Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
NPJ Vaccines ; 8(1): 135, 2023 Sep 14.
Article in English | MEDLINE | ID: mdl-37709900
2.
mBio ; 14(5): e0094823, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37650625

ABSTRACT

IMPORTANCE: Biofilms are the communal way of life that microbes adopt to increase survival. Key to our ability to systematically promote or ablate biofilm formation is a detailed understanding of the biofilm matrix macromolecules. Here, we identify the first two essential steps in the Bacillus subtilis biofilm matrix exopolysaccharide (EPS) synthesis pathway. Together, our studies and approaches provide the foundation for the sequential characterization of the steps in EPS biosynthesis, using prior steps to enable chemoenzymatic synthesis of the undecaprenyl diphosphate-linked glycan substrates.


Subject(s)
Bacillus subtilis , Biofilms , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Extracellular Polymeric Substance Matrix/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
3.
Nat Chem Biol ; 19(7): 795-796, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37336978
4.
PLoS One ; 18(6): e0286540, 2023.
Article in English | MEDLINE | ID: mdl-37267309

ABSTRACT

Bacteria produce a variety of polysaccharides with functional roles in cell surface coating, surface and host interactions, and biofilms. We have identified an 'Orphan' bacterial cellulose synthase catalytic subunit (BcsA)-like protein found in four model pseudomonads, P. aeruginosa PA01, P. fluorescens SBW25, P. putida KT2440 and P. syringae pv. tomato DC3000. Pairwise alignments indicated that the Orphan and BcsA proteins shared less than 41% sequence identity suggesting they may not have the same structural folds or function. We identified 112 Orphans among soil and plant-associated pseudomonads as well as in phytopathogenic and human opportunistic pathogenic strains. The wide distribution of these highly conserved proteins suggest they form a novel family of synthases producing a different polysaccharide. In silico analysis, including sequence comparisons, secondary structure and topology predictions, and protein structural modelling, revealed a two-domain transmembrane ovoid-like structure for the Orphan protein with a periplasmic glycosyl hydrolase family GH17 domain linked via a transmembrane region to a cytoplasmic glycosyltransferase family GT2 domain. We suggest the GT2 domain synthesises ß-(1,3)-glucan that is transferred to the GH17 domain where it is cleaved and cyclised to produce cyclic-ß-(1,3)-glucan (CßG). Our structural models are consistent with enzymatic characterisation and recent molecular simulations of the PaPA01 and PpKT2440 GH17 domains. It also provides a functional explanation linking PaPAK and PaPA14 Orphan (also known as NdvB) transposon mutants with CßG production and biofilm-associated antibiotic resistance. Importantly, cyclic glucans are also involved in osmoregulation, plant infection and induced systemic suppression, and our findings suggest this novel family of CßG synthases may provide similar range of adaptive responses for pseudomonads.


Subject(s)
Polysaccharides , beta-Glucans , Humans , Polysaccharides/metabolism , Glucans , Protein Structure, Secondary , Biofilms , Pseudomonas aeruginosa/metabolism , Bacterial Proteins/metabolism , beta-Glucans/metabolism
5.
bioRxiv ; 2023 Feb 22.
Article in English | MEDLINE | ID: mdl-36865097

ABSTRACT

The Bacillus subtilis extracellular biofilm matrix includes an exopolysaccharide that is critical for the architecture and function of the community. To date, our understanding of the biosynthetic machinery and the molecular composition of the exopolysaccharide of B. subtilis remains unclear and incomplete. This report presents synergistic biochemical and genetic studies built from a foundation of comparative sequence analyses targeted at elucidating the activities of the first two membrane-committed steps in the exopolysaccharide biosynthetic pathway. By taking this approach, we determined the nucleotide sugar donor and lipid-linked acceptor substrates for the first two enzymes in the B. subtilis biofilm exopolysaccharide biosynthetic pathway. EpsL catalyzes the first phosphoglycosyl transferase step using UDP-di- N -acetyl bacillosamine as phospho-sugar donor. EpsD is a GT-B fold glycosyl transferase that facilitates the second step in the pathway that utilizes the product of EpsL as an acceptor substrate and UDP- N -acetyl glucosamine as the sugar donor. Thus, the study defines the first two monosaccharides at the reducing end of the growing exopolysaccharide unit. In doing so we provide the first evidence of the presence of bacillosamine in an exopolysaccharide synthesized by a Gram-positive bacterium. IMPORTANCE: Biofilms are the communal way of life that microbes adopt to increase survival. Key to our ability to systematically promote or ablate biofilm formation is a detailed understanding of the biofilm matrix macromolecules. Here we identify the first two essential steps in the Bacillus subtilis biofilm matrix exopolysaccharide synthesis pathway. Together our studies and approaches provide the foundation for the sequential characterization of the steps in exopolysaccharide biosynthesis, using prior steps to enable chemoenzymatic synthesis of the undecaprenol diphosphate-linked glycan substrates.

6.
NPJ Vaccines ; 8(1): 48, 2023 Mar 28.
Article in English | MEDLINE | ID: mdl-36977677

ABSTRACT

The Group A Carbohydrate (GAC) is a defining feature of Group A Streptococcus (Strep A) or Streptococcus pyogenes. It is a conserved and simple polysaccharide, comprising a rhamnose backbone and GlcNAc side chains, further decorated with glycerol phosphate on approximately 40% GlcNAc residues. Its conservation, surface exposure and antigenicity have made it an interesting focus on Strep A vaccine design. Glycoconjugates containing this conserved carbohydrate should be a key approach towards the successful mission to build a universal Strep A vaccine candidate. In this review, a brief introduction to GAC, the main carbohydrate component of Strep A bacteria, and a variety of published carrier proteins and conjugation technologies are discussed. Components and technologies should be chosen carefully for building affordable Strep A vaccine candidates, particularly for low- and middle-income countries (LMICs). Towards this, novel technologies are discussed, such as the prospective use of bioconjugation with PglB for rhamnose polymer conjugation and generalised modules for membrane antigens (GMMA), particularly as low-cost solutions to vaccine production. Rational design of "double-hit" conjugates encompassing species specific glycan and protein components would be beneficial and production of a conserved vaccine to target Strep A colonisation without invoking an autoimmune response would be ideal.

7.
PLoS One ; 18(2): e0281839, 2023.
Article in English | MEDLINE | ID: mdl-36795789

ABSTRACT

The Fructobacillus genus is a group of obligately fructophilic lactic acid bacteria (FLAB) that requires the use of fructose or another electron acceptor for their growth. In this work, we performed a comparative genomic analysis within the genus Fructobacillus by using 24 available genomes to evaluate genomic and metabolic differences among these organisms. In the genome of these strains, which varies between 1.15- and 1.75-Mbp, nineteen intact prophage regions, and seven complete CRISPR-Cas type II systems were found. Phylogenetic analyses located the studied genomes in two different clades. A pangenome analysis and a functional classification of their genes revealed that genomes of the first clade presented fewer genes involved in the synthesis of amino acids and other nitrogen compounds. Moreover, the presence of genes strictly related to the use of fructose and electron acceptors was variable within the genus, although these variations were not always related to the phylogeny.


Subject(s)
Lactobacillales , Leuconostocaceae , Fructose/metabolism , Phylogeny , Leuconostocaceae/genetics , Leuconostocaceae/metabolism , Lactobacillales/genetics , Genomics
8.
Elife ; 112022 04 08.
Article in English | MEDLINE | ID: mdl-35394422

ABSTRACT

Protein N-glycosylation is a post-translational modification found in organisms of all domains of life. The crenarchaeal N-glycosylation begins with the synthesis of a lipid-linked chitobiose core structure, identical to that in Eukaryotes, although the enzyme catalyzing this reaction remains unknown. Here, we report the identification of a thermostable archaeal ß-1,4-N-acetylglucosaminyltransferase, named archaeal glycosylation enzyme 24 (Agl24), responsible for the synthesis of the N-glycan chitobiose core. Biochemical characterization confirmed its function as an inverting ß-D-GlcNAc-(1→4)-α-D-GlcNAc-diphosphodolichol glycosyltransferase. Substitution of a conserved histidine residue, found also in the eukaryotic and bacterial homologs, demonstrated its functional importance for Agl24. Furthermore, bioinformatics and structural modeling revealed similarities of Agl24 to the eukaryotic Alg14/13 and a distant relation to the bacterial MurG, which are catalyzing the same or a similar reaction, respectively. Phylogenetic analysis of Alg14/13 homologs indicates that they are ancient in Eukaryotes, either as a lateral transfer or inherited through eukaryogenesis.


Subject(s)
Archaea , Eukaryota , Archaea/genetics , Disaccharides , Phylogeny , Polysaccharides
9.
Biochem J ; 478(12): 2385-2397, 2021 06 25.
Article in English | MEDLINE | ID: mdl-34096588

ABSTRACT

Endolysins are peptidoglycan (PG) hydrolases that function as part of the bacteriophage (phage) lytic system to release progeny phage at the end of a replication cycle. Notably, endolysins alone can produce lysis without phage infection, which offers an attractive alternative to traditional antibiotics. Endolysins from phage that infect Gram-positive bacterial hosts contain at least one enzymatically active domain (EAD) responsible for hydrolysis of PG bonds and a cell wall binding domain (CBD) that binds a cell wall epitope, such as a surface carbohydrate, providing some degree of specificity for the endolysin. Whilst the EADs typically cluster into conserved mechanistic classes with well-defined active sites, relatively little is known about the nature of the CBDs and only a few binding epitopes for CBDs have been elucidated. The major cell wall components of many streptococci are the polysaccharides that contain the polyrhamnose (pRha) backbone modified with species-specific and serotype-specific glycosyl side chains. In this report, using molecular genetics, microscopy, flow cytometry and lytic activity assays, we demonstrate the interaction of PlyCB, the CBD subunit of the streptococcal PlyC endolysin, with the pRha backbone of the cell wall polysaccharides, Group A Carbohydrate (GAC) and serotype c-specific carbohydrate (SCC) expressed by the Group A Streptococcus and Streptococcus mutans, respectively.


Subject(s)
Bacteriophages/physiology , Carbohydrates/physiology , Enzymes/metabolism , N-Acetylmuramoyl-L-alanine Amidase/metabolism , Peptidoglycan/metabolism , Streptococcus pyogenes/metabolism , Viral Proteins/metabolism , Carbohydrates/chemistry , Catalytic Domain , Cell Wall/chemistry , Cell Wall/metabolism , Enzymes/genetics , Hydrolysis , N-Acetylmuramoyl-L-alanine Amidase/genetics , Protein Conformation , Streptococcus pyogenes/genetics , Streptococcus pyogenes/growth & development , Viral Proteins/genetics
10.
R Soc Open Sci ; 8(3): 201991, 2021 Mar 10.
Article in English | MEDLINE | ID: mdl-33959354

ABSTRACT

Streptococcus pyogenes, also known as Group A Streptococcus (GAS), is a Gram-positive human-exclusive pathogen, responsible for more than 500 000 deaths annually worldwide. Upon infection, GAS commonly triggers mild symptoms such as pharyngitis, pyoderma and fever. However, recurrent infections or prolonged exposure to GAS might lead to life-threatening conditions. Necrotizing fasciitis, streptococcal toxic shock syndrome and post-immune mediated diseases, such as poststreptococcal glomerulonephritis, acute rheumatic fever and rheumatic heart disease, contribute to very high mortality rates in non-industrialized countries. Though an initial reduction in GAS infections was observed in high-income countries, global outbreaks of GAS, causing rheumatic fever and acute poststreptococcal glomerulonephritis, have been reported over the last decade. At the same time, our understanding of GAS pathogenesis and transmission has vastly increased, with detailed insight into the various stages of infection, beginning with adhesion, colonization and evasion of the host immune system. Despite deeper knowledge of the impact of GAS on the human body, the development of a successful vaccine for prophylaxis of GAS remains outstanding. In this review, we discuss the challenges involved in identifying a universal GAS vaccine and describe several potential vaccine candidates that we believe warrant pursuit.

11.
Nat Chem Biol ; 17(8): 878-887, 2021 08.
Article in English | MEDLINE | ID: mdl-34045745

ABSTRACT

In ovoid-shaped, Gram-positive bacteria, MapZ guides FtsZ-ring positioning at cell equators. The cell wall of the ovococcus Streptococcus mutans contains peptidoglycan decorated with serotype c carbohydrates (SCCs). In the present study, we identify the major cell separation autolysin AtlA as an SCC-binding protein. AtlA binding to SCC is attenuated by the glycerol phosphate (GroP) modification. Using fluorescently labeled AtlA constructs, we mapped SCC distribution on the streptococcal surface, revealing enrichment of GroP-deficient immature SCCs at the cell poles and equators. The immature SCCs co-localize with MapZ at the equatorial rings throughout the cell cycle. In GroP-deficient mutants, AtlA is mislocalized, resulting in dysregulated cellular autolysis. These mutants display morphological abnormalities associated with MapZ mislocalization, leading to FtsZ-ring misplacement. Altogether, our data support a model in which maturation of a cell wall polysaccharide provides the molecular cues for the recruitment of cell division machinery, ensuring proper daughter cell separation and FtsZ-ring positioning.


Subject(s)
Cell Wall/metabolism , Polysaccharides/metabolism , Streptococcus mutans/metabolism , Cell Division , Cell Wall/chemistry , Polysaccharides/chemistry , Streptococcus mutans/cytology
12.
Biochem J ; 478(4): 685-701, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33599745

ABSTRACT

Rhamnose is an important 6-deoxy sugar present in many natural products, glycoproteins, and structural polysaccharides. Whilst predominantly found as the l-enantiomer, instances of d-rhamnose are also found in nature, particularly in the Pseudomonads bacteria. Interestingly, rhamnose is notably absent from humans and other animals, which poses unique opportunities for drug discovery targeted towards rhamnose utilizing enzymes from pathogenic bacteria. Whilst the biosynthesis of nucleotide-activated rhamnose (NDP-rhamnose) is well studied, the study of rhamnosyltransferases that synthesize rhamnose-containing glycoconjugates is the current focus amongst the scientific community. In this review, we describe where rhamnose has been found in nature, as well as what is known about TDP-ß-l-rhamnose, UDP-ß-l-rhamnose, and GDP-α-d-rhamnose biosynthesis. We then focus on examples of rhamnosyltransferases that have been characterized using both in vivo and in vitro approaches from plants and bacteria, highlighting enzymes where 3D structures have been obtained. The ongoing study of rhamnose and rhamnosyltransferases, in particular in pathogenic organisms, is important to inform future drug discovery projects and vaccine development.


Subject(s)
Glycoconjugates/biosynthesis , Hexosyltransferases/physiology , Rhamnose/biosynthesis , Uridine Diphosphate Sugars/biosynthesis , Arabidopsis Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Capsid/metabolism , Eukaryotic Cells/metabolism , Flavonoids/metabolism , Glycoconjugates/chemistry , Glycolipids/biosynthesis , Glycosylation , Gram-Negative Bacteria/metabolism , Gram-Negative Bacteria/pathogenicity , Gram-Positive Bacteria/metabolism , Gram-Positive Bacteria/pathogenicity , Hexosyltransferases/chemistry , Hexosyltransferases/genetics , Models, Molecular , O Antigens/metabolism , Plant Proteins/metabolism , Polysaccharides, Bacterial/metabolism , Prokaryotic Cells/metabolism , Protein Conformation , Protein Processing, Post-Translational , Viral Proteins/metabolism , Virulence
13.
Cell Surf ; 6: 100047, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33294751

ABSTRACT

Despite the importance of fungal cell walls as the principle determinant of fungal morphology and the defining element determining fungal interactions with other cells, few scalar models have been developed that reconcile chemical and microscopic attributes of its structure. The cell wall of the fungal pathogen Candida albicans is comprised of an amorphous inner skeletal layer of ß(1,3)- and ß(1,6)-glucan and chitin and an outer fibrillar layer thought to be dominated by highly mannosylated cell wall proteins. The architecture of these two layers can be resolved at the electron microscopy level, but the visualised structure of the wall has not yet been defined precisely in chemical terms. We have therefore examined the precise structure, location and molecular sizes of the cell wall components using transmission electron microscopy and tomography and tested predictions of the cell wall models using mutants and agents that perturb the normal cell wall structure. We demonstrate that the fibrils are comprised of a frond of N-linked outer chain mannans linked to a basal layer of GPI-proteins concentrated in the mid-wall region and that the non-elastic chitin microfibrils are cantilevered with sufficient lengths of non-fibrillar chitin and/or ß-glucan to enable the chitin-glucan cage to flex, e.g. during morphogenesis and osmotic swelling. We present the first three-dimensional nano-scalar model of the C. albicans cell wall which can be used to test hypotheses relating to the structure-function relationships that underpin the pathobiology of this fungal pathogen.

14.
J Biol Chem ; 294(42): 15237-15256, 2019 10 18.
Article in English | MEDLINE | ID: mdl-31506299

ABSTRACT

Group A carbohydrate (GAC) is a bacterial peptidoglycan-anchored surface rhamnose polysaccharide (RhaPS) that is essential for growth of Streptococcus pyogenes and contributes to its ability to infect the human host. In this study, using molecular and synthetic biology approaches, biochemistry, radiolabeling techniques, and NMR and MS analyses, we examined the role of GacB, encoded in the S. pyogenes GAC gene cluster, in the GAC biosynthesis pathway. We demonstrate that GacB is the first characterized α-d-GlcNAc-ß-1,4-l-rhamnosyltransferase that synthesizes the committed step in the biosynthesis of the GAC virulence determinant. Importantly, the substitution of S. pyogenes gacB with the homologous gene from Streptococcus agalactiae (Group B Streptococcus), Streptococcus equi subsp. zooepidemicus (Group C Streptococcus), Streptococcus dysgalactiae subsp. equisimilis (Group G Streptococcus), or Streptococcus mutans complemented the GAC biosynthesis pathway. These results, combined with those from extensive in silico studies, reveal a common phylogenetic origin of the genes required for this priming step in >40 pathogenic species of the Streptococcus genus, including members from the Lancefield Groups B, C, D, E, G, and H. Importantly, this priming step appears to be unique to streptococcal ABC transporter-dependent RhaPS biosynthesis, whereas the Wzx/Wzy-dependent streptococcal capsular polysaccharide pathways instead require an α-d-Glc-ß-1,4-l-rhamnosyltransferase. The insights into the RhaPS priming step obtained here open the door to targeting the early steps of the group carbohydrate biosynthesis pathways in species of the Streptococcus genus of high clinical and veterinary importance.


Subject(s)
Antigens, Bacterial/biosynthesis , Bacterial Proteins/metabolism , Hexosyltransferases/metabolism , Polysaccharides, Bacterial/biosynthesis , Streptococcal Infections/microbiology , Streptococcus pyogenes/enzymology , Bacterial Proteins/genetics , Hexosyltransferases/genetics , Multigene Family , Phylogeny , Polysaccharides, Bacterial/genetics , Rhamnose/metabolism , Streptococcus/classification , Streptococcus/enzymology , Streptococcus/genetics , Streptococcus pyogenes/genetics , Streptococcus pyogenes/metabolism
15.
J Biol Chem ; 294(23): 9172-9185, 2019 06 07.
Article in English | MEDLINE | ID: mdl-31010825

ABSTRACT

The 6-deoxy sugar l-rhamnose (l-Rha) is found widely in plant and microbial polysaccharides and natural products. The importance of this and related compounds in host-pathogen interactions often means that l-Rha plays an essential role in many organisms. l-Rha is most commonly biosynthesized as the activated sugar nucleotide uridine 5'-diphospho-ß-l-rhamnose (UDP-ß-l-Rha) or thymidine 5'-diphospho-ß-l-rhamnose (TDP-ß-l-Rha). Enzymes involved in the biosynthesis of these sugar nucleotides have been studied in some detail in bacteria and plants, but the activated form of l-Rha and the corresponding biosynthetic enzymes have yet to be explored in algae. Here, using sugar-nucleotide profiling in two representative algae, Euglena gracilis and the toxin-producing microalga Prymnesium parvum, we show that levels of UDP- and TDP-activated l-Rha differ significantly between these two algal species. Using bioinformatics and biochemical methods, we identified and characterized a fusion of the RmlC and RmlD proteins, two bacteria-like enzymes involved in TDP-ß-l-Rha biosynthesis, from P. parvum Using this new sequence and also others, we explored l-Rha biosynthesis among algae, finding that although most algae contain sequences orthologous to plant-like l-Rha biosynthesis machineries, instances of the RmlC-RmlD fusion protein identified here exist across the Haptophyta and Gymnodiniaceae families of microalgae. On the basis of these findings, we propose potential routes for the evolution of nucleoside diphosphate ß-l-Rha (NDP-ß-l-Rha) pathways among algae.


Subject(s)
Algal Proteins/metabolism , Carbohydrate Epimerases/metabolism , Haptophyta/metabolism , Rhamnose/biosynthesis , Algal Proteins/genetics , Carbohydrate Epimerases/classification , Carbohydrate Epimerases/genetics , Phylogeny , Plastids/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Rhamnose/chemistry , Symbiosis
16.
Mol Microbiol ; 111(4): 951-964, 2019 04.
Article in English | MEDLINE | ID: mdl-30600561

ABSTRACT

Biosynthesis of the nucleotide sugar precursor dTDP-L-rhamnose is critical for the viability and virulence of many human pathogenic bacteria, including Streptococcus pyogenes (Group A Streptococcus; GAS), Streptococcus mutans and Mycobacterium tuberculosis. Streptococcal pathogens require dTDP-L-rhamnose for the production of structurally similar rhamnose polysaccharides in their cell wall. Via heterologous expression in S. mutans, we confirmed that GAS RmlB and RmlC are critical for dTDP-L-rhamnose biosynthesis through their action as dTDP-glucose-4,6-dehydratase and dTDP-4-keto-6-deoxyglucose-3,5-epimerase enzymes respectively. Complementation with GAS RmlB and RmlC containing specific point mutations corroborated the conservation of previous identified catalytic residues. Bio-layer interferometry was used to identify and confirm inhibitory lead compounds that bind to GAS dTDP-rhamnose biosynthesis enzymes RmlB, RmlC and GacA. One of the identified compounds, Ri03, inhibited growth of GAS, other rhamnose-dependent streptococcal pathogens as well as M. tuberculosis with an IC50 of 120-410 µM. Importantly, we confirmed that Ri03 inhibited dTDP-L-rhamnose formation in a concentration-dependent manner through a biochemical assay with recombinant rhamnose biosynthesis enzymes. We therefore conclude that inhibitors of dTDP-L-rhamnose biosynthesis, such as Ri03, affect streptococcal and mycobacterial viability and can serve as lead compounds for the development of a new class of antibiotics that targets dTDP-rhamnose biosynthesis in pathogenic bacteria.


Subject(s)
Anti-Bacterial Agents/isolation & purification , Hydro-Lyases/metabolism , Nucleoside Diphosphate Sugars/biosynthesis , Racemases and Epimerases/metabolism , Streptococcus/enzymology , Thymine Nucleotides/biosynthesis , Anti-Bacterial Agents/pharmacology , Biosynthetic Pathways , Hydro-Lyases/genetics , Inhibitory Concentration 50 , Racemases and Epimerases/genetics , Streptococcus/drug effects
17.
Mol Microbiol ; 98(5): 946-62, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26278404

ABSTRACT

The sugar nucleotide dTDP-L-rhamnose is critical for the biosynthesis of the Group A Carbohydrate, the molecular signature and virulence determinant of the human pathogen Group A Streptococcus (GAS). The final step of the four-step dTDP-L-rhamnose biosynthesis pathway is catalyzed by dTDP-4-dehydrorhamnose reductases (RmlD). RmlD from the Gram-negative bacterium Salmonella is the only structurally characterized family member and requires metal-dependent homo-dimerization for enzymatic activity. Using a biochemical and structural biology approach, we demonstrate that the only RmlD homologue from GAS, previously renamed GacA, functions in a novel monomeric manner. Sequence analysis of 213 Gram-negative and Gram-positive RmlD homologues predicts that enzymes from all Gram-positive species lack a dimerization motif and function as monomers. The enzymatic function of GacA was confirmed through heterologous expression of gacA in a S. mutans rmlD knockout, which restored attenuated growth and aberrant cell division. Finally, analysis of a saturated mutant GAS library using Tn-sequencing and generation of a conditional-expression mutant identified gacA as an essential gene for GAS. In conclusion, GacA is an essential monomeric enzyme in GAS and representative of monomeric RmlD enzymes in Gram-positive bacteria and a subset of Gram-negative bacteria. These results will help future screens for novel inhibitors of dTDP-L-rhamnose biosynthesis.


Subject(s)
Bacterial Proteins/metabolism , Carbohydrate Dehydrogenases/genetics , Carbohydrate Dehydrogenases/metabolism , Streptococcus pyogenes/enzymology , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Carbohydrate Dehydrogenases/chemistry , Carbohydrate Epimerases/metabolism , Cloning, Molecular , Gram-Positive Bacteria/enzymology , High-Throughput Nucleotide Sequencing , Humans , Models, Molecular , Mutation , Nucleoside Diphosphate Sugars/biosynthesis , Protein Structure, Tertiary , Rhamnose/analogs & derivatives , Rhamnose/biosynthesis , Rhamnose/metabolism , Sequence Alignment , Streptococcus pyogenes/genetics , Thymine Nucleotides/biosynthesis , Thymine Nucleotides/metabolism
18.
J Biol Chem ; 289(33): 23020-23028, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24942743

ABSTRACT

Chitin synthases (CHS) produce chitin, an essential component of the fungal cell wall. The molecular mechanism of processive chitin synthesis is not understood, limiting the discovery of new inhibitors of this enzyme class. We identified the bacterial glycosyltransferase NodC as an appropriate model system to study the general structure and reaction mechanism of CHS. A high throughput screening-compatible novel assay demonstrates that a known inhibitor of fungal CHS also inhibit NodC. A structural model of NodC, on the basis of the recently published BcsA cellulose synthase structure, enabled probing of the catalytic mechanism by mutagenesis, demonstrating the essential roles of the DD and QXXRW catalytic motifs. The NodC membrane topology was mapped, validating the structural model. Together, these approaches give insight into the CHS structure and mechanism and provide a platform for the discovery of inhibitors for this antifungal target.


Subject(s)
Bacterial Proteins , Chitin Synthase , Chitin , Models, Biological , Models, Molecular , Sinorhizobium meliloti/enzymology , Amino Acid Motifs , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Chitin/biosynthesis , Chitin/chemistry , Chitin Synthase/chemistry , Chitin Synthase/metabolism , Glucosyltransferases/chemistry , Glucosyltransferases/metabolism
19.
Biochem J ; 457(3): 497-502, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24256146

ABSTRACT

Inhibitors of OGT (O-GlcNAc transferase) are valuable tools to study the cell biology of protein O-GlcNAcylation. We report OGT bisubstrate-linked inhibitors (goblins) in which the acceptor serine in the peptide VTPVSTA is covalently linked to UDP, eliminating the GlcNAc pyranoside ring. Goblin1 co-crystallizes with OGT, revealing an ordered C3 linker and retained substrate-binding modes, and binds the enzyme with micromolar affinity, inhibiting glycosyltransfer on to protein and peptide substrates.


Subject(s)
Drug Design , Enzyme Inhibitors/pharmacology , Models, Molecular , N-Acetylglucosaminyltransferases/antagonists & inhibitors , Oligopeptides/pharmacology , Uridine Diphosphate/analogs & derivatives , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Binding Sites , Crystallography, X-Ray , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Glycosylation/drug effects , Humans , Interferometry , Kinetics , N-Acetylglucosaminyltransferases/chemistry , N-Acetylglucosaminyltransferases/metabolism , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Oligopeptides/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Processing, Post-Translational/drug effects , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Serine/chemistry , Uridine Diphosphate/chemistry , Uridine Diphosphate/metabolism , Uridine Diphosphate/pharmacology
20.
Open Biol ; 3(10): 130021, 2013 Oct 02.
Article in English | MEDLINE | ID: mdl-24088714

ABSTRACT

The dynamic modification of proteins by O-linked N-acetylglucosamine (O-GlcNAc) is an essential posttranslational modification present in higher eukaryotes. Removal of O-GlcNAc is catalysed by O-GlcNAcase, a multi-domain enzyme that has been reported to be bifunctional, possessing both glycoside hydrolase and histone acetyltransferase (AT) activity. Insights into the mechanism, protein substrate recognition and inhibition of the hydrolase domain of human OGA (hOGA) have been obtained via the use of the structures of bacterial homologues. However, the molecular basis of AT activity of OGA, which has only been reported in vitro, is not presently understood. Here, we describe the crystal structure of a putative acetyltransferase (OgpAT) that we identified in the genome of the marine bacterium Oceanicola granulosus, showing homology to the hOGA C-terminal AT domain (hOGA-AT). The structure of OgpAT in complex with acetyl coenzyme A (AcCoA) reveals that, by homology modelling, hOGA-AT adopts a variant AT fold with a unique loop creating a deep tunnel. The structures, together with mutagenesis and surface plasmon resonance data, reveal that while the bacterial OgpAT binds AcCoA, the hOGA-AT does not, as explained by the lack of key residues normally required to bind AcCoA. Thus, the C-terminal domain of hOGA is a catalytically incompetent 'pseudo'-AT.


Subject(s)
Acetylglucosamine/metabolism , Histone Acetyltransferases/chemistry , Histone Acetyltransferases/genetics , Rhodobacteraceae/enzymology , beta-N-Acetylhexosaminidases/chemistry , Acetyl Coenzyme A/chemistry , Acetyl Coenzyme A/metabolism , Acetylglucosamine/chemistry , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Histone Acetyltransferases/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Multifunctional Enzymes/chemistry , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Operon , Protein Binding , Protein Folding , Protein Processing, Post-Translational , Protein Structure, Secondary , Rhodobacteraceae/genetics , Sequence Alignment , Substrate Specificity , Surface Plasmon Resonance , beta-N-Acetylhexosaminidases/genetics , beta-N-Acetylhexosaminidases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...