Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 411
Filter
1.
Environ Microbiol Rep ; 14(4): 570-576, 2022 08.
Article in English | MEDLINE | ID: mdl-35403341

ABSTRACT

Previously we described the discovery of a Bacillus spp. specific peptidase activity related to d-stereospecific peptidases (DSPs). The peptidase showed a strong preference for d-leucine and d-valine amino acids. These amino acids are present in the structure of the non-ribosomal peptide (NRP) antibiotics gramicidin A, B and C and polymyxin E. To examine if the Bacillus spp. DSP-related peptidase can hydrolyze these NRPs, the effect of gramicidin A and C and polymyxin E on peptidase activity in Bacillus anthracis culture supernatant was monitored. It was found that both gramicidins inhibited the DSP-related activity in a competitive manner. MALDI-TOF analysis revealed that upon incubation with B. anthracis culture supernatant gramicidin A hydrolyzation products appeared. This study shows that the Bacillus spp. specific DSP-like peptidase was potentially produced by the bacteria to gain intrinsic resistance against NRP antibiotics. These results are of utmost importance in research towards antimicrobial resistance, whereas transfer of DSP-related activity to other clinically relevant pathogens can be a serious threat to human health.


Subject(s)
Bacillus anthracis , Gramicidin , Peptide Hydrolases , Amino Acids/metabolism , Anti-Bacterial Agents/pharmacology , Bacillus anthracis/enzymology , Colistin/pharmacology , Gramicidin/metabolism , Peptide Hydrolases/metabolism
2.
J Bacteriol ; 203(24): e0041521, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34570623

ABSTRACT

Anthrax disease is caused by infection with the bacteria Bacillus anthracis which, if left untreated, can result in fatal bacteremia and toxemia. Current treatment for infection requires prolonged administration of antibiotics. Despite this, inhalational and gastrointestinal anthrax still result in lethal disease. By identifying key metabolic steps that B. anthracis uses to grow in host-like environments, new targets for antibacterial strategies can be identified. Here, we report that the ilvD gene, which encodes dihydroxyacid dehydratase in the putative pathway for synthesizing branched chain amino acids, is necessary for B. anthracis to synthesize isoleucine de novo in an otherwise limiting microenvironment. We observed that ΔilvD B. anthracis cannot grow in media lacking isoleucine, but growth is restored when exogenous isoleucine is added. In addition, ΔilvD bacilli are unable to utilize human hemoglobin or serum albumin to overcome isoleucine auxotrophy, but can when provided with the murine forms. This species-specific effect is due to the lack of isoleucine in human hemoglobin. Furthermore, even when supplemented with physiological levels of human serum albumin, apotransferrin, fibrinogen, and IgG, the ilvD knockout strain grew poorly relative to nonsupplemented wild type. In addition, comparisons upon infecting humanized mice suggest that murine hemoglobin is a key source of isoleucine for both WT and ΔilvD bacilli. Further growth comparisons in murine and human blood show that the auxotrophy is detrimental for growth in human blood, not murine. This report identifies ilvD as necessary for isoleucine production in B. anthracis, and that it plays a key role in allowing the bacilli to effectively grow in isoleucine poor hosts. IMPORTANCE Anthrax disease, caused by B. anthracis, can cause lethal bacteremia and toxemia, even following treatment with antibiotics. This report identifies the ilvD gene, which encodes a dihydroxyacid dehydratase, as necessary for B. anthracis to synthesize the amino acid isoleucine in a nutrient-limiting environment, such as its mammalian host. The use of this strain further demonstrated a unique species-dependent utilization of hemoglobin as an exogenous source of extracellular isoleucine. By identifying mechanisms that B. anthracis uses to grow in host-like environments, new targets for therapeutic intervention are revealed.


Subject(s)
Bacillus anthracis/enzymology , Gene Expression Regulation, Bacterial/physiology , Gene Expression Regulation, Enzymologic/physiology , Hydro-Lyases/metabolism , Animals , Bacillus anthracis/genetics , Bacillus anthracis/metabolism , Blood Proteins/chemistry , Blood Proteins/metabolism , Culture Media/chemistry , Gene Deletion , Hemoglobins/chemistry , Hemoglobins/metabolism , Humans , Hydro-Lyases/genetics , Mice , Mutation
3.
Int J Biol Macromol ; 177: 337-350, 2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33582216

ABSTRACT

This study evaluated the inhibitory potential of various beta-lactamase inhibitors such as mechanism-based inhibitors (MBIs), carbapenems, monobactam, and non-beta-lactam inhibitors against Bla1, a class-A beta-lactamase encoded by Bacillus anthracis. The binding potential of different inhibitors was estimated using competitive kinetic assay, isothermal titration calorimetry, and Biolayer interferometry. We observed that tazobactam has better inhibition among other MBIs with a characteristics inhibition dissociation constant of 0.51 ± 0.13 µM. Avibactam was also identified as good inhibitor with an inhibition efficiency of 0.6 ± 0.04 µM. All the MBIs (KD = 1.90E-04 M, 2.05E-05 M, 3.55E-04 M for clavulanate, sulbactam and tazobactam) showed significantly better binding potential than carbapenems (KD = 1.02E-03 M, 2.74E-03 M, 1.24E-03 M for ertapenem, imipenem and biapenem respectively). Molecular dynamics simulations were carried out using Bla1-inhibitor complexes to understand the dynamics and stability. The minimum inhibitory concentration (MIC) was carried out by taking various substrates and inhibitors, and later it was followed by cell viability assay. Together, our study helps develop a proper understanding of Bla1 beta-lactamase and its interaction with inhibitory molecules. This study would facilitate comprehending the catalytic divergence of beta-lactamases and the newly emergent resistant strains, focusing on the new generation of therapeutics being less prone to antimicrobial resistance.


Subject(s)
Azabicyclo Compounds/chemistry , Bacillus anthracis/enzymology , Bacterial Proteins , beta-Lactam Resistance , beta-Lactamase Inhibitors/chemistry , beta-Lactamases/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry
4.
Nat Microbiol ; 5(12): 1464-1471, 2020 12.
Article in English | MEDLINE | ID: mdl-32895527

ABSTRACT

Anthrax lethal toxin (LT), produced by Bacillus anthracis, comprises a receptor-binding moiety, protective antigen and the lethal factor (LF) protease1,2. Although LF is known to cleave mitogen-activated protein kinase kinases (MEKs/MKKs) and some variants of the NLRP1 inflammasome sensor, targeting of these pathways does not explain the lethality of anthrax toxin1,2. Here we report that the regulatory subunits of phosphoinositide-3 kinase (PI3K)-p85α (PIK3R1) and p85ß (PIK3R2)3,4-are substrates of LF. Cleavage of these proteins in a proline-rich region between their N-terminal Src homology and Bcr homology domains disrupts homodimer formation and impacts PI3K signalling. Mice carrying a mutated p85α that cannot be cleaved by LF show a greater resistance to anthrax toxin challenge. The LF(W271A) mutant cleaves p85α with lower efficiency and is non-toxic to mice but can regain lethality when combined with PI3K pathway inhibitors. We provide evidence that LF targets two signalling pathways that are essential for growth and metabolism and that the disabling of both pathways is likely necessary for lethal anthrax infection.


Subject(s)
Anthrax/enzymology , Antigens, Bacterial/metabolism , Antigens, Bacterial/toxicity , Bacillus anthracis/enzymology , Bacillus anthracis/metabolism , Bacterial Toxins/metabolism , Bacterial Toxins/toxicity , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Peptide Hydrolases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Amino Acid Motifs , Animals , Anthrax/genetics , Anthrax/microbiology , Class Ia Phosphatidylinositol 3-Kinase/chemistry , Class Ia Phosphatidylinositol 3-Kinase/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Peptide Hydrolases/genetics , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol 3-Kinases/genetics
5.
Proteins ; 88(11): 1394-1400, 2020 11.
Article in English | MEDLINE | ID: mdl-32501594

ABSTRACT

Sortases are a group of enzymes displayed on the cell-wall of Gram-positive bacteria. They are responsible for the attachment of virulence factors onto the peptidoglycan in a transpeptidation reaction through recognition of a pentapeptide substrate. Most housekeeping sortases recognize one specific pentapeptide motif; however, Streptococcus pyogenes sortase A (SpSrtA WT) recognizes LPETG, LPETA and LPKLG motifs. Here, we examined SpSrtA's flexible substrate specificity by investigating the role of the ß7/ß8 loop in determining substrate specificity. We exchanged the ß7/ß8 loop in SpSrtA with corresponding ß7/ß8 loops from Staphylococcus aureus (SaSrtA WT) and Bacillus anthracis (BaSrtA WT). While the BaSrtA-derived variant showed no enzymatic activity toward either LPETG or LPETA substrates, the activity of the SaSrtA-derived mutant toward the LPETA substrate was completely abolished. Instead, the mutant had an improved activity toward LPETG, the preferred substrate of SaSrtA WT.


Subject(s)
Aminoacyltransferases/chemistry , Bacillus anthracis/enzymology , Bacterial Proteins/chemistry , Cysteine Endopeptidases/chemistry , Oligopeptides/chemistry , Protein Engineering/methods , Staphylococcus aureus/enzymology , Streptococcus pyogenes/enzymology , Amino Acid Motifs , Aminoacyltransferases/genetics , Aminoacyltransferases/metabolism , Bacillus anthracis/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Cloning, Molecular , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Models, Molecular , Oligopeptides/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Staphylococcus aureus/chemistry , Streptococcus pyogenes/chemistry , Substrate Specificity
6.
J Biol Inorg Chem ; 25(4): 571-582, 2020 06.
Article in English | MEDLINE | ID: mdl-32296998

ABSTRACT

Correct protein metallation in the complex mixture of the cell is a prerequisite for metalloprotein function. While some metals, such as Cu, are commonly chaperoned, specificity towards metals earlier in the Irving-Williams series is achieved through other means, the determinants of which are poorly understood. The dimetal carboxylate family of proteins provides an intriguing example, as different proteins, while sharing a common fold and the same 4-carboxylate 2-histidine coordination sphere, are known to require either a Fe/Fe, Mn/Fe or Mn/Mn cofactor for function. We previously showed that the R2lox proteins from this family spontaneously assemble the heterodinuclear Mn/Fe cofactor. Here we show that the class Ib ribonucleotide reductase R2 protein from Bacillus anthracis spontaneously assembles a Mn/Mn cofactor in vitro, under both aerobic and anoxic conditions, when the metal-free protein is subjected to incubation with MnII and FeII in equal concentrations. This observation provides an example of a protein scaffold intrinsically predisposed to defy the Irving-Williams series and supports the assumption that the Mn/Mn cofactor is the biologically relevant cofactor in vivo. Substitution of a second coordination sphere residue changes the spontaneous metallation of the protein to predominantly form a heterodinuclear Mn/Fe cofactor under aerobic conditions and a Mn/Mn metal center under anoxic conditions. Together, the results describe the intrinsic metal specificity of class Ib RNR and provide insight into control mechanisms for protein metallation.


Subject(s)
Bacillus anthracis/enzymology , Bacterial Proteins/metabolism , Iron/metabolism , Manganese/metabolism , Ribonucleotide Reductases/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Crystallography, X-Ray , Iron/chemistry , Manganese/chemistry , Models, Molecular , Protein Conformation , Ribonucleotide Reductases/chemistry , Ribonucleotide Reductases/genetics
7.
J Infect Dis ; 221(4): 660-667, 2020 02 03.
Article in English | MEDLINE | ID: mdl-31574153

ABSTRACT

BACKGROUND: Lethal and edema toxins are critical virulence factors of Bacillus anthracis. Few data are available on their presence in the early stage of intranasal infection. METHODS: To investigate the diffusion of edema factor (EF) and lethal factor (LF), we use sensitive quantitative methods to measure their enzymatic activities in mice intranasally challenged with a wild-type B anthracis strain or with an isogenic mutant deficient for the protective antigen. RESULTS: One hour after mouse challenge, although only 7% of mice presented bacteremia, LF and EF were detected in the blood of 100% and 42% of mice, respectively. Protective antigen facilitated the diffusion of LF and EF into the blood compartment. Toxins played a significant role in the systemic dissemination of B anthracis in the blood, spleen, and liver. A mouse model of intoxination further confirmed that LT and ET could diffuse rapidly in the circulation, independently of bacteria. CONCLUSIONS: In this inhalational model, toxins have disseminated rapidly in the blood, playing a significant and novel role in the early systemic diffusion of bacteria, demonstrating that they may represent a very early target for the diagnosis and the treatment of anthrax.


Subject(s)
Anthrax/metabolism , Antigens, Bacterial/blood , Bacillus anthracis/pathogenicity , Bacterial Toxins/blood , Nasal Absorption , Virulence Factors/blood , Animals , Animals, Outbred Strains , Anthrax/microbiology , Bacillus anthracis/enzymology , Bacteremia , Biomarkers/blood , Disease Models, Animal , Enzyme Activation , Enzyme Assays , Female , Mice , Virulence
8.
FEBS J ; 287(11): 2235-2255, 2020 06.
Article in English | MEDLINE | ID: mdl-31750992

ABSTRACT

Tyrosine biosynthesis via the shikimate pathway is absent in humans and other animals, making it an attractive target for next-generation antibiotics, which is increasingly important due to the looming proliferation of multidrug-resistant pathogens. Tyrosine biosynthesis is also of commercial importance for the environmentally friendly production of numerous compounds, such as pharmaceuticals, opioids, aromatic polymers, and petrochemical aromatics. Prephenate dehydrogenase (PDH) catalyzes the penultimate step of tyrosine biosynthesis in bacteria: the oxidative decarboxylation of prephenate to 4-hydroxyphenylpyruvate. The majority of PDHs are competitively inhibited by tyrosine and consist of a nucleotide-binding domain and a dimerization domain. Certain PDHs, including several from pathogens on the World Health Organization priority list of antibiotic-resistant bacteria, possess an additional ACT domain. However, biochemical and structural knowledge was lacking for these enzymes. In this study, we successfully established a recombinant protein expression system for PDH from Bacillus anthracis (BaPDH), the causative agent of anthrax, and determined the structure of a BaPDH ternary complex with NAD+ and tyrosine, a binary complex with tyrosine, and a structure of an isolated ACT domain dimer. We also conducted detailed kinetic and biophysical analyses of the enzyme. We show that BaPDH is allosterically regulated by tyrosine binding to the ACT domains, resulting in an asymmetric conformation of the BaDPH dimer that sterically prevents prephenate binding to either active site. The presented mode of allosteric inhibition is unique compared to both the competitive inhibition established for other PDHs and to the allosteric mechanisms for other ACT-containing enzymes. This study provides new structural and mechanistic insights that advance our understanding of tyrosine biosynthesis in bacteria. ENZYMES: Prephenate dehydrogenase from Bacillus anthracis (PDH): EC database ID: 1.3.1.12. DATABASES: Coordinates and structure factors have been deposited in the Protein Data Bank (PDB) with accession numbers PDB ID: 6U60 (BaPDH complex with NAD+ and tyrosine), PDB ID: 5UYY (BaPDH complex with tyrosine), and PDB ID: 5V0S (BaPDH isolated ACT domain dimer). The diffraction images are available at http://proteindiffraction.org with DOIs: https://doi.org/10.18430/M35USC, https://doi.org/10.18430/M35UYY, and https://doi.org/10.18430/M35V0S.


Subject(s)
Bacillus anthracis/enzymology , Prephenate Dehydrogenase/genetics , Tyrosine/pharmacology , Bacillus anthracis/chemistry , Bacillus anthracis/ultrastructure , Catalysis/drug effects , Catalytic Domain/drug effects , Crystallography, X-Ray , Cyclohexanecarboxylic Acids/chemistry , Cyclohexenes/chemistry , Humans , Prephenate Dehydrogenase/ultrastructure , Protein Domains/drug effects , Tyrosine/chemistry
9.
Int J Biol Macromol ; 145: 510-526, 2020 Feb 15.
Article in English | MEDLINE | ID: mdl-31874266

ABSTRACT

Bacillus anthracis, a potent pathogen of anthrax is becoming resistant to many beta-lactam antibiotics because of the expression of two chromosomally encoded beta-lactamases Bla1 and Bla2. Bla1 is a class A beta-lactamase whereas Bla2 is a Metallo beta-lactamase. In the current study, we have attempted in-detailed characterization of Bla1 beta-lactamase by taking interdisciplinary approaches. Our study includes structure and sequence comparison of this enzyme with other members of the class, to know the conservation pattern that includes active site residues, secondary structure, conserved fold, evolutionary relationships, etc. Dynamic characterizations of the enzyme, unfolding kinetics were determined with the help of Molecular dynamics simulation. Detailed enzyme stability and catalytic activity towards various physical (Temperature and pH), and chemical parameters (Urea, GnHCl) were performed. Together, our study helps to develop a proper understanding of this beta-lactamase by characterizing its biochemical, biophysical, dynamic, kinetic and thermodynamic properties. This would help contribute towards a better understanding of beta-lactamase based AMR emergence.


Subject(s)
Bacillus anthracis/enzymology , Kinetics , beta-Lactamases/genetics , Anti-Bacterial Agents/chemistry , Bacillus anthracis/pathogenicity , Humans , Microbial Sensitivity Tests , Molecular Dynamics Simulation , Thermodynamics , beta-Lactamases/chemistry
10.
Biochemistry ; 58(44): 4447-4455, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31617352

ABSTRACT

Gyrase and topoisomerase IV are the targets of fluoroquinolone antibacterials. However, the rise in antimicrobial resistance has undermined the clinical use of this important drug class. Therefore, it is critical to identify new agents that maintain activity against fluoroquinolone-resistant strains. One approach is to develop non-fluoroquinolone drugs that also target gyrase and topoisomerase IV but interact differently with the enzymes. This has led to the development of the "novel bacterial topoisomerase inhibitor" (NBTI) class of antibacterials. Despite the clinical potential of NBTIs, there is a relative paucity of data describing their mechanism of action against bacterial type II topoisomerases. Consequently, we characterized the activity of GSK126, a naphthyridone/aminopiperidine-based NBTI, against a variety of Gram-positive and Gram-negative bacterial type II topoisomerases, including gyrase from Mycobacterium tuberculosis and gyrase and topoisomerase IV from Bacillus anthracis and Escherichia coli. GSK126 enhanced single-stranded DNA cleavage and suppressed double-stranded cleavage mediated by these enzymes. It was also a potent inhibitor of gyrase-catalyzed DNA supercoiling and topoisomerase IV-catalyzed decatenation. Thus, GSK126 displays a similar bimodal mechanism of action across a variety of species. In contrast, GSK126 displayed a variable ability to overcome fluoroquinolone resistance mutations across these same species. Our results suggest that NBTIs elicit their antibacterial effects by two different mechanisms: inhibition of gyrase/topoisomerase IV catalytic activity or enhancement of enzyme-mediated DNA cleavage. Furthermore, the relative importance of these two mechanisms appears to differ from species to species. Therefore, we propose that the mechanistic basis for the antibacterial properties of NBTIs is bimodal in nature.


Subject(s)
Anti-Bacterial Agents/chemistry , DNA Cleavage/drug effects , Indoles/chemistry , Naphthyridines/chemistry , Piperidines/chemistry , Pyridones/chemistry , Topoisomerase II Inhibitors/chemistry , Bacillus anthracis/enzymology , DNA Breaks, Double-Stranded/drug effects , DNA Gyrase/chemistry , DNA Topoisomerase IV/antagonists & inhibitors , DNA, Bacterial/drug effects , DNA, Single-Stranded/drug effects , Escherichia coli/enzymology , Mycobacterium tuberculosis/enzymology
11.
J Biol Inorg Chem ; 24(6): 849-861, 2019 09.
Article in English | MEDLINE | ID: mdl-31410573

ABSTRACT

Class Ib ribonucleotide reductases (RNR) utilize a di-nuclear manganese or iron cofactor for reduction of superoxide or molecular oxygen, respectively. This generates a stable tyrosyl radical (Y·) in the R2 subunit (NrdF), which is further used for ribonucleotide reduction in the R1 subunit of RNR. Here, we report high-resolution crystal structures of Bacillus anthracis NrdF in the metal-free form (1.51 Å) and in complex with manganese (MnII/MnII, 1.30 Å). We also report three structures of the protein in complex with iron, either prepared anaerobically (FeII/FeII form, 1.32 Å), or prepared aerobically in the photo-reduced FeII/FeII form (1.63 Å) and with the partially oxidized metallo-cofactor (1.46 Å). The structures reveal significant conformational dynamics, likely to be associated with the generation, stabilization, and transfer of the radical to the R1 subunit. Based on observed redox-dependent structural changes, we propose that the passage for the superoxide, linking the FMN cofactor of NrdI and the metal site in NrdF, is closed upon metal oxidation, blocking access to the metal and radical sites. In addition, we describe the structural mechanics likely to be involved in this process.


Subject(s)
Bacillus anthracis/enzymology , Bacillus anthracis/metabolism , Iron/metabolism , Manganese/metabolism , Metalloproteases/metabolism , Crystallography, X-Ray , FMN Reductase/chemistry , FMN Reductase/genetics , FMN Reductase/metabolism , Ferritins/chemistry , Ferritins/metabolism , Flavin Mononucleotide/chemistry , Flavin Mononucleotide/genetics , Flavin Mononucleotide/metabolism , Metalloproteases/chemistry , Metalloproteases/genetics , Ribonucleotide Reductases
12.
Mol Microbiol ; 112(2): 515-531, 2019 08.
Article in English | MEDLINE | ID: mdl-31063630

ABSTRACT

A challenge common to all bacterial pathogens is to acquire nutrients from hostile host environments. Iron is an important cofactor required for essential cellular processes such as DNA repair, energy production and redox balance. Within a mammalian host, most iron is sequestered within heme, which in turn is predominantly bound by hemoglobin. While little is understood about the mechanisms by which bacterial hemophores attain heme from host-hemoglobin, even less is known about intracellular heme processing. Bacillus anthracis, the causative agent of anthrax, displays a remarkable ability to grow in mammalian hosts. Hypothesizing this pathogen harbors robust ways to catabolize heme, we characterize two new intracellular heme-binding proteins that are distinct from the previously described IsdG heme monooxygenase. The first of these, HmoA, binds and degrades heme, is necessary for heme detoxification and facilitates growth on heme iron sources. The second protein, HmoB, binds and degrades heme too, but is not necessary for heme utilization or virulence. The loss of both HmoA and IsdG renders B. anthracis incapable of causing anthrax disease. The additional loss of HmoB in this background increases clearance of bacilli in lungs, which is consistent with this protein being important for survival in alveolar macrophages.


Subject(s)
Anthrax/microbiology , Bacillus anthracis/metabolism , Heme/metabolism , Anthrax/metabolism , Bacillus anthracis/enzymology , Bacillus anthracis/genetics , Bacillus anthracis/growth & development , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Humans , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Protein Binding
13.
J Biol Chem ; 294(22): 8930-8941, 2019 05 31.
Article in English | MEDLINE | ID: mdl-30952697

ABSTRACT

Bacillus anthracis is the causative agent of anthrax in humans, bovine, and other animals. B. anthracis pathogenesis requires differentiation of dormant spores into vegetative cells. The spores inherit cellular components as phenotypic memory from the parent cell, and this memory plays a critical role in facilitating the spores' revival. Because metabolism initiates at the beginning of spore germination, here we metabolically reprogrammed B. anthracis cells to understand the role of glycolytic enzymes in this process. We show that increased expression of enolase (Eno) in the sporulating mother cell decreases germination efficiency. Eno is phosphorylated by the conserved Ser/Thr protein kinase PrkC which decreases the catalytic activity of Eno. We found that phosphorylation also regulates Eno expression and localization, thereby controlling the overall spore germination process. Using MS analysis, we identified the sites of phosphorylation in Eno, and substitution(s) of selected phosphorylation sites helped establish the functional correlation between phosphorylation and Eno activity. We propose that PrkC-mediated regulation of Eno may help sporulating B. anthracis cells in adapting to nutrient deprivation. In summary, to the best of our knowledge, our study provides the first evidence that in sporulating B. anthracis, PrkC imprints phenotypic memory that facilitates the germination process.


Subject(s)
Bacillus anthracis/physiology , Bacterial Proteins/metabolism , Phosphopyruvate Hydratase/metabolism , Protein Serine-Threonine Kinases/metabolism , Spores, Bacterial/metabolism , Bacillus anthracis/enzymology , Bacterial Proteins/genetics , Kinetics , Magnesium/metabolism , Mutagenesis, Site-Directed , Phosphopyruvate Hydratase/genetics , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
14.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 4): 312-320, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30950833

ABSTRACT

Ba0331 is a putative polysaccharide deacetylase from Bacillus anthracis, the etiological agent of the disease anthrax, that contributes to adaptation of the bacterium under extreme conditions and to maintenance of the cell shape. In the present study, the crystal structure of Ba0331 was determined at 2.6 Šresolution. The structure consists of two domains: a fibronectin type 3-like (Fn3-like) domain and a NodB catalytic domain. The latter is present in all carbohydrate esterase family 4 enzymes, while a comparative analysis of the Fn3-like domain revealed structural plasticity despite the retention of the conserved Fn3-like domain characteristics.


Subject(s)
Amidohydrolases/chemistry , Amidohydrolases/genetics , Bacillus anthracis/enzymology , Gene Expression , Amino Acid Sequence , Binding Sites , Cloning, Molecular , Crystallization , Crystallography, X-Ray , Models, Molecular , Protein Domains , Static Electricity , Zinc/metabolism
15.
Science ; 364(6435)2019 04 05.
Article in English | MEDLINE | ID: mdl-30872533

ABSTRACT

Inflammasomes are multiprotein platforms that initiate innate immunity by recruitment and activation of caspase-1. The NLRP1B inflammasome is activated upon direct cleavage by the anthrax lethal toxin protease. However, the mechanism by which cleavage results in NLRP1B activation is unknown. In this study, we find that cleavage results in proteasome-mediated degradation of the amino-terminal domains of NLRP1B, liberating a carboxyl-terminal fragment that is a potent caspase-1 activator. Proteasome-mediated degradation of NLRP1B is both necessary and sufficient for NLRP1B activation. Consistent with our functional degradation model, we identify IpaH7.8, a Shigella flexneri ubiquitin ligase secreted effector, as an enzyme that induces NLRP1B degradation and activation. Our results provide a unified mechanism for NLRP1B activation by diverse pathogen-encoded enzymatic activities.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antigens, Bacterial/metabolism , Apoptosis Regulatory Proteins/metabolism , Bacterial Proteins/metabolism , Host-Pathogen Interactions/immunology , Immunity, Innate , Inflammasomes/immunology , Peptide Hydrolases/metabolism , Proteolysis , Shigella flexneri/pathogenicity , Ubiquitin-Protein Ligases/metabolism , Animals , Bacillus anthracis/enzymology , Bacterial Toxins/metabolism , CARD Signaling Adaptor Proteins/chemistry , CARD Signaling Adaptor Proteins/metabolism , Caspase 1/metabolism , Death Domain Receptor Signaling Adaptor Proteins/chemistry , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Enzyme Activation , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , NLR Proteins , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Domains , Protein Subunits , RAW 264.7 Cells , Shigella flexneri/enzymology
16.
J Appl Microbiol ; 126(6): 1700-1707, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30776160

ABSTRACT

AIMS: To develop a gel formulation to trigger a visual signal for rapid disclosure of the location and extent of surface contamination with viable Bacillus anthracis spores. METHODS AND RESULTS: Methylumbelliferyl-α-d-glucopyranoside was combined with hyaluronic acid to produce a gel that could be applied to a surface as a coating. It remained hydrated for a sufficient time for α-glucosidase activity present in intact B. anthracis spores to cleave the substrate and release the fluorescent product, methylumbelliferone. The presence of B. anthracis spores could be disclosed at 5 × 104 CFU per reaction test well (0·32 cm2 ) both visually and using fluorescence detection equipment. CONCLUSIONS: The disclosure gel provides a rapid, visual response to the presence of B. anthracis spores on a surface. SIGNIFICANCE AND IMPACT OF THE STUDY: The disclosure gel demonstrates the first steps towards the development of a formulation that can provide nonspecialist users with a visual alert to the presence of B. anthracis spores on a surface. It is envisioned that such a formulation would be beneficial in scenarios where exposure to spore release is a risk, and could be used in the initial assessment of equipment to aid prioritization and localized execution of a decontamination strategy.


Subject(s)
Bacillus anthracis/isolation & purification , Decontamination/methods , Environmental Exposure/prevention & control , Microbiological Techniques/methods , Spores, Bacterial/isolation & purification , Bacillus anthracis/enzymology , Bacillus anthracis/metabolism , Hyaluronic Acid/chemistry , Hymecromone/chemistry , Hymecromone/metabolism , Indicators and Reagents , Spores, Bacterial/enzymology , Spores, Bacterial/metabolism , alpha-Glucosidases/metabolism
17.
Int J Biol Macromol ; 127: 585-593, 2019 Apr 15.
Article in English | MEDLINE | ID: mdl-30660563

ABSTRACT

A homology model of ferrochelatase (HemH), the heme biosynthesis terminal step enzyme from Salmonella Typhi was generated to understand the mechanism of metal insertion into protoporphyrin IX for heme biosynthesis. The overall fold of membrane associated ferrochelatase (StFc) from S. Typhi is similar to human and Yeast ferrochelatase than Bacillus subtilis, and Bacillus anthracis. An insertion of 16 amino acid residues in helical switch having hydrophobic patch proposed to interact with membrane lipids and in opening and closing of heme binding cleft. The sequence analysis and the docking study revealed that the protoporphyrin binding site in StFc has a crucial replacement of Tyr/Met to Leu13 unique in comparison to other known structures, where Tyr13 observed in B. subtilis/B. anthracis while Met76 in human/yeast play important role in holding protoporphyrin in optimal orientation for metalation. A sitting-a-top (SAT) complex mechanism for metalation is proposed with His194 and Glu264 lie at the bottom and Leu13 on the top of the porphyrin ring. In addition, an entry and exit mechanism is also proposed for protoporphyrin binding into cavity by opening and closing of helical switch using molecular dynamics simulation studies of Apo and heme complexed model structure of S. Typhi HemH.


Subject(s)
Bacterial Proteins/chemistry , Ferrochelatase/chemistry , Molecular Dynamics Simulation , Salmonella typhi/enzymology , Bacillus anthracis/enzymology , Bacillus subtilis/enzymology , Humans , Protoporphyrins/chemistry
18.
Appl Environ Microbiol ; 85(6)2019 03 15.
Article in English | MEDLINE | ID: mdl-30658980

ABSTRACT

Acylation of epsilon amino groups of lysyl side chains is a widespread modification of proteins and small molecules in cells of all three domains of life. Recently, we showed that Bacillus subtilis and Bacillus anthracis encode the GCN5-related N-acetyltransferase (GNAT) SatA that can acetylate and inactivate streptothricin, which is a broad-spectrum antibiotic produced by actinomycetes in the soil. To determine functionally relevant residues of B. subtilis SatA (BsSatA), a mutational screen was performed, highlighting the importance of a conserved area near the C terminus. Upon inspection of the crystal structure of the B. anthracis Ames SatA (BaSatA; PDB entry 3PP9), this area appears to form a pocket with multiple conserved aromatic residues; we hypothesized this region contains the streptothricin-binding site. Chemical and site-directed mutagenesis was used to introduce missense mutations into satA, and the functionality of the variants was assessed using a heterologous host (Salmonella enterica). Results of isothermal titration calorimetry experiments showed that residue Y164 of BaSatA was important for binding streptothricin. Results of size exclusion chromatography analyses showed that residue D160 was important for dimerization. Together, these data advance our understanding of how SatA interacts with streptothricin.IMPORTANCE This work provides insights into how an abundant antibiotic found in soil is bound to the enzyme that inactivates it. This work identifies residues for the binding of the antibiotic and probes the contributions of substituting side chains for those in the native protein, providing information regarding hydrophobicity, size, and flexibility of the antibiotic binding site.


Subject(s)
Acetyltransferases/metabolism , Anti-Bacterial Agents/metabolism , Bacillus anthracis/enzymology , Bacillus subtilis/enzymology , Bacterial Proteins/metabolism , Streptothricins/metabolism , Acetylation , Acetyltransferases/chemistry , Acetyltransferases/genetics , Anti-Bacterial Agents/chemistry , Bacillus anthracis/genetics , Bacillus anthracis/metabolism , Bacillus subtilis/chemistry , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Streptothricins/chemistry
19.
ChemMedChem ; 14(1): 78-82, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30376607

ABSTRACT

Antibiotics with novel mechanisms of action are desperately needed to combat the increasing rates of multidrug-resistant infections. Bacterial pantothenate kinase (PanK) has emerged as a target of interest to cut off the biosynthesis of coenzyme A. Herein we report the results of an in vitro high-throughput screen of over 10 000 small molecules against Bacillus anthracis PanK, as well as a follow-up screen of hits against PanK isolated from Pseudomonas aeruginosa and Burkholderia cenocepacia. Nine hits are structurally categorized and analyzed to set the stage for future drug development.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacillus anthracis/drug effects , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Bacillus anthracis/enzymology , Dose-Response Relationship, Drug , Microbial Sensitivity Tests , Molecular Structure , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
20.
Adv Exp Med Biol ; 1111: 189-203, 2019.
Article in English | MEDLINE | ID: mdl-30267305

ABSTRACT

The zinc-dependent metalloprotease anthrax lethal factor (LF) is the enzymatic component of a toxin thought to have a major role in Bacillus anthracis infections. Like many bacterial toxins, LF is a secreted protein that functions within host cells. LF is a highly selective protease that cleaves a limited number of substrates in a site-specific manner, thereby impacting host signal transduction pathways. The major substrates of LF are mitogen-activated protein kinase kinases (MKKs), which lie in the middle of three-component phosphorylation cascades mediating numerous functions in a variety of cells and tissues. How LF targets its limited substrate repertoire has been an active area of investigation. LF recognizes a specific sequence motif surrounding the scissile bonds of substrate proteins. X-ray crystallography of the protease in complex with peptide substrates has revealed the structural basis of selectivity for the LF cleavage site motif. In addition to having interactions proximal to the cleavage site, LF binds directly to a more distal region in its substrates through a so-called exosite interaction. This exosite has been mapped to a surface within a non-catalytic domain of LF with previously unknown function. A putative LF-binding site has likewise been identified on the catalytic domains of MKKs. Here we review our current state of understanding of LF-substrate interactions and discuss the implications for the design and discovery of inhibitors that may have utility as anthrax therapeutics.


Subject(s)
Antigens, Bacterial/metabolism , Bacillus anthracis/enzymology , Bacterial Toxins/metabolism , Metalloproteases/metabolism , Humans , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...