Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.349
Filter
1.
Nature ; 613(7945): 729-734, 2023 01.
Article in English | MEDLINE | ID: mdl-36450357

ABSTRACT

Peptidoglycan and almost all surface glycopolymers in bacteria are built in the cytoplasm on the lipid carrier undecaprenyl phosphate (UndP)1-4. These UndP-linked precursors are transported across the membrane and polymerized or directly transferred to surface polymers, lipids or proteins. UndP is then flipped to regenerate the pool of cytoplasmic-facing UndP. The identity of the flippase that catalyses transport has remained unknown. Here, using the antibiotic amphomycin that targets UndP5-7, we identified two broadly conserved protein families that affect UndP recycling. One (UptA) is a member of the DedA superfamily8; the other (PopT) contains the domain DUF368. Genetic, cytological and syntenic analyses indicate that these proteins are UndP transporters. Notably, homologues from Gram-positive and Gram-negative bacteria promote UndP transport in Bacillus subtilis, indicating that recycling activity is broadly conserved among family members. Inhibitors of these flippases could potentiate the activity of antibiotics targeting the cell envelope.


Subject(s)
Bacterial Proteins , Carrier Proteins , Conserved Sequence , Evolution, Molecular , Gram-Negative Bacteria , Gram-Positive Bacteria , Polyisoprenyl Phosphates , Anti-Bacterial Agents/pharmacology , Bacillus subtilis/cytology , Bacillus subtilis/drug effects , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/classification , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Carrier Proteins/chemistry , Carrier Proteins/classification , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Gram-Negative Bacteria/cytology , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/genetics , Gram-Negative Bacteria/metabolism , Gram-Positive Bacteria/cytology , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/genetics , Gram-Positive Bacteria/metabolism , Polyisoprenyl Phosphates/metabolism , Synteny , Peptidoglycan/metabolism , Cell Wall/chemistry , Cell Wall/metabolism
2.
Nature ; 590(7846): 463-467, 2021 02.
Article in English | MEDLINE | ID: mdl-33536618

ABSTRACT

Actinobacteria produce numerous antibiotics and other specialized metabolites that have important applications in medicine and agriculture1. Diffusible hormones frequently control the production of such metabolites by binding TetR family transcriptional repressors (TFTRs), but the molecular basis for this remains unclear2. The production of methylenomycin antibiotics in Streptomyces coelicolor A3(2) is initiated by the binding of 2-alkyl-4-hydroxymethylfuran-3-carboxylic acid (AHFCA) hormones to the TFTR MmfR3. Here we report the X-ray crystal structure of an MmfR-AHFCA complex, establishing the structural basis for hormone recognition. We also elucidate the mechanism for DNA release upon hormone binding through the single-particle cryo-electron microscopy structure of an MmfR-operator complex. DNA binding and release assays with MmfR mutants and synthetic AHFCA analogues define the role of individual amino acid residues and hormone functional groups in ligand recognition and DNA release. These findings will facilitate the exploitation of actinobacterial hormones and their associated TFTRs in synthetic biology and in the discovery of new antibiotics.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Furans/metabolism , Streptomyces coelicolor/metabolism , Apoproteins/chemistry , Apoproteins/metabolism , Apoproteins/ultrastructure , Bacterial Proteins/chemistry , Bacterial Proteins/classification , Bacterial Proteins/metabolism , Bacterial Proteins/ultrastructure , Cryoelectron Microscopy , Crystallography, X-Ray , DNA/chemistry , DNA/genetics , DNA/metabolism , DNA/ultrastructure , Furans/chemistry , Hormones/chemistry , Hormones/classification , Hormones/metabolism , Ligands , Models, Molecular , Peptides/metabolism , Repressor Proteins/chemistry , Repressor Proteins/classification , Repressor Proteins/metabolism , Repressor Proteins/ultrastructure , Signal Transduction , Streptomyces coelicolor/chemistry , Streptomyces coelicolor/genetics , Structure-Activity Relationship
3.
Mol Cell ; 68(1): 15-25, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28985502

ABSTRACT

CRISPR-Cas systems defend prokaryotes against bacteriophages and mobile genetic elements and serve as the basis for revolutionary tools for genetic engineering. Class 2 CRISPR-Cas systems use single Cas endonucleases paired with guide RNAs to cleave complementary nucleic acid targets, enabling programmable sequence-specific targeting with minimal machinery. Recent discoveries of previously unidentified CRISPR-Cas systems have uncovered a deep reservoir of potential biotechnological tools beyond the well-characterized Type II Cas9 systems. Here we review the current mechanistic understanding of newly discovered single-protein Cas endonucleases. Comparison of these Cas effectors reveals substantial mechanistic diversity, underscoring the phylogenetic divergence of related CRISPR-Cas systems. This diversity has enabled further expansion of CRISPR-Cas biotechnological toolkits, with wide-ranging applications from genome editing to diagnostic tools based on various Cas endonuclease activities. These advances highlight the exciting prospects for future tools based on the continually expanding set of CRISPR-Cas systems.


Subject(s)
Bacterial Proteins/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Endonucleases/genetics , Gene Editing/methods , Genome , Bacteria/genetics , Bacteria/immunology , Bacteria/virology , Bacterial Proteins/chemistry , Bacterial Proteins/classification , Bacterial Proteins/metabolism , Bacteriophages/growth & development , Endonucleases/chemistry , Endonucleases/classification , Endonucleases/metabolism , Genetic Engineering , Humans , Models, Molecular , Protein Conformation , Protein Domains , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , Transcription, Genetic
4.
Proc Natl Acad Sci U S A ; 119(28): e2118260119, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35763567

ABSTRACT

Type VI CRISPR-Cas systems have been repurposed for various applications such as gene knockdown, viral interference, and diagnostics. However, the identification and characterization of thermophilic orthologs will expand and unlock the potential of diverse biotechnological applications. Herein, we identified and characterized a thermostable ortholog of the Cas13a family from the thermophilic organism Thermoclostridium caenicola (TccCas13a). We show that TccCas13a has a close phylogenetic relation to the HheCas13a ortholog from the thermophilic bacterium Herbinix hemicellulosilytica and shares several properties such as thermostability and inability to process its own pre-CRISPR RNA. We demonstrate that TccCas13a possesses robust cis and trans activities at a broad temperature range of 37 to 70 °C, compared with HheCas13a, which has a more limited range and lower activity. We harnessed TccCas13a thermostability to develop a sensitive, robust, rapid, and one-pot assay, named OPTIMA-dx, for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) detection. OPTIMA-dx exhibits no cross-reactivity with other viruses and a limit of detection of 10 copies/µL when using a synthetic SARS-CoV-2 genome. We used OPTIMA-dx for SARS-CoV-2 detection in clinical samples, and our assay showed 95% sensitivity and 100% specificity compared with qRT-PCR. Furthermore, we demonstrated that OPTIMA-dx is suitable for multiplexed detection and is compatible with the quick extraction protocol. OPTIMA-dx exhibits critical features that enable its use at point of care (POC). Therefore, we developed a mobile phone application to facilitate OPTIMA-dx data collection and sharing of patient sample results. This work demonstrates the power of CRISPR-Cas13 thermostable enzymes in enabling key applications in one-pot POC diagnostics and potentially in transcriptome engineering, editing, and therapies.


Subject(s)
Bacterial Proteins , COVID-19 , CRISPR-Associated Proteins , Clostridiales , Endodeoxyribonucleases , Point-of-Care Testing , SARS-CoV-2 , Bacterial Proteins/chemistry , Bacterial Proteins/classification , Bacterial Proteins/genetics , Biotechnology , COVID-19/diagnosis , CRISPR-Associated Proteins/chemistry , CRISPR-Associated Proteins/classification , CRISPR-Associated Proteins/genetics , Clostridiales/enzymology , Endodeoxyribonucleases/chemistry , Endodeoxyribonucleases/classification , Endodeoxyribonucleases/genetics , Enzyme Stability , Hot Temperature , Humans , Phylogeny , SARS-CoV-2/isolation & purification
5.
Proc Natl Acad Sci U S A ; 119(38): e2203593119, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36095213

ABSTRACT

Outer membrane porins in Gram-negative bacteria facilitate antibiotic influx. In Klebsiella pneumoniae, modifications in the porin OmpK36 are implicated in increasing resistance to carbapenems. An analysis of large K. pneumoniae genome collections, encompassing major healthcare-associated clones, revealed the recurrent emergence of a synonymous cytosine-to-thymine transition at position 25 (25c > t) in ompK36. We show that the 25c > t transition increases carbapenem resistance through depletion of OmpK36 from the outer membrane. The mutation attenuates K. pneumoniae in a murine pneumonia model, which accounts for its limited clonal expansion observed by phylogenetic analysis. However, in the context of carbapenem treatment, the 25c > t transition tips the balance toward treatment failure, thus accounting for its recurrent emergence. Mechanistically, the 25c > t transition mediates an intramolecular messenger RNA (mRNA) interaction between a uracil encoded by 25t and the first adenine within the Shine-Dalgarno sequence. This specific interaction leads to the formation of an RNA stem structure, which obscures the ribosomal binding site thus disrupting translation. While mutations reducing OmpK36 expression via transcriptional silencing are known, we uniquely demonstrate the repeated selection of a synonymous ompK36 mutation mediating translational suppression in response to antibiotic pressure.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Carbapenems , Klebsiella pneumoniae , Porins , beta-Lactam Resistance , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/classification , Bacterial Proteins/genetics , Carbapenems/pharmacology , Carbapenems/therapeutic use , Disease Models, Animal , Klebsiella Infections/drug therapy , Klebsiella Infections/microbiology , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/genetics , Mice , Microbial Sensitivity Tests , Mutation , Phylogeny , Pneumonia, Bacterial/drug therapy , Pneumonia, Bacterial/microbiology , Porins/classification , Porins/genetics , RNA, Messenger/metabolism , beta-Lactam Resistance/genetics
6.
J Biol Chem ; 299(7): 104902, 2023 07.
Article in English | MEDLINE | ID: mdl-37302554

ABSTRACT

Fusobacterium nucleatum is an opportunistic oral pathogen that is associated with various cancers. To fulfill its essential need for iron, this anaerobe will express heme uptake machinery encoded at a single genetic locus. The heme uptake operon includes HmuW, a class C radical SAM-dependent methyltransferase that degrades heme anaerobically to release Fe2+ and a linear tetrapyrrole called anaerobilin. The last gene in the operon, hmuF encodes a member of the flavodoxin superfamily of proteins. We discovered that HmuF and a paralog, FldH, bind tightly to both FMN and heme. The structure of Fe3+-heme-bound FldH (1.6 Å resolution) reveals a helical cap domain appended to the ⍺/ß core of the flavodoxin fold. The cap creates a hydrophobic binding cleft that positions the heme planar to the si-face of the FMN isoalloxazine ring. The ferric heme iron is hexacoordinated to His134 and a solvent molecule. In contrast to flavodoxins, FldH and HmuF do not stabilize the FMN semiquinone but instead cycle between the FMN oxidized and hydroquinone states. We show that heme-loaded HmuF and heme-loaded FldH traffic heme to HmuW for degradation of the protoporphyrin ring. Both FldH and HmuF then catalyze multiple reductions of anaerobilin through hydride transfer from the FMN hydroquinone. The latter activity eliminates the aromaticity of anaerobilin and the electrophilic methylene group that was installed through HmuW turnover. Hence, HmuF provides a protected path for anaerobic heme catabolism, offering F. nucleatum a competitive advantage in the colonization of anoxic sites of the human body.


Subject(s)
Flavodoxin , Fusobacterium nucleatum , Heme , Tetrapyrroles , Humans , Flavin Mononucleotide/metabolism , Flavodoxin/chemistry , Flavodoxin/classification , Flavodoxin/genetics , Flavodoxin/metabolism , Fusobacterium nucleatum/chemistry , Fusobacterium nucleatum/genetics , Fusobacterium nucleatum/metabolism , Heme/metabolism , Iron/metabolism , Oxidation-Reduction , Tetrapyrroles/metabolism , Biological Transport , Genes, Bacterial , Bacterial Proteins/chemistry , Bacterial Proteins/classification , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Protein Domains , Fusobacterium Infections/microbiology
7.
Nature ; 557(7706): 503-509, 2018 05.
Article in English | MEDLINE | ID: mdl-29769716

ABSTRACT

One-third of all protein-coding genes from bacterial genomes cannot be annotated with a function. Here, to investigate the functions of these genes, we present genome-wide mutant fitness data from 32 diverse bacteria across dozens of growth conditions. We identified mutant phenotypes for 11,779 protein-coding genes that had not been annotated with a specific function. Many genes could be associated with a specific condition because the gene affected fitness only in that condition, or with another gene in the same bacterium because they had similar mutant phenotypes. Of the poorly annotated genes, 2,316 had associations that have high confidence because they are conserved in other bacteria. By combining these conserved associations with comparative genomics, we identified putative DNA repair proteins; in addition, we propose specific functions for poorly annotated enzymes and transporters and for uncharacterized protein families. Our study demonstrates the scalability of microbial genetics and its utility for improving gene annotations.


Subject(s)
Bacteria/genetics , Genes, Bacterial/genetics , Molecular Sequence Annotation , Mutation , Phenotype , Uncertainty , Bacteria/cytology , Bacterial Proteins/classification , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Conserved Sequence , DNA Repair/genetics , Genetic Fitness , Genome, Bacterial/genetics , Mutant Proteins/classification , Mutant Proteins/genetics , Mutant Proteins/physiology
8.
Nucleic Acids Res ; 50(D1): D848-D857, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34551435

ABSTRACT

From industry to food to health, bacteria play an important role in all facets of life. Some of the most important bacteria have been purposely engineered to produce commercial quantities of antibiotics and therapeutics, and non-classical secretion systems are at the forefront of these technologies. Unlike the classical Sec or Tat pathways, non-classically secreted proteins share few common characteristics and use much more diverse secretion pathways for protein transport. Systematically categorizing and investigating the non-classically secreted proteins will enable a deeper understanding of their associated secretion mechanisms and provide a landscape of the Gram-positive secretion pathway distribution. We therefore developed PncsHub (https://pncshub.erc.monash.edu/), the first universal platform for comprehensively annotating and analyzing Gram-positive bacterial non-classically secreted proteins. PncsHub catalogs 4,914 non-classically secreted proteins, which are delicately categorized into 8 subtypes (including the 'unknown' subtype) and annotated with data compiled from up to 26 resources and visualisation tools. It incorporates state-of-the-art predictors to identify new and homologous non-classically secreted proteins and includes three analytical modules to visualise the relationships between known and putative non-classically secreted proteins. As such, PncsHub aims to provide integrated services for investigating, predicting and identifying non-classically secreted proteins to promote hypothesis-driven laboratory-based experiments.


Subject(s)
Bacterial Proteins/genetics , Databases, Protein , Gram-Positive Bacteria/genetics , User-Computer Interface , Bacterial Proteins/classification , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Gram-Positive Bacteria/classification , Gram-Positive Bacteria/metabolism , Internet , Molecular Sequence Annotation , Phylogeny , Protein Transport
9.
Nucleic Acids Res ; 50(D1): D560-D570, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34664670

ABSTRACT

The success of protein engineering and design has extensively expanded the protein space, which presents a promising strategy for creating next-generation proteins of diverse functions. Among these proteins, the synthetic binding proteins (SBPs) are smaller, more stable, less immunogenic, and better of tissue penetration than others, which make the SBP-related data attracting extensive interest from worldwide scientists. However, no database has been developed to systematically provide the valuable information of SBPs yet. In this study, a database named 'Synthetic Binding Proteins for Research, Diagnosis, and Therapy (SYNBIP)' was thus introduced. This database is unique in (a) comprehensively describing thousands of SBPs from the perspectives of scaffolds, biophysical & functional properties, etc.; (b) panoramically illustrating the binding targets & the broad application of each SBP and (c) enabling a similarity search against the sequences of all SBPs and their binding targets. Since SBP is a human-made protein that has not been found in nature, the discovery of novel SBPs relied heavily on experimental protein engineering and could be greatly facilitated by in-silico studies (such as AI and computational modeling). Thus, the data provided in SYNBIP could lay a solid foundation for the future development of novel SBPs. The SYNBIP is accessible without login requirement at both official (https://idrblab.org/synbip/) and mirror (http://synbip.idrblab.net/) sites.


Subject(s)
Bacterial Proteins/classification , Carrier Proteins/genetics , Databases, Protein , Proteins/classification , Bacterial Proteins/chemistry , Carrier Proteins/classification , Computer Simulation , Humans , Protein Conformation , Protein Engineering/trends , Proteins/chemistry
10.
Nucleic Acids Res ; 50(D1): D758-D764, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34614159

ABSTRACT

With their photosynthetic ability and established genetic modification systems, cyanobacteria are essential for fundamental and biotechnological research. Till now, hundreds of cyanobacterial genomes have been sequenced, and transcriptomic analysis has been frequently applied in the functional genomics of cyanobacteria. However, the massive omics data have not been extensively mined and integrated. Here, we describe CyanoOmicsDB (http://www.cyanoomics.cn/), a database aiming to provide comprehensive functional information for each cyanobacterial gene. CyanoOmicsDB consists of 8 335 261 entries of cyanobacterial genes from 928 genomes. It provides multiple gene identifiers, visualized genomic location, and DNA sequences for each gene entry. For protein-encoding genes, CyanoOmicsDB can provide predicted gene function, amino acid sequences, homologs, protein-domain super-families, and accession numbers for various public protein function databases. CyanoOmicsDB integrates both transcriptional and translational profiles of Synechocystis sp. PCC 6803 under various environmental culture coditions and genetic backgrounds. Moreover, CyanoOmicsDB includes 23 689 gene transcriptional start sites, 94 644 identified peptides, and 16 778 post-translation modification sites obtained from transcriptomes or proteomes of several model cyanobacteria. Compared with other existing cyanobacterial databases, CyanoOmicsDB comprises more datasets and more comprehensive functional information. CyanoOmicsDB will provide researchers in this field with a convenient way to retrieve functional information on cyanobacterial genes.


Subject(s)
Bacterial Proteins/genetics , Cyanobacteria/genetics , Databases, Genetic , Genome, Bacterial , Software , Synechocystis/genetics , Bacterial Proteins/classification , Bacterial Proteins/metabolism , Cyanobacteria/classification , Cyanobacteria/metabolism , Data Mining , Gene Expression Profiling , Genomics/methods , Internet , Photosynthesis/genetics , Proteomics/methods , Synechocystis/metabolism , Transcriptome
11.
Nucleic Acids Res ; 50(D1): D912-D917, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34850947

ABSTRACT

The virulence factor database (VFDB, http://www.mgc.ac.cn/VFs/) is dedicated to presenting a comprehensive knowledge base and a versatile analysis platform for bacterial virulence factors (VFs). Recent developments in sequencing technologies have led to increasing demands to analyze potential VFs within microbiome data that always consist of many different bacteria. Nevertheless, the current classification of VFs from various pathogens is based on different schemes, which create a chaotic situation and form a barrier for the easy application of the VFDB dataset for future panbacterial metagenomic analyses. Therefore, based on extensive literature mining, we recently proposed a general category of bacterial VFs in the database and reorganized the VFDB dataset accordingly. Thus, all known bacterial VFs from 32 genera of common bacterial pathogens collected in the VFDB are well grouped into 14 basal categories along with over 100 subcategories in a hierarchical architecture. The new coherent and well-defined VFDB dataset will be feasible and applicable for future panbacterial analysis in terms of virulence factors. In addition, we introduced a redesigned JavaScript-independent web interface for the VFDB website to make the database readily accessible to all users with various client settings worldwide.


Subject(s)
Bacteria/genetics , Bacteria/pathogenicity , Bacterial Proteins/genetics , Databases, Genetic , Genome, Bacterial , Software , Virulence Factors/genetics , Bacteria/classification , Bacteria/metabolism , Bacterial Proteins/classification , Bacterial Proteins/metabolism , Data Mining , Gene Expression Regulation, Bacterial , Gene Regulatory Networks , Humans , Internet , Knowledge Bases , Molecular Sequence Annotation , Phylogeny , Protein Interaction Mapping , Virulence Factors/classification , Virulence Factors/metabolism
12.
Nucleic Acids Res ; 50(D1): D875-D882, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34664671

ABSTRACT

Bacillus subtilis is a Gram-positive model bacterium with extensive documented annotation. However, with the rise of high-throughput techniques, the amount of complex data being generated every year has been increasing at a fast pace. Thus, having platforms ready to integrate and give a representation to these data becomes a priority. To address it, SubtiWiki (http://subtiwiki.uni-goettingen.de/) was created in 2008 and has been growing in data and viewership ever since. With millions of requests every year, it is the most visited B. subtilis database, providing scientists all over the world with curated information about its genes and proteins, as well as intricate protein-protein interactions, regulatory elements, expression data and metabolic pathways. However, there is still a large portion of annotation to be unveiled for some biological elements. Thus, to facilitate the development of new hypotheses for research, we have added a Homology section covering potential protein homologs in other organisms. Here, we present the recent developments of SubtiWiki and give a guided tour of our database and the current state of the data for this organism.


Subject(s)
Bacillus subtilis/genetics , Bacteria/genetics , Bacterial Proteins/genetics , Databases, Genetic , Genome, Bacterial , Software , Amino Acid Sequence , Bacillus subtilis/metabolism , Bacteria/classification , Bacteria/metabolism , Bacterial Proteins/classification , Bacterial Proteins/metabolism , Base Sequence , Gene Expression Regulation, Bacterial , Gene Regulatory Networks , Internet , Metabolic Networks and Pathways/genetics , Models, Molecular , Molecular Sequence Annotation , Phylogeny , Protein Conformation , Protein Interaction Mapping , Sequence Homology, Amino Acid
13.
J Biol Chem ; 298(10): 102392, 2022 10.
Article in English | MEDLINE | ID: mdl-35988643

ABSTRACT

Enzymes involved in Staphylococcus aureus amino acid metabolism have recently gained traction as promising targets for the development of new antibiotics, however, not all aspects of this process are understood. The ATP-grasp superfamily includes enzymes that predominantly catalyze the ATP-dependent ligation of various carboxylate and amine substrates. One subset, ʟ-amino acid ligases (LALs), primarily catalyze the formation of dipeptide products in Gram-positive bacteria, however, their involvement in S. aureus amino acid metabolism has not been investigated. Here, we present the characterization of the putative ATP-grasp enzyme (SAOUHSC_02373) from S. aureus NCTC 8325 and its identification as a novel LAL. First, we interrogated the activity of SAOUHSC_02373 against a panel of ʟ-amino acid substrates. As a result, we identified SAOUHSC_02373 as an LAL with high selectivity for ʟ-aspartate and ʟ-methionine substrates, specifically forming an ʟ-aspartyl-ʟ-methionine dipeptide. Thus, we propose that SAOUHSC_02373 be assigned as ʟ-aspartate-ʟ-methionine ligase (LdmS). To further understand this unique activity, we investigated the mechanism of LdmS by X-ray crystallography, molecular modeling, and site-directed mutagenesis. Our results suggest that LdmS shares a similar mechanism to other ATP-grasp enzymes but possesses a distinctive active site architecture that confers selectivity for the ʟ-Asp and ʟ-Met substrates. Phylogenetic analysis revealed LdmS homologs are highly conserved in Staphylococcus and closely related Gram-positive Firmicutes. Subsequent genetic analysis upstream of the ldmS operon revealed several trans-acting regulatory elements associated with control of Met and Cys metabolism. Together, these findings support a role for LdmS in Staphylococcal sulfur amino acid metabolism.


Subject(s)
Bacterial Proteins , Cysteine , Methionine , Peptide Synthases , Staphylococcus aureus , Adenosine Triphosphate/metabolism , Aspartic Acid/chemistry , Aspartic Acid/metabolism , Dipeptides/biosynthesis , Methionine/chemistry , Methionine/metabolism , Phylogeny , Staphylococcus aureus/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/classification , Bacterial Proteins/genetics , Peptide Synthases/chemistry , Peptide Synthases/classification , Peptide Synthases/genetics , Cysteine/chemistry , Cysteine/metabolism
14.
Mol Cell ; 59(2): 309-20, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26166706

ABSTRACT

Sirtuins are an ancient family of NAD(+)-dependent deacylases connected with the regulation of fundamental cellular processes including metabolic homeostasis and genome integrity. We show the existence of a hitherto unrecognized class of sirtuins, found predominantly in microbial pathogens. In contrast to earlier described classes, these sirtuins exhibit robust protein ADP-ribosylation activity. In our model organisms, Staphylococcus aureus and Streptococcus pyogenes, the activity is dependent on prior lipoylation of the target protein and can be reversed by a sirtuin-associated macrodomain protein. Together, our data describe a sirtuin-dependent reversible protein ADP-ribosylation system and establish a crosstalk between lipoylation and mono-ADP-ribosylation. We propose that these posttranslational modifications modulate microbial virulence by regulating the response to host-derived reactive oxygen species.


Subject(s)
Adenosine Diphosphate Ribose/metabolism , Bacterial Proteins/classification , Sirtuins/classification , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Catalytic Domain , Crystallography, X-Ray , Genes, Bacterial , HEK293 Cells , Host-Pathogen Interactions , Humans , Lactobacillales/enzymology , Lactobacillales/genetics , Lipoylation , Models, Molecular , Operon , Oxidative Stress , Phylogeny , Protein Conformation , Sirtuins/chemistry , Sirtuins/genetics , Staphylococcus aureus/enzymology , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Streptococcus pyogenes/enzymology , Streptococcus pyogenes/genetics , Streptococcus pyogenes/pathogenicity
15.
Cell Mol Life Sci ; 79(3): 179, 2022 Mar 06.
Article in English | MEDLINE | ID: mdl-35253091

ABSTRACT

ATP synthases are unique rotatory molecular machines that supply biochemical reactions with adenosine triphosphate (ATP)-the universal "currency", which cells use for synthesis of vital molecules and sustaining life. ATP synthases of F-type (FOF1) are found embedded in bacterial cellular membrane, in thylakoid membranes of chloroplasts, and in mitochondrial inner membranes in eukaryotes. The main functions of ATP synthases are control of the ATP synthesis and transmembrane potential. Although the key subunits of the enzyme remain highly conserved, subunit composition and structural organization of ATP synthases and their assemblies are significantly different. In addition, there are hypotheses that the enzyme might be involved in the formation of the mitochondrial permeability transition pore and play a role in regulation of the cell death processes. Dysfunctions of this enzyme lead to numerous severe disorders with high fatality levels. In our review, we focus on FOF1-structure-based approach towards development of new therapies by using FOF1 structural features inherited by the representatives of this enzyme family from different taxonomy groups. We analyzed and systematized the most relevant information about the structural organization of FOF1 to discuss how this approach might help in the development of new therapies targeting ATP synthases and design tools for cellular bioenergetics control.


Subject(s)
Drug Design , Proton-Translocating ATPases/metabolism , Adenosine Triphosphate/metabolism , Bacteria/metabolism , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/classification , Bacterial Proteins/metabolism , Chloroplasts/metabolism , Eukaryota/metabolism , Phylogeny , Protein Subunits/metabolism , Proton-Translocating ATPases/antagonists & inhibitors , Proton-Translocating ATPases/classification , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism
16.
Nucleic Acids Res ; 49(D1): D274-D281, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33167031

ABSTRACT

The Clusters of Orthologous Genes (COG) database, also referred to as the Clusters of Orthologous Groups of proteins, was created in 1997 and went through several rounds of updates, most recently, in 2014. The current update, available at https://www.ncbi.nlm.nih.gov/research/COG, substantially expands the scope of the database to include complete genomes of 1187 bacteria and 122 archaea, typically, with a single genome per genus. In addition, the current version of the COGs includes the following new features: (i) the recently deprecated NCBI's gene index (gi) numbers for the encoded proteins are replaced with stable RefSeq or GenBank\ENA\DDBJ coding sequence (CDS) accession numbers; (ii) COG annotations are updated for >200 newly characterized protein families with corresponding references and PDB links, where available; (iii) lists of COGs grouped by pathways and functional systems are added; (iv) 266 new COGs for proteins involved in CRISPR-Cas immunity, sporulation in Firmicutes and photosynthesis in cyanobacteria are included; and (v) the database is made available as a web page, in addition to FTP. The current release includes 4877 COGs. Future plans include further expansion of the COG collection by adding archaeal COGs (arCOGs), splitting the COGs containing multiple paralogs, and continued refinement of COG annotations.


Subject(s)
Archaea/genetics , Bacteria/genetics , Databases, Genetic , Genome, Archaeal , Genome, Bacterial , Archaea/metabolism , Archaeal Proteins/classification , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Bacteria/immunology , Bacteria/metabolism , Bacterial Proteins/classification , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , CRISPR-Cas Systems , Gene Ontology , Humans , Molecular Sequence Annotation , Spores, Bacterial/genetics , Spores, Bacterial/growth & development
17.
Crit Rev Biochem Mol Biol ; 55(6): 716-728, 2020 12.
Article in English | MEDLINE | ID: mdl-33003953

ABSTRACT

Although transcription by RNA polymerase (RNAP) is highly processive, elongation can be transiently halted by RNAP pausing. Pausing provides time for diverse regulatory events to occur such as RNA folding and regulatory factor binding. The transcription elongation factors NusA and NusG dramatically affect the frequency and duration of RNAP pausing, and hence regulation of transcription. NusG is the only transcription factor conserved in all three domains of life; its homolog in archaea and eukaryotes is Spt5. This review focuses on NusG-dependent pausing, which is a common occurrence in Bacillus subtilis. B. NusG induces pausing about once per 3 kb at a consensus TTNTTT motif in the non-template DNA strand within the paused transcription bubble. A conserved region of NusG contacts the TTNTTT motif to stabilize the paused transcription elongation complex (TEC) in multiple catalytically inactive RNAP conformations. The density of NusG-dependent pause sites is 3-fold higher in untranslated regions, suggesting that pausing could regulate the expression of hundreds of genes in B. subtilis. We describe how pausing in 5' leader regions contributes to regulating the expression of B. subtilis genes by transcription attenuation and translation control mechanisms. As opposed to the broadly accepted view that NusG is an anti-pausing factor, phylogenetic analyses suggest that NusG-dependent pausing is a widespread mechanism in bacteria. This function of NusG is consistent with the well-established role of its eukaryotic homolog Spt5 in promoter-proximal pausing. Since NusG is present in all domains of life, NusG-dependent pausing could be a conserved mechanism in all organisms.


Subject(s)
Bacillus subtilis/metabolism , DNA-Directed RNA Polymerases/metabolism , Transcription Factors/metabolism , Transcriptional Elongation Factors/metabolism , Bacillus subtilis/genetics , Bacterial Proteins/classification , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , DNA-Directed RNA Polymerases/genetics , Gene Expression Regulation, Bacterial , Phylogeny , Transcription Factors/genetics , Transcriptional Elongation Factors/genetics
18.
J Bacteriol ; 204(2): e0044921, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34898261

ABSTRACT

Two-component systems (TCS) are signaling pathways that allow bacterial cells to sense, respond to, and adapt to fluctuating environments. Among the classical TCS of Escherichia coli, HprSR has recently been shown to be involved in the regulation of msrPQ, which encodes the periplasmic methionine sulfoxide reductase system. In this study, we demonstrated that hypochlorous acid (HOCl) induces the expression of msrPQ in an HprSR-dependent manner, whereas H2O2, NO, and paraquat (a superoxide generator) do not. Therefore, HprS appears to be an HOCl-sensing histidine kinase. Using a directed mutagenesis approach, we showed that Met residues located in the periplasmic loop of HprS are important for its activity: we provide evidence that as HOCl preferentially oxidizes Met residues, HprS could be activated via the reversible oxidation of its methionine residues, meaning that MsrPQ plays a role in switching HprSR off. We propose that the activation of HprS by HOCl could occur through a Met redox switch. HprSR appears to be the first characterized TCS able to detect reactive chlorine species (RCS) in E. coli. This study represents an important step toward understanding the mechanisms of RCS resistance in prokaryotes. IMPORTANCE Understanding how bacteria respond to oxidative stress at the molecular level is crucial in the fight against pathogens. HOCl is one of the most potent industrial and physiological microbicidal oxidants. Therefore, bacteria have developed counterstrategies to survive HOCl-induced stress. Over the last decade, important insights into these bacterial protection factors have been obtained. Our work establishes HprSR as a reactive chlorine species-sensing, two-component system in Escherichia coli MG1655, which regulates the expression of msrPQ, two genes encoding, a repair system for HOCl-oxidized proteins. Moreover, we provide evidence suggesting that HOCl could activate HprS through a methionine redox switch.


Subject(s)
Chlorine/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Oxidative Stress/physiology , Bacterial Proteins/classification , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Escherichia coli/chemistry , Escherichia coli/drug effects , Hydrogen Peroxide/pharmacology , Hypochlorous Acid/pharmacology , Nitric Oxide/pharmacology , Oxidation-Reduction , Oxidative Stress/drug effects , Phosphoenolpyruvate Sugar Phosphotransferase System/classification , Phosphoenolpyruvate Sugar Phosphotransferase System/genetics , Phosphoenolpyruvate Sugar Phosphotransferase System/metabolism , Signal Transduction
19.
PLoS Comput Biol ; 17(1): e1008487, 2021 01.
Article in English | MEDLINE | ID: mdl-33406089

ABSTRACT

Investigating metabolic functional capability of a human gut microbiome enables the quantification of microbiome changes, which can cause a phenotypic change of host physiology and disease. One possible way to estimate the functional capability of a microbial community is through inferring metagenomic content from 16S rRNA gene sequences. Genome-scale models (GEMs) can be used as scaffold for functional estimation analysis at a systematic level, however up to date, there is no integrative toolbox based on GEMs for uncovering metabolic functions. Here, we developed the MetGEMs (metagenome-scale models) toolbox, an open-source application for inferring metabolic functions from 16S rRNA gene sequences to facilitate the study of the human gut microbiome by the wider scientific community. The developed toolbox was validated using shotgun metagenomic data and shown to be superior in predicting functional composition in human clinical samples compared to existing state-of-the-art tools. Therefore, the MetGEMs toolbox was subsequently applied for annotating putative enzyme functions and metabolic routes related in human disease using atopic dermatitis as a case study.


Subject(s)
Bacteria , Gastrointestinal Microbiome/genetics , Metagenome/genetics , Metagenomics/methods , Software , Bacteria/enzymology , Bacteria/genetics , Bacteria/metabolism , Bacterial Proteins/classification , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , DNA, Bacterial/genetics , Feces/microbiology , Humans , RNA, Ribosomal, 16S/genetics
20.
Biochem J ; 478(8): 1585-1603, 2021 04 30.
Article in English | MEDLINE | ID: mdl-33909028

ABSTRACT

Epithelial surfaces throughout the body are coated by mucins, a class of proteins carrying domains characterized by a high density of O-glycosylated serine and threonine residues. The resulting mucosal layers form crucial host-microbe interfaces that prevent the translocation of microbes while also selecting for distinct bacteria via the presented glycan repertoire. The intricate interplay between mucus production and breakdown thus determines the composition of the microbiota maintained within these mucosal environments, which can have a large influence on the host during both homeostasis and disease. Most research to date on mucus breakdown has focused on glycosidases that trim glycan structures to release monosaccharides as a source of nutrients. More recent work has uncovered the existence of mucin-type O-glycosylation-dependent proteases that are secreted by pathogens, commensals, and mutualists to facilitate mucosal colonization and penetration. Additionally, immunoglobulin A (IgA) proteases promote bacterial colonization in the presence of neutralizing secretory IgA through selective cleavage of the heavily O-glycosylated hinge region. In this review, we summarize families of O-glycoproteases and IgA proteases, discuss known structural features, and review applications of these enzymes to glycobiology.


Subject(s)
Bacterial Proteins/metabolism , Metalloendopeptidases/metabolism , Mucin-1/metabolism , Mucins/metabolism , Amino Acid Sequence , Bacteria/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/classification , Bacterial Proteins/genetics , Carbohydrate Sequence , Gene Expression , Host-Pathogen Interactions/genetics , Humans , Metalloendopeptidases/chemistry , Metalloendopeptidases/classification , Metalloendopeptidases/genetics , Mucin-1/chemistry , Mucin-1/genetics , Mucins/chemistry , Multigene Family , Protein Domains , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL