Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Angew Chem Int Ed Engl ; 62(29): e202304989, 2023 07 17.
Article in English | MEDLINE | ID: mdl-37222528

ABSTRACT

The S-glycosyltransferase LmbT, involved in the biosynthesis of lincomycin A, is the only known enzyme that catalyzes the enzymatic incorporation of rare amino acid L-ergothioneine (EGT) into secondary metabolites. Here, we show the structure and function analyses of LmbT. Our in vitro analysis of LmbT revealed that the enzyme shows promiscuous substrate specificity toward nitrogenous base moieties in the generation of unnatural nucleotide diphosphate (NDP)-D-α-D-lincosamides. Furthermore, the X-ray crystal structures of LmbT in its apo form and in complex with substrates indicated that the large conformational changes of the active site occur upon binding of the substrates, and that EGT is strictly recognized by salt-bridge and cation-π interactions with Arg260 and Trp101, respectively. The structure of LmbT in complex with its substrates, the docking model with the EGT-S-conjugated lincosamide, and the structure-based site-directed mutagenesis analysis revealed the structural details of the LmbT-catalyzed SN 2-like S-glycosylation reaction with EGT.


Subject(s)
Anti-Bacterial Agents , Lincomycin , Glycosylation , Anti-Bacterial Agents/chemistry , Lincosamides/chemistry , Lincosamides/metabolism , Lincomycin/chemistry , Glycosyltransferases/metabolism , Crystallography, X-Ray
2.
Microbiol Res ; 265: 127186, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36155963

ABSTRACT

Here we provide a review on TldD/TldE family proteins, summarizing current knowledge and outlining further research perspectives. Despite being widely distributed in bacteria and archaea, TldD/TldE proteins have been escaping attention for a long time until several recent reports pointed to their unique features. Specifically, TldD/TldE generally act as peptidases, though some of them turned out to be N-deacetylases. Biological function of TldD/TldE has been extensively described in bacterial specialized metabolism, in which they participate in the biosynthesis of lincosamide antibiotics (as N-deacetylases), and in the biosynthesis of ribosomally synthesized and post-translationally modified bioactive peptides (as peptidases). These enzymes possess special position in the relevant biosynthesis since they convert non-bioactive intermediates into bioactive metabolites. Further, based on a recent study of Escherichia coli TldD/TldE, these heterodimeric metallopeptidases possess a new protein fold exhibiting several structural features with no precedent in the Protein Data Bank. The most interesting ones are structural elements forming metal-containing active site on the inner surface of the catalytically active subunit TldD, in which substrates bind through ß sheet interactions in the sequence-independent manner. It results in relaxed substrate specificity of TldD/TldE, which is counterbalanced by enclosing the active centre within the hollow core of the heterodimer and only appropriate substrates can entry through a narrow channel. Based on the published data, we hypothesize a yet unrecognized central metabolic function of TldD/TldE in the degradation of (partially) unfolded proteins, i.e., in protein quality control.


Subject(s)
Escherichia coli , Peptide Hydrolases , Anti-Bacterial Agents/metabolism , Bacteria/genetics , Bacteria/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Lincosamides/metabolism , Metalloproteases/metabolism , Peptide Hydrolases/metabolism , Peptides/chemistry
3.
Proc Natl Acad Sci U S A ; 117(40): 24794-24801, 2020 10 06.
Article in English | MEDLINE | ID: mdl-32958639

ABSTRACT

The structure of lincomycin A consists of the unusual eight-carbon thiosugar core methyllincosamide (MTL) decorated with a pendent N-methylprolinyl moiety. Previous studies on MTL biosynthesis have suggested GDP-ᴅ-erythro-α-ᴅ-gluco-octose and GDP-ᴅ-α-ᴅ-lincosamide as key intermediates in the pathway. However, the enzyme-catalyzed reactions resulting in the conversion of GDP-ᴅ-erythro-α-ᴅ-gluco-octose to GDP-ᴅ-α-ᴅ-lincosamide have not yet been elucidated. Herein, a biosynthetic subpathway involving the activities of four enzymes-LmbM, LmbL, CcbZ, and CcbS (the LmbZ and LmbS equivalents in the closely related celesticetin pathway)-is reported. These enzymes catalyze the previously unknown biosynthetic steps including 6-epimerization, 6,8-dehydration, 4-epimerization, and 6-transamination that convert GDP-ᴅ-erythro-α-ᴅ-gluco-octose to GDP-ᴅ-α-ᴅ-lincosamide. Identification of these reactions completes the description of the entire lincomycin biosynthetic pathway. This work is significant since it not only resolves the missing link in octose core assembly of a thiosugar-containing natural product but also showcases the sophistication in catalytic logic of enzymes involved in carbohydrate transformations.


Subject(s)
Lincomycin/biosynthesis , Streptomyces/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biosynthetic Pathways , Lincomycin/chemistry , Lincosamides/chemistry , Lincosamides/metabolism , Streptomyces/chemistry , Streptomyces/enzymology , Streptomyces/genetics
4.
Prep Biochem Biotechnol ; 50(6): 529-537, 2020.
Article in English | MEDLINE | ID: mdl-31916478

ABSTRACT

As a lincosamide antibiotic, lincomycin is still important for treating diseases caused by Gram-positive bacteria. Manufacturing of lincomycin needs efforts to, e.g. maximize desirable species and minimizing unwanted fermentation byproducts. Analysis of the lincomycin biosynthetic gene cluster of Streptomyces lincolnensis, lmbB1, was shown to catalyze the conversion of L-dopa but not of L-tyrosine and then further generated the precursor of lincomycin A. Based on the principle of directed breeding, a strain termed as S. lincolnensis 24-2, was obtained in this work. By overexpressing the lmbB1 gene, this strain produces efficacious lincomycin A and suppresses melanin generation, whereas contains unwanted lincomycin B. The good fermentation performance of the mutant-lmbB1 (M-lmbB1) was also confirmed in a 15 L-scale bioreactor, which increased the lincomycin A production by 37.6% compared with control of 6435 u/mL and reduced the accumulation of melanin by 29.9% and lincomycin B by 73.4%. This work demonstrated that the amplification of lmbB1 gene mutation and metabolic engineering could promote lincomycin biosynthesis and might be helpful for reducing the production of other industrially unnecessary byproduct.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Bacterial Proteins/biosynthesis , Fermentation/genetics , Lincomycin/biosynthesis , Metabolic Engineering/methods , Multigene Family , Streptomyces/genetics , Streptomyces/metabolism , Bioreactors , Escherichia coli/genetics , Gene Expression , Levodopa/metabolism , Lincosamides/metabolism , Melanins/biosynthesis , Microorganisms, Genetically-Modified , Transcription, Genetic/genetics
5.
Chemosphere ; 193: 611-617, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29169137

ABSTRACT

Lincomycin mycelial residue (LMR) is the restricted resource because it contains residual lincomycin, which is producing potential risks to the environment and human health. In this study, lincomycin-degrading strain LCM-B was isolated and identified as Clostridium sp. in the LMR. Strain LCM-B was able to degrade 62.03% of lincomycin at the initial concentration of 100 mg L-1 after incubation for 10 d, while only 15.61% of lincomycin was removed at the initial concentration of 500 mg L-1. The removal efficiency of lincomycin by strain LCM-B decreased as the initial concentration increased. Gene lnuB (which encodes the nucleotidyl transferase) was detected in the isolated strain, and it was proven to participate in lincomycin biodegradation based on the analysis of degradation products and pathway. The results provide a relatively complete understanding of lincomycin biodegradation mechanism. Strain LCM-B is promising to eliminate lincomycin from the LMR.


Subject(s)
Anti-Bacterial Agents/metabolism , Biodegradation, Environmental , Clostridium/physiology , Lincomycin/metabolism , Clostridium/isolation & purification , Humans , Lincosamides/metabolism
6.
J Mol Biol ; 427(12): 2229-43, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25900373

ABSTRACT

One of the main mechanisms of resistance to lincosamide and aminoglycoside antibiotics is their inactivation by O-nucleotidylyltransferases (NTases). Significant sequence variation of lincomycin nucleotidylyltransferase (Lnu) and aminoglycoside nucleotidylyltransferase (ANT) enzymes plus lack of detailed information about the molecular basis for specificity of these enzymes toward chemically distinct antibiotic scaffolds hinders development of a general strategy to curb this resistance mechanism. We conducted an extensive sequence analysis identifying 129 putative antibiotic NTases constituting six distinct subfamilies represented by Lnu(A), Lnu(B), Lnu(C), Lnu(D), Lnu(F)/(G) plus ANT(2") enzymes. Since only the Lnu(B) enzyme has been previously studied in detail, we biochemically characterized the Lnu(A) and Lnu(D) enzymes, with the former representing the most sequence distinct Lnu ortholog. We also determined the crystal structure of the Lnu(A) enzyme in complex with a lincosamide. These data suggested that, while sharing the N-terminal nucleotidylyltransferase domain, the groups of antibiotic NTases feature structurally distinct C-terminal domains (CTDs) adapted to accommodate antibiotics. Comparative structural analysis among antibiotic NTases rationalized their specificity toward lincosamides versus aminoglycosides through active-site plasticity, which allows retention of general catalytic activity while accepting alterations at multiple, specific positions contributed by both domains. Based on this structural analysis, we suggest that antibiotic NTases evolved from an ancestral nucleotidylyltransferase along independent paths according to the identified groups, characterized by structural changes in the active site and recruitment of structurally diverse CTDs. These data show the complexity of enzyme-driven antibiotic resistance and provide a basis for broadly active inhibitors by identifying the key unifying features of antibiotic NTases.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Drug Resistance, Bacterial , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Catalytic Domain , Cluster Analysis , Crystallography, X-Ray , Lincosamides/chemistry , Lincosamides/metabolism , Molecular Sequence Data , Nucleotidyltransferases/genetics , Phylogeny , Protein Binding , Protein Conformation , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity
7.
Nature ; 518(7537): 115-9, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25607359

ABSTRACT

Low-molecular-mass thiols in organisms are well known for their redox-relevant role in protection against various endogenous and exogenous stresses. In eukaryotes and Gram-negative bacteria, the primary thiol is glutathione (GSH), a cysteinyl-containing tripeptide. In contrast, mycothiol (MSH), a cysteinyl pseudo-disaccharide, is dominant in Gram-positive actinobacteria, including antibiotic-producing actinomycetes and pathogenic mycobacteria. MSH is equivalent to GSH, either as a cofactor or as a substrate, in numerous biochemical processes, most of which have not been characterized, largely due to the dearth of information concerning MSH-dependent proteins. Actinomycetes are able to produce another thiol, ergothioneine (EGT), a histidine betaine derivative that is widely assimilated by plants and animals for variable physiological activities. The involvement of EGT in enzymatic reactions, however, lacks any precedent. Here we report that the unprecedented coupling of two bacterial thiols, MSH and EGT, has a constructive role in the biosynthesis of lincomycin A, a sulfur-containing lincosamide (C8 sugar) antibiotic that has been widely used for half a century to treat Gram-positive bacterial infections. EGT acts as a carrier to template the molecular assembly, and MSH is the sulfur donor for lincomycin maturation after thiol exchange. These thiols function through two unusual S-glycosylations that program lincosamide transfer, activation and modification, providing the first paradigm for EGT-associated biochemical processes and for the poorly understood MSH-dependent biotransformations, a newly described model that is potentially common in the incorporation of sulfur, an element essential for life and ubiquitous in living systems.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Cysteine/metabolism , Ergothioneine/metabolism , Glycopeptides/metabolism , Inositol/metabolism , Lincomycin/biosynthesis , Streptomyces/metabolism , Biological Products/metabolism , Biosynthetic Pathways/genetics , Biotransformation , Cysteine/chemistry , Ergothioneine/chemistry , Glycopeptides/chemistry , Glycosylation , Inositol/chemistry , Lincosamides/metabolism , Molecular Sequence Data , Streptomyces/genetics
8.
J Photochem Photobiol B ; 138: 324-30, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-25033467

ABSTRACT

The interaction of clindamycin phosphate (CP) with bovine serum albumin (BSA) is studied by using fluorescence spectra, UV-visible absorption, synchronous fluorescence spectra (SFS), CD, 3D fluorescence spectra and lifetime measurements under simulated physiological conditions. CP effectively quenched intrinsic fluorescence of BSA. The binding constants KA values are 2.540×10(5), 4.960×10(5), 7.207×10(5) L mol(-1), the number of binding sites n and corresponding thermodynamic parameters ΔG(o), ΔH(o) and ΔS(o) between CP and BSA were calculated at different temperatures. The interaction between CP and BSA occurs through dynamic quenching and the effect of CP on the conformation of BSA was also analyzed using SFS. The average binding distance r between the donor (BSA) and acceptor (CP) was determined based on Förster's theory. The results of fluorescence spectra, UV-vis absorption spectra and SFS show that the secondary structure of the protein has been changed in the presence of CP.


Subject(s)
Anti-Bacterial Agents/chemistry , Clindamycin/analogs & derivatives , Metals/chemistry , Serum Albumin, Bovine/chemistry , Animals , Anti-Bacterial Agents/metabolism , Binding Sites , Cattle , Clindamycin/chemistry , Clindamycin/metabolism , Energy Transfer , Ions/chemistry , Kinetics , Lincosamides/chemistry , Lincosamides/metabolism , Metals/metabolism , Protein Binding , Serum Albumin, Bovine/metabolism , Thermodynamics
10.
Food Microbiol ; 30(1): 59-67, 2012 May.
Article in English | MEDLINE | ID: mdl-22265284

ABSTRACT

Enterococcus faecalis UGRA10, a new AS-48-producer strain, has been isolated from a Spanish sheep's cheese. The inhibitory substance produced by E. faecalis UGRA10 was purified and characterized using matrix-assisted laser desorption ionization-time of flight mass spectrometry, confirming its identity with AS-48 enterocin (7.150 Da). Subsequent genetic analysis showed the existence of the as-48 gene cluster on a plasmid of approximately 70-kb. The UGRA10 strain was examined for safety properties such as enterococci virulence genes, biogenic amine production, and antibiotic resistance. As for most E. faecalis strains, PCR amplification revealed the existence of gene encoding for GelE, Asa1, Esp, EfaA, and Ace antigens and for tyrosine decarboxylase. This strain was sensitive to most of the antibiotics tested, being resistant only to aminoglycosides, lincosamide, and pristinamicins. In addition, UGRA10 developed an ability to form biofilms and to adhere to Caco 2 and HeLa 229 cells. More interestingly, this strain shows a high ability to interfere with the adhesion of Listeria monocytogenes to Caco 2 cells. Altogether, the results suggest that this broad-spectrum bacteriocin-producing strain has biotechnological potential to be developed as a protective agent in food preservation and as a probiotic.


Subject(s)
Bacteriocins/isolation & purification , Cheese/microbiology , DNA, Bacterial/isolation & purification , Enterococcus faecalis/isolation & purification , Probiotics/metabolism , Aminoglycosides/metabolism , Animals , Bacterial Adhesion , Bacteriocins/pharmacology , Caco-2 Cells , Cheese/standards , Consumer Product Safety , Drug Resistance, Bacterial/drug effects , Enterococcus faecalis/genetics , Enterococcus faecalis/metabolism , Food Contamination/analysis , Food Microbiology/methods , HeLa Cells , Humans , Lincosamides/metabolism , Listeria monocytogenes/pathogenicity , Microbial Sensitivity Tests , Multigene Family , Sheep , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL