(P)G-aldolase from S. solfataricus [Ss-KD(P)G-aldolase] is an enzyme that catalyzes the cleavage of both KDG and 2-keto-3-deoxy-6-phosphogluconate (KDPG), with a preference for KDPG. The structural basis for the high specificity of Pt-KDG-aldolase for KDG as compared to the more promiscuous Ss-KD(P)G-aldolase has not been analyzed before. In this work, we report the elucidation of the structure of Ss-KD(P)G-aldolase in complex with KDPG at 2.35 Å and that of KDG-aldolase from P. torridus at 2.50 Å resolution. By superimposition of the active sites of the two enzymes, and subsequent site-directed mutagenesis studies, a network of four amino acids, namely, Arg106, Tyr132, Arg237, and Ser241, was identified in Ss-KD(P)G-aldolase that interact with the negatively charged phosphate group of KDPG, thereby increasing the affinity of the enzyme for KDPG. This KDPG-binding network is absent in Pt-KDG-aldolase, which explains the low catalytic efficiency of KDPG cleavage.


Subject(s)
Aldehyde-Lyases/chemistry , Archaeal Proteins/chemistry , Gluconates/chemistry , Sulfolobus solfataricus/enzymology , Thermoplasmales/enzymology , Models, Molecular , Protein Domains , Structure-Activity Relationship
3.
Oncotarget ; 7(50): 83017-83030, 2016 Dec 13.
Article in English | MEDLINE | ID: mdl-27806324

ABSTRACT

PRIMA-1Met is the methylated PRIMA-1 (p53 reactivation and induction of massive apoptosis) and could restore tumor suppressor function of mutant p53 and induce p53 dependent apoptosis in cancer cells harboring mutant p53. However, p53 independent activity of PRIMA-1Met remains elusive. Here we reported that PRIMA-1Met attenuated colorectal cancer cell growth irrespective of p53 status. Kinase profiling revealed that mitogen-activated or extracellular signal-related protein kinase (MEK) might be a potential target of PRIMA-1Met. Pull-down binding and ATP competitive assay showed that PRIMA-1Met directly bound MEK in vitro and in cells. Furthermore, the direct binding sites of PRIMA-1Met were explored by using a computational docking model. Treatment of colorectal cancer cells with PRIMA-1Met inhibited p53-independent phosphorylation of MEK, which in turn impaired anchorage-independent cell growth in vitro. Moreover, PRIMA-1Met suppressed colorectal cancer growth in xenograft mouse model by inhibiting MEK1 activity.Taken together, our findings demonstrate a novel p53-independent activity of PRIMA-1Met to inhibit MEK and suppress colorectal cancer growth.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , MAP Kinase Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Quinuclidines/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/metabolism , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , HCT116 Cells , HT29 Cells , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinases/metabolism , Mice, Nude , Molecular Docking Simulation , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/metabolism , Quinuclidines/metabolism , Signal Transduction/drug effects , Time Factors , Tumor Burden/drug effects , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
5.
J Biol Chem ; 290(46): 27736-48, 2015 Nov 13.
Article in English | MEDLINE | ID: mdl-26370075

ABSTRACT

Streptococcus pneumoniae is an important human pathogen that causes a range of disease states. Sialidases are important bacterial virulence factors. There are three pneumococcal sialidases: NanA, NanB, and NanC. NanC is an unusual sialidase in that its primary reaction product is 2-deoxy-2,3-didehydro-N-acetylneuraminic acid (Neu5Ac2en, also known as DANA), a nonspecific hydrolytic sialidase inhibitor. The production of Neu5Ac2en from α2-3-linked sialosides by the catalytic domain is confirmed within a crystal structure. A covalent complex with 3-fluoro-ß-N-acetylneuraminic acid is also presented, suggesting a common mechanism with other sialidases up to the final step of product formation. A conformation change in an active site hydrophobic loop on ligand binding constricts the entrance to the active site. In addition, the distance between the catalytic acid/base (Asp-315) and the ligand anomeric carbon is unusually short. These features facilitate a novel sialidase reaction in which the final step of product formation is direct abstraction of the C3 proton by the active site aspartic acid, forming Neu5Ac2en. NanC also possesses a carbohydrate-binding module, which is shown to bind α2-3- and α2-6-linked sialosides, as well as N-acetylneuraminic acid, which is captured in the crystal structure following hydration of Neu5Ac2en by NanC. Overall, the pneumococcal sialidases show remarkable mechanistic diversity while maintaining a common structural scaffold.


Subject(s)
Bacterial Proteins/chemistry , Enzyme Inhibitors/chemical synthesis , Neuraminidase/chemistry , Streptococcus pneumoniae/enzymology , Sugar Acids/chemical synthesis , Bacterial Proteins/ultrastructure , Catalytic Domain , Crystallography, X-Ray , Neuraminidase/ultrastructure
6.
BMC Struct Biol ; 15: 15, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26289431

ABSTRACT

BACKGROUND: Streptococcus pneumoniae Neuraminidase A (NanA) is a multi-domain protein anchored to the bacterial surface. Upstream of the catalytic domain of NanA is a domain that conforms to the sialic acid-recognising CBM40 family of the CAZY (carbohydrate-active enzymes) database. This domain has been identified to play a critical role in allowing the bacterium to promote adhesion and invasion of human brain microvascular endothelial cells, and hence may play a key role in promoting bacterial meningitis. In addition, the CBM40 domain has also been reported to activate host chemokines and neutrophil recruitment during infection. RESULTS: Crystal structures of both apo- and holo- forms of the NanA CBM40 domain (residues 121 to 305), have been determined to 1.8 Å resolution. The domain shares the fold of other CBM40 domains that are associated with sialidases. When in complex with α2,3- or α2,6-sialyllactose, the domain is shown to interact only with the terminal sialic acid. Significantly, a deep acidic pocket adjacent to the sialic acid-binding site is identified, which is occupied by a lysine from a symmetry-related molecule in the crystal. This pocket is adjacent to a region that is predicted to be involved in protein-protein interactions. CONCLUSIONS: The structural data provide the details of linkage-independent sialyllactose binding by NanA CBM40 and reveal striking surface features that may hold the key to recognition of binding partners on the host cell surface. The structure also suggests that small molecules or sialic acid analogues could be developed to fill the acidic pocket and hence provide a new therapeutic avenue against meningitis caused by S. pneumoniae.


Subject(s)
Bacterial Proteins/chemistry , Neuraminidase/chemistry , Streptococcus pneumoniae/enzymology , Virulence Factors/chemistry , Bacterial Proteins/metabolism , Binding Sites , Lactose/analogs & derivatives , Lactose/metabolism , Models, Molecular , Neuraminidase/metabolism , Protein Binding , Protein Folding , Protein Structure, Tertiary , Sialic Acids/metabolism , Streptococcus pneumoniae/chemistry , Virulence Factors/metabolism
7.
Nat Commun ; 6: 7624, 2015 Jul 08.
Article in English | MEDLINE | ID: mdl-26154892

ABSTRACT

The gastrointestinal mucus layer is colonized by a dense community of microbes catabolizing dietary and host carbohydrates during their expansion in the gut. Alterations in mucosal carbohydrate availability impact on the composition of microbial species. Ruminococcus gnavus is a commensal anaerobe present in the gastrointestinal tract of >90% of humans and overrepresented in inflammatory bowel diseases (IBD). Using a combination of genomics, enzymology and crystallography, we show that the mucin-degrader R. gnavus ATCC 29149 strain produces an intramolecular trans-sialidase (IT-sialidase) that cleaves off terminal α2-3-linked sialic acid from glycoproteins, releasing 2,7-anhydro-Neu5Ac instead of sialic acid. Evidence of IT-sialidases in human metagenomes indicates that this enzyme occurs in healthy subjects but is more prevalent in IBD metagenomes. Our results uncover a previously unrecognized enzymatic activity in the gut microbiota, which may contribute to the adaptation of intestinal bacteria to the mucosal environment in health and disease.


Subject(s)
Adaptation, Physiological/physiology , Gene Expression Regulation, Bacterial/physiology , Glycoproteins/metabolism , Intestinal Mucosa/microbiology , Neuraminidase/metabolism , Ruminococcus/enzymology , Gene Expression Regulation, Enzymologic/physiology , Glycoproteins/genetics , Humans , Mucins/metabolism , Neuraminidase/genetics , Ruminococcus/genetics , Ruminococcus/metabolism
8.
Antimicrob Agents Chemother ; 59(3): 1495-504, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25534734

ABSTRACT

Compounds that target the cellular factors essential for influenza virus replication represent an innovative approach to antiviral therapy. Sp2CBMTD is a genetically engineered multivalent protein that masks sialic acid-containing cellular receptors on the respiratory epithelium, which are recognized by influenza viruses. Here, we evaluated the antiviral potential of Sp2CBMTD against lethal infection in mice with an emerging A/Anhui/1/2013 (H7N9) influenza virus and addressed the mechanistic basis of its activity in vivo. Sp2CBMTD was administered to mice intranasally as a single or repeated dose (0.1, 1, 10, or 100 µg) before (day -7, -3, and/or -1) or after (6 or 24 h) H7N9 virus inoculation. A single Sp2CBMTD dose (10 or 100 µg) protected 80% to 100% of the mice when administered 7 days before the H7N9 lethal challenge. Repeated Sp2CBMTD administration conferred the highest protection, resulting in 100% survival of the mice even at the lowest dose tested (0.1 µg). When treatment began 24 h after exposure to the H7N9 virus, a single administration of 100 µg of Sp2CBMTD protected 40% of the mice from death. The administration of Sp2CBMTD induced the pulmonary expression of proinflammatory mediators (interleukin-6 [IL-6], IL-1ß, RANTES, monocyte chemotactic protein-1 [MCP-1], macrophage inflammatory protein-1α [MIP-1α], and inducible protein [IP-10]) and recruited neutrophils to the respiratory tract before H7N9 virus infection, which resulted in less pronounced inflammation and rapid virus clearance from mouse lungs. Sp2CBMTD administration did not affect the virus-specific adaptive immune response, which was sufficient to protect against reinfection with a higher dose of homologous H7N9 virus or heterologous H5N1 virus. Thus, Sp2CBMTD was effective in preventing H7N9 infections in a lethal mouse model and holds promise as a prophylaxis option against zoonotic influenza viruses.


Subject(s)
Antiviral Agents/therapeutic use , Carrier Proteins/therapeutic use , Influenza A Virus, H7N9 Subtype , Orthomyxoviridae Infections/drug therapy , Receptors, Cell Surface/physiology , Sialic Acids/metabolism , Animals , Chemokines/biosynthesis , Cytokines/biosynthesis , Female , Lung/immunology , Lung/virology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Virus Replication
9.
Proc Natl Acad Sci U S A ; 111(17): 6401-6, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24733924

ABSTRACT

There is a need for new approaches for the control of influenza given the burden caused by annual seasonal outbreaks, the emergence of viruses with pandemic potential, and the development of resistance to current antiviral drugs. We show that multivalent biologics, engineered using carbohydrate-binding modules specific for sialic acid, mask the cell-surface receptor recognized by the influenza virus and protect mice from a lethal challenge with 2009 pandemic H1N1 influenza virus. The most promising biologic protects mice when given as a single 1-µg intranasal dose 7 d in advance of viral challenge. There also is sufficient virus replication to establish an immune response, potentially protecting the animal from future exposure to the virus. Furthermore, the biologics appear to stimulate inflammatory mediators, and this stimulation may contribute to their protective ability. Our results suggest that this host-targeted approach could provide a front-line prophylactic that has the potential to protect against any current and future influenza virus and possibly against other respiratory pathogens that use sialic acid as a receptor.


Subject(s)
Influenza, Human/metabolism , Influenza, Human/prevention & control , Protein Engineering , Receptors, Virus/metabolism , Animals , Body Weight , Chemokines/metabolism , Dogs , Humans , Inflammation Mediators/metabolism , Influenza A Virus, H1N1 Subtype/physiology , Lung/pathology , Lung/virology , Madin Darby Canine Kidney Cells , Mice , N-Acetylneuraminic Acid/metabolism , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology , Receptors, Cell Surface/metabolism , Survival Analysis
10.
Biochemistry ; 52(51): 9177-86, 2013 Dec 23.
Article in English | MEDLINE | ID: mdl-24295366

ABSTRACT

Aspergillus fumigatus is an airborne fungal pathogen. We previously cloned and characterized an exo-sialidase from A. fumigatus and showed that it preferred 2-keto-3-deoxynononic acid (KDN) as a substrate to N-acetylneuraminic acid (Neu5Ac). The purpose of this study was to investigate the structure-function relationships of critical catalytic site residues. Site-directed mutagenesis was used to create three mutant recombinant enzymes: the catalytic nucleophile (Y358H), the general acid/base catalyst (D84A), and an enlargement of the binding pocket to attempt to accommodate the N-acetyl group of Neu5Ac (R171L). Crystal structures for all enzymes were determined. The D84A mutation had an effect in decreasing the activity of AfKDNase that was stronger than that of the same mutation in the structurally similar sialidase from the bacterium Micromonospora viridifaciens. These data suggest that the catalytic acid is more important in the reaction of AfKDNase and that catalysis is less dependent on nucleophilic or electrostatic stabilization of the developing positive charge at the transition state for hydrolysis. Removal of the catalytic nucleophile (Y358H) significantly lowered the activity of the enzyme, but this mutant remained a retaining glycosidase as demonstrated by nuclear magnetic resonance spectroscopic analysis. This is a novel finding that has not been shown with other sialidases. Kinetic activity measured at pH 5.2 revealed that R171L had higher activity on a Neu5Ac-based substrate than wild-type KDNase; hence, leucine in place of arginine in the binding pocket improved catalysis toward Neu5Ac substrates. Hence, whether a sialidase is primarily a KDNase or a neuraminidase is due in part to the presence of an amino acid that creates a steric clash with the N-acetyl group.


Subject(s)
Aspergillus fumigatus/enzymology , Fungal Proteins/metabolism , Glycoside Hydrolases/metabolism , Micromonospora/enzymology , Models, Molecular , Mutant Proteins/metabolism , Neuraminidase/metabolism , Amino Acid Substitution , Biocatalysis , Catalytic Domain , Crystallography, X-Ray , Fungal Proteins/chemistry , Fungal Proteins/genetics , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/genetics , Hydrogen-Ion Concentration , Hydrolysis , Keto Acids/chemistry , Keto Acids/metabolism , Kinetics , Molecular Conformation , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , N-Acetylneuraminic Acid/chemistry , N-Acetylneuraminic Acid/metabolism , Neuraminidase/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Substrate Specificity
11.
BMC Biochem ; 13: 19, 2012 Oct 02.
Article in English | MEDLINE | ID: mdl-23031230

ABSTRACT

BACKGROUNDS: Streptococcus pneumoniae expresses three distinct sialidases, NanA, NanB, and NanC, that are believed to be key virulence factors and thus, potential important drug targets. We previously reported that the three enzymes release different products from sialosides, but could share a common catalytic mechanism before the final step of product formation. However, the kinetic investigations of the three sialidases have not been systematically done thus far, due to the lack of an easy and steady measurement of sialidase reaction rate. RESULTS: In this work, we present further kinetic characterization of pneumococcal sialidases by using a direct spectrophotometric method with the chromogenic substrate p-nitrophenyl-N-acetylneuraminic acid (p-NP-Neu5Ac). Using our assay, the measured kinetic parameters of the three purified pneumococcal sialidase, NanA, NanB and NanC, were obtained and were in perfect agreement with the previously published data. The major advantage of this alternative method resides in the direct measurement of the released product, allowing to readily determine of initial reaction rates and record complete hydrolysis time courses. CONCLUSION: We developed an accurate, fast and sensitive spectrophotometric method to investigate the kinetics of sialidase-catalyzed reactions. This fast, sensitive, inexpensive and accurate method could benefit the study of the kinetics and inhibition of sialidases in general.


Subject(s)
Neuraminidase/antagonists & inhibitors , Biocatalysis , Buffers , Hydrogen-Ion Concentration , Hydrolysis , Kinetics , N-Acetylneuraminic Acid/metabolism , Neuraminidase/genetics , Neuraminidase/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spectrophotometry, Ultraviolet , Streptococcus pneumoniae/enzymology , Substrate Specificity
12.
Chembiochem ; 13(16): 2374-83, 2012 Nov 05.
Article in English | MEDLINE | ID: mdl-23070966

ABSTRACT

The major human pathogen Streptococcus pneumoniae plays a key role in several disease states including septicaemia, meningitis and community-acquired pneumonia. Although vaccines against S. pneumoniae are available as prophylactics, there remains a need to identify and characterise novel chemical entities that can treat the diseases caused by this pathogen. S. pneumoniae expresses three sialidases, enzymes that cleave sialic acid from carbohydrate-based surface molecules. Two of these enzymes, NanA and NanB, have been implicated in the pathogenesis of S. pneumoniae and are considered to be validated drug targets. Here we report our studies on the synthesis and structural characterisation of novel NanB-selective inhibitors that are inspired by the ß-amino-sulfonic acid family of buffers.


Subject(s)
Enzyme Inhibitors/pharmacology , Hymecromone/analogs & derivatives , Neuraminidase/antagonists & inhibitors , Streptococcus pneumoniae/enzymology , Sulfonic Acids/pharmacology , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Hymecromone/chemical synthesis , Hymecromone/chemistry , Hymecromone/pharmacology , Models, Molecular , Molecular Structure , Neuraminidase/chemistry , Neuraminidase/metabolism , Structure-Activity Relationship , Sulfonic Acids/chemical synthesis , Sulfonic Acids/chemistry
13.
J Virol ; 86(23): 12923-32, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22993159

ABSTRACT

The E2 envelope glycoprotein of hepatitis C virus (HCV) binds to the host entry factor CD81 and is the principal target for neutralizing antibodies (NAbs). Most NAbs recognize hypervariable region 1 on E2, which undergoes frequent mutation, thereby allowing the virus to evade neutralization. Consequently, there is great interest in NAbs that target conserved epitopes. One such NAb is AP33, a mouse monoclonal antibody that recognizes a conserved, linear epitope on E2 and potently neutralizes a broad range of HCV genotypes. In this study, the X-ray structure of AP33 Fab in complex with an epitope peptide spanning residues 412 to 423 of HCV E2 was determined to 1.8 Å. In the complex, the peptide adopts a ß-hairpin conformation and docks into a deep binding pocket on the antibody. The major determinants of antibody recognition are E2 residues L413, N415, G418, and W420. The structure is compared to the recently described HCV1 Fab in complex with the same epitope. Interestingly, the antigen-binding sites of HCV1 and AP33 are completely different, whereas the peptide conformation is very similar in the two structures. Mutagenesis of the peptide-binding residues on AP33 confirmed that these residues are also critical for AP33 recognition of whole E2, confirming that the peptide-bound structure truly represents AP33 interaction with the intact glycoprotein. The slightly conformation-sensitive character of the AP33-E2 interaction was explored by cross-competition analysis and alanine-scanning mutagenesis. The structural details of this neutralizing epitope provide a starting point for the design of an immunogen capable of eliciting AP33-like antibodies.


Subject(s)
Antibodies, Neutralizing/immunology , Hepatitis C/prevention & control , Models, Molecular , Tetraspanin 28/immunology , Viral Envelope Proteins/immunology , Viral Hepatitis Vaccines/immunology , Viral Proteins/immunology , Animals , Antibodies, Neutralizing/chemistry , Crystallography, X-Ray , Epitopes/genetics , Mice , Mutagenesis , Tetraspanin 28/metabolism , Viral Envelope Proteins/metabolism , Viral Proteins/metabolism
14.
Glycobiology ; 22(2): 174-80, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21846691

ABSTRACT

The hemagglutinin-neuraminidase (HN) glycoprotein is utilized by human parainfluenza viruses for binding to the host cell. By the use of glycan array assays, we demonstrate that, in addition to the first catalytic-binding site, the HN of human parainfluenza virus type 1 has a second site for binding covered by N-linked glycan. Our data suggest that attachment of the first site to sialic acid (SA)-linked receptors triggers exposure of the second site. We found that both sites bind to α2-3-linked SAs with a preference for a sialyl-Lewis(x) motif. Binding to α2-3-linked SAs with a sulfated sialyl-Lewis motif as well as to α2-8-linked SAs was unique for the second binding site. Neither site recognizes α2-6-linked oligosaccharides.


Subject(s)
Enzyme Inhibitors/metabolism , HN Protein/chemistry , HN Protein/metabolism , Mutation/genetics , Neuraminidase/chemistry , Parainfluenza Virus 1, Human/metabolism , Receptors, Cell Surface/metabolism , Azides/chemistry , Azides/metabolism , Binding Sites , Enzyme Inhibitors/chemistry , HN Protein/genetics , Hexuronic Acids/chemistry , Hexuronic Acids/metabolism , Humans , N-Acetylneuraminic Acid/analogs & derivatives , N-Acetylneuraminic Acid/chemistry , N-Acetylneuraminic Acid/metabolism , Neuraminidase/antagonists & inhibitors , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Parainfluenza Virus 1, Human/genetics , Protein Binding , Receptors, Cell Surface/chemistry , Sialyl Lewis X Antigen
15.
J Mol Biol ; 409(4): 496-503, 2011 Jun 17.
Article in English | MEDLINE | ID: mdl-21514303

ABSTRACT

The human pathogen Streptococcus pneumoniae is the major cause of bacterial meningitis, respiratory tract infection, septicemia, and otitis media. The bacterium expresses neuraminidase (NA) proteins that contribute to pathogenesis by cleaving sialic acids from host glycoconjugates, thereby enhancing biofilm formation and colonization. Recent in vivo experiments have shown that antiviral compounds, widely used in clinics and designed to inhibit influenza NA, significantly reduce biofilm formation and nasopharyngeal colonization of S. pneumoniae in mice. Here, we present the structural basis for the beneficial effect of these compounds against pneumococcal infection. Crystal structures of pneumococcal NanA in complex with zanamivir and oseltamivir carboxylate are discussed, correlated with measured inhibitory constants K(i), and compared with the binding modes of the inhibitors in the viral enzyme. Inhibitor structures show for the first time how clinically approved anti-influenza compounds interact with an NA of the human pathogen S. pneumoniae and give a rational explanation for their antibacterial effects.


Subject(s)
Antiviral Agents/pharmacology , Neuraminidase/antagonists & inhibitors , Neuraminidase/chemistry , Oseltamivir/analogs & derivatives , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/enzymology , Zanamivir/pharmacology , Animals , Carboxylic Acids/chemistry , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Molecular Sequence Data , Molecular Structure , Neuraminidase/genetics , Neuraminidase/metabolism , Oseltamivir/pharmacology , Protein Conformation
16.
J Biol Chem ; 286(12): 10783-92, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21247893

ABSTRACT

Aspergillus fumigatus is a filamentous fungus that can cause severe respiratory disease in immunocompromised individuals. A putative sialidase from A. fumigatus was recently cloned and shown to be relatively poor in cleaving N-acetylneuraminic acid (Neu5Ac) in comparison with bacterial sialidases. Here we present the first crystal structure of a fungal sialidase. When the apo structure was compared with bacterial sialidase structures, the active site of the Aspergillus enzyme suggested that Neu5Ac would be a poor substrate because of a smaller pocket that normally accommodates the acetamido group of Neu5Ac in sialidases. A sialic acid with a hydroxyl in place of an acetamido group is 2-keto-3-deoxynononic acid (KDN). We show that KDN is the preferred substrate for the A. fumigatus sialidase and that A. fumigatus can utilize KDN as a sole carbon source. A 1.45-Å resolution crystal structure of the enzyme in complex with KDN reveals KDN in the active site in a boat conformation and nearby a second binding site occupied by KDN in a chair conformation, suggesting that polyKDN may be a natural substrate. The enzyme is not inhibited by the sialidase transition state analog 2-deoxy-2,3-dehydro-N-acetylneuraminic acid (Neu5Ac2en) but is inhibited by the related 2,3-didehydro-2,3-dideoxy-D-glycero-D-galacto-nonulosonic acid that we show bound to the enzyme in a 1.84-Å resolution crystal structure. Using a fluorinated KDN substrate, we present a 1.5-Å resolution structure of a covalently bound catalytic intermediate. The A. fumigatus sialidase is therefore a KDNase with a similar catalytic mechanism to Neu5Ac exosialidases, and this study represents the first structure of a KDNase.


Subject(s)
Aspergillus fumigatus/enzymology , Fungal Proteins/chemistry , Glycoside Hydrolases/chemistry , Neuraminidase/chemistry , Binding Sites , Crystallography, X-Ray , Fungal Proteins/metabolism , Glycoside Hydrolases/metabolism , N-Acetylneuraminic Acid/chemistry , N-Acetylneuraminic Acid/metabolism , Neuraminidase/metabolism , Protein Structure, Tertiary , Structure-Activity Relationship
17.
J Am Chem Soc ; 133(6): 1718-21, 2011 Feb 16.
Article in English | MEDLINE | ID: mdl-21244006

ABSTRACT

Streptococcus penumoniae is a major human pathogen responsible for respiratory tract infections, septicemia, and meningitis and continues to produce numerous cases of disease with relatively high mortalities. S. pneumoniae encodes up to three sialidases, NanA, NanB, and NanC, that have been implicated in pathogenesis and are potential drug targets. NanA has been shown to be a promiscuous sialidase, hydrolyzing the removal of Neu5Ac from a variety of glycoconjugates with retention of configuration at the anomeric center, as we confirm by NMR. NanB is an intramolecular trans-sialidase producing 2,7-anhydro-Neu5Ac selectively from α2,3-sialosides. Here, we show that the first product of NanC is 2-deoxy-2,3-didehydro-N-acetylneuraminic acid (Neu5Ac2en) that can be slowly hydrated by the enzyme to Neu5Ac. We propose that the three pneumococcal sialidases share a common catalytic mechanism up to the final product formation step, and speculate on the roles of the enzymes in the lifecycle of the bacterium.


Subject(s)
Neuraminidase/metabolism , Streptococcus pneumoniae/enzymology , Biocatalysis , Catalytic Domain , Kinetics , Models, Molecular , Neuraminidase/chemistry
18.
Acta Crystallogr D Biol Crystallogr ; 66(Pt 4): 325-38, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20382985

ABSTRACT

When collecting X-ray diffraction data from a crystal, we measure the intensities of the diffracted waves scattered from a series of planes that we can imagine slicing through the crystal in all directions. From these intensities we derive the amplitudes of the scattered waves, but in the experiment we lose the phase information; that is, how we offset these waves when we add them together to reconstruct an image of our molecule. This is generally known as the 'phase problem'. We can only derive the phases from some knowledge of the molecular structure. In small-molecule crystallography, some basic assumptions about atomicity give rise to relationships between the amplitudes from which phase information can be extracted. In protein crystallography, these ab initio methods can only be used in the rare cases in which there are data to at least 1.2 A resolution. For the majority of cases in protein crystallography phases are derived either by using the atomic coordinates of a structurally similar protein (molecular replacement) or by finding the positions of heavy atoms that are intrinsic to the protein or that have been added (methods such as MIR, MIRAS, SIR, SIRAS, MAD, SAD or combinations of these). The pioneering work of Perutz, Kendrew, Blow, Crick and others developed the methods of isomorphous replacement: adding electron-dense atoms to the protein without disturbing the protein structure. Nowadays, methods from small-molecule crystallography can be used to find the heavy-atom substructure and the phases for the whole protein can be bootstrapped from this prior knowledge. More recently, improved X-ray sources, detectors and software have led to the routine use of anomalous scattering to obtain phase information from either incorporated selenium or intrinsic sulfurs. In the best cases, only a single set of X-ray data (SAD) is required to provide the positions of the anomalous scatters, which together with density-modification procedures can reveal the structure of the complete protein.


Subject(s)
Crystallography, X-Ray/methods , Proteins/analysis , Computer Simulation , Models, Molecular , Probability , Protein Structure, Tertiary , Proteins/chemistry
19.
J Struct Funct Genomics ; 11(2): 167-80, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20419351

ABSTRACT

The Scottish Structural Proteomics Facility was funded to develop a laboratory scale approach to high throughput structure determination. The effort was successful in that over 40 structures were determined. These structures and the methods harnessed to obtain them are reported here. This report reflects on the value of automation but also on the continued requirement for a high degree of scientific and technical expertise. The efficiency of the process poses challenges to the current paradigm of structural analysis and publication. In the 5 year period we published ten peer-reviewed papers reporting structural data arising from the pipeline. Nevertheless, the number of structures solved exceeded our ability to analyse and publish each new finding. By reporting the experimental details and depositing the structures we hope to maximize the impact of the project by allowing others to follow up the relevant biology.


Subject(s)
Laboratories/organization & administration , Proteins/chemistry , Proteins/metabolism , Proteomics/organization & administration , Computational Biology , Crystallization , Humans , Proteins/genetics , Scotland
20.
J Mol Biol ; 392(3): 763-73, 2009 Sep 25.
Article in English | MEDLINE | ID: mdl-19631226

ABSTRACT

CqsA is an enzyme involved in the biosynthesis of cholerae autoinducer-1 (CAI-1), the major Vibrio cholerae autoinducer engaged in quorum sensing. The amino acid sequence of CqsA suggests that it belongs to the family of alpha-oxoamine synthases that catalyse the condensation of an amino acid to an acyl-CoA substrate. Here we present the apo- and PLP-bound crystal structures of CqsA and confirm that it shares structural homology with the dimeric alpha-oxoamine synthases, including a conserved PLP-binding site. The chemical structure of CAI-1 suggests that decanoyl-CoA may be one substrate of CqsA and that another substrate may be l-threonine or l-2-aminobutyric acid. A crystal structure of CqsA at 1.9-A resolution obtained in the presence of PLP and l-threonine reveals an external aldimine that has lost the l-threonine side chain. Similarly, a 1.9-A-resolution crystal structure of CqsA in the presence of PLP, l-threonine, and decanoyl-CoA shows a trapped external aldimine intermediate, suggesting that the condensation and decarboxylation steps have occurred, again with loss of the l-threonine side chain. It is suggested that this side-chain loss, an observation supported by mass spectrometry, is due to a retro-aldol reaction. Although no structural data have been obtained on CqsA using l-2-aminobutyric acid and decanoyl-CoA as substrates, mass spectrometry confirms the expected product of the enzyme reaction. It is proposed that a region of structure that is disordered in the apo structure is involved in the release of product. While not confirming if CqsA alone is able to synthesize CAI-1, these results suggest possible synthetic routes.


Subject(s)
Bacterial Proteins/biosynthesis , Bacterial Proteins/chemistry , Ketones , Protein Conformation , Vibrio cholerae/enzymology , Amino Acid Sequence , Bacterial Proteins/genetics , Binding Sites , Crystallography, X-Ray , Humans , Ketones/chemistry , Ketones/metabolism , Models, Molecular , Molecular Sequence Data , Molecular Structure , Schiff Bases/chemistry , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...