Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Nucleic Acids Res ; 51(13): 6554-6565, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37283083

ABSTRACT

UdgX excises uracil from uracil-containing DNA to concurrently form a covalent bond with the resulting AP-DNA. Structurally, UdgX is highly similar to family-4 UDGs (F4-UDGs). However, UdgX is unique in possessing a flexible R-loop (105KRRIH109). Among the class-defining motifs, while its motif A (51GEQPG55) diverged to possess Q53 in place of A53/G53 in F4-UDGs, motif B [178HPS(S/A)(L/V)(L/V)R184] has remained unchanged. Previously, we proposed an SN1 mechanism resulting in a covalent bond between H109 and AP-DNA. In this study, we investigated several single/double mutants of UdgX. The H109A, H109S, H109G, H109Q, H109C and H109K mutants gain conventional UDG activity to varying levels. The crystal structures of UdgX mutants show topological changes in their active sites, rationalizing their UDG activities. The E52Q, E52N and E52A mutants reveal that E52 forms a catalytic dyad with H109 to enhance its nucleophilicity. The Q53A mutant supports that UdgX specific evolution of Q53 occurred essentially to stabilize the R-loop conformation. The R184A mutation (motif B) supports the role of R184 in substrate-binding. Taken together, the structural, bioinformatics, and mutational studies suggest that UdgX diverged from F4-UDGs, and the emergence of the characteristic R-loop in UdgX is functionally assisted by A53/G53 to Q53 changes in motif A.


Subject(s)
Uracil-DNA Glycosidase , Catalytic Domain , DNA/chemistry , DNA Repair , Mutation , Uracil , Uracil-DNA Glycosidase/chemistry , Uracil-DNA Glycosidase/genetics
2.
J Biol Chem ; 299(3): 102933, 2023 03.
Article in English | MEDLINE | ID: mdl-36690275

ABSTRACT

SigA (σA) is an essential protein and the primary sigma factor in Mycobacterium tuberculosis (Mtb). However, due to the absence of genetic tools, our understanding of the role and regulation of σA activity and its molecular attributes that help modulate Mtb survival is scant. Here, we generated a conditional gene replacement of σA in Mtb and showed that its depletion results in a severe survival defect in vitro, ex vivo, and in vivo in a murine infection model. Our RNA-seq analysis suggests that σA either directly or indirectly regulates ∼57% of the Mtb transcriptome, including ∼28% of essential genes. Surprisingly, we note that despite having ∼64% similarity with σA, overexpression of the primary-like σ factor SigB (σB) fails to compensate for the absence of σA, suggesting minimal functional redundancy. RNA-seq analysis of the Mtb σB deletion mutant revealed that 433 genes are regulated by σB, of which 283 overlap with the σA transcriptome. Additionally, surface plasmon resonance, in vitro transcription, and functional complementation experiments reveal that σA residues between 132-179 that are disordered and missing from all experimentally determined σA-RNAP structural models are imperative for σA function. Moreover, phosphorylation of σA in the intrinsically disordered N-terminal region plays a regulatory role in modulating its activity. Collectively, these observations and analysis provide a rationale for the centrality of σA for the survival and pathogenicity of this bacillus.


Subject(s)
Bacterial Proteins , Microbial Viability , Mycobacterium tuberculosis , Sigma Factor , Sigma Factor/genetics , Sigma Factor/metabolism , Animals , Mice , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Transcriptome , Tuberculosis/microbiology , Sequence Deletion , Microbial Viability/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial/genetics
3.
Appl Environ Microbiol ; 90(5): e0002124, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38606981

ABSTRACT

Extracytoplasmic function (ECF) σ factors selectively upregulate expression of specific genes in bacteria. These σ factors, belonging to the σ70 family, are much smaller than the primary, housekeeping σ factor with two helical domains that interact with the Pribnow box and the -35 element of the promoter DNA. Structural studies reveal that promoter specificity in a σ factor is determined by the interactions between a loop (L3) and the Pribnow box element. Similarly, the efficiency of transcription initiation is governed by the polypeptide linker between the two promoter-binding domains. Both these polypeptide segments are dynamic and poorly conserved among ECF σ factor homologs. This feature hitherto limited insights from protein-DNA interactions to be correlated with transcription initiation efficiency. Here, we describe an approach to characterize these features that govern the dynamic range of gene expression using chimeric Escherichia coli σE. The L3 loop and linker polypeptides in these σE chimeras were replaced by the corresponding segments from 10 annotated and functional Mycobacterium tuberculosis ECF σ's. In vitro and in vivo measurements to determine the effect of these polypeptide replacements provided an experimentally validated σE chimera- gene expression level data set. We illustrate the utility of this chimeric σE library in improving the efficiency of a biosynthetic pathway in E. coli. In a two-enzyme step, unaffected by feedback inhibition and substrate concentration, we show an increase in desired product levels by altering the relative intracellular levels of the target enzymes using this library of σ factors. The chimeric σE library thus demonstrates the feasibility of engineering σ factors to achieve bespoke expression levels of target genes for diverse applications in synthetic microbiology. IMPORTANCE: The synthesis of organic compounds involves the action of multiple enzymes in a biosynthetic pathway. Incorporating such biosynthetic pathways into microbes often leads to substantial cellular and metabolic stress resulting in low titers of the target compound. This limitation can be offset, in part, by optimizing enzyme efficiency and cellular enzyme concentration. The former involves significant efforts to achieve improvements in catalytic efficiency with the caveat that the metabolic load on a microbial cell imposed by the overexpression of the exogenous enzyme could result in reduced cell fitness. Here, we demonstrate the feasibility of engineered σ factors to modulate gene expression levels without significant genetic engineering. We note that changing the sequence of two flexible polypeptide loops without any changes to the structural scaffold of the transcription initiation factor σE could modulate the expression levels of the target genes. This ability provides a route to improve the efficiency of a biosynthetic pathway without altering the overall genomic makeup. The σE chimera library thus provides an avenue for pre-determined conditional gene expression of specific genes in Escherichia coli.


Subject(s)
Bacterial Proteins , Escherichia coli , Gene Expression Regulation, Bacterial , Sigma Factor , Sigma Factor/genetics , Sigma Factor/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Promoter Regions, Genetic , Protein Engineering/methods
4.
Bioorg Med Chem Lett ; 76: 129008, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36174837

ABSTRACT

Mycobacterium tuberculosis uracil-DNA glycosylase (MtUng), a key DNA repair enzyme, represents an attractive target for the design of new antimycobacterial agents. However, only a limited number of weak MtUng inhibitors are reported, primarily based on the uracil ring, and hence, lack diversity. We report the first structure-based virtual screening (SBVS) using three separate libraries consisting of uracil and non-uracil small molecules, together with the FDA-approved drugs. Twenty diverse virtual hits with the highest predicted binding were procured and screened using a fluorescence-based assay to evaluate their potential to inhibit MtUng. Several of these molecules were found to inhibit MtUng activity at low mM and µM levels, comparable to or better than several other reported Ung inhibitors. Thus, these molecules represent a diverse set of scaffolds for developing next-generation MtUng inhibitors. The most active uracil-based compound 5 (IC50 = 0.14 mM) was found to be âˆ¼ 15-fold more potent than the positive control, uracil. The binding stability and conformation of compound 5 in complex with the enzyme were further confirmed using molecular dynamics simulation.


Subject(s)
Mycobacterium tuberculosis , Uracil-DNA Glycosidase , Uracil-DNA Glycosidase/chemistry , Uracil-DNA Glycosidase/genetics , Uracil-DNA Glycosidase/metabolism , Molecular Dynamics Simulation , Uracil/pharmacology , Uracil/metabolism , Anti-Bacterial Agents
5.
Mol Divers ; 2022 Dec 27.
Article in English | MEDLINE | ID: mdl-36574164

ABSTRACT

Virtual screening (VS) is an important approach in drug discovery and relies on the availability of a virtual library of synthetically tractable molecules. Ugi reaction (UR) represents an important multi-component reaction (MCR) that reliably produces a peptidomimetic scaffold. Recent literature shows that a tactically assembled Ugi adduct can be subjected to further chemical modifications to yield a variety of rings and scaffolds, thus, renewing the interest in this old reaction. Given the reliability and efficiency of UR, we collated an UR derived library (URDL) of small molecules (total = 5773) for VS. The synthesis of the majority of URDL molecules may be carried out in 1-2 pots in a time and cost-effective manner. The detailed analysis of the average property and chemical space of URDL was also carried out using the open-source Datawarrior program. The comparison with FDA-approved oral drugs and inhibitors of protein-protein interactions (iPPIs) suggests URDL molecules are 'clean', drug-like, and conform to a structurally distinct space from the other two categories. The average physicochemical properties of compounds in the URDL library lie closer to iPPI molecules than oral drugs thus suggesting that the URDL resource can be applied to discover novel iPPI molecules. The URDL molecules consist of diverse ring systems, many of which have not been exploited yet for drug design. Thus, URDL represents a small virtual library of drug-like molecules with unexplored chemical space designed for VS. The structures of all molecules of URDL, oral drugs, and iPPI compounds are being made freely accessible as supplementary information for broader application.

6.
J Biol Chem ; 295(49): 16863-16876, 2020 12 04.
Article in English | MEDLINE | ID: mdl-32994223

ABSTRACT

RNase J enzymes are metallohydrolases that are involved in RNA maturation and RNA recycling, govern gene expression in bacteria, and catalyze both exonuclease and endonuclease activity. The catalytic activity of RNase J is regulated by multiple mechanisms which include oligomerization, conformational changes to aid substrate recognition, and the metal cofactor at the active site. However, little is known of how RNase J paralogs differ in expression and activity. Here we describe structural and biochemical features of two Staphylococcus epidermidis RNase J paralogs, RNase J1 and RNase J2. RNase J1 is a homodimer with exonuclease activity aided by two metal cofactors at the active site. RNase J2, on the other hand, has endonuclease activity and one metal ion at the active site and is predominantly a monomer. We note that the expression levels of these enzymes vary across Staphylococcal strains. Together, these observations suggest that multiple interacting RNase J paralogs could provide a strategy for functional improvisation utilizing differences in intracellular concentration, quaternary structure, and distinct active site architecture despite overall structural similarity.


Subject(s)
Bacterial Proteins/metabolism , Ribonucleases/metabolism , Staphylococcus epidermidis/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/classification , Bacterial Proteins/genetics , Biocatalysis , Catalytic Domain , Coenzymes/chemistry , Coenzymes/metabolism , Crystallography, X-Ray , Dimerization , Gene Expression Regulation, Bacterial , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Phylogeny , Protein Structure, Quaternary , RNA/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Ribonucleases/chemistry , Ribonucleases/classification , Ribonucleases/genetics , Substrate Specificity
7.
Biochem Biophys Res Commun ; 521(4): 900-906, 2020 01 22.
Article in English | MEDLINE | ID: mdl-31711645

ABSTRACT

Extra Cytoplasmic Function (ECF) σ factor/regulatory protein (anti-σ factor) pairs govern environment mediated changes in gene expression in bacteria. The release of the ECF σ factor from an inactive σ/anti-σ factor complex is triggered by specific environmental stimuli. The free σ factor then associates with the RNA polymerase and drives the expression of genes in its target regulon. Multiple ECF σ/anti-σ pairs ensure calibrated changes in the expression profile by correlating diverse environmental stimuli with changes in the intracellular levels of different ECF σ factors. Specificity in σ/anti-σ factor interaction is thus essential for accurate signal transduction. Here we describe experiments to evaluate interactions between different M. tuberculosis σ and anti-σ proteins in vitro. The interaction parameters suggest that cross-talk between non-cognate σ/anti-σ pairs is likely. The sequence and conformational determinants that govern interaction specificity in a σ/anti-σ complex are not immediately evident due to substantial structural conservation. Sequence-structure analysis of all σ/anti-σ pairs suggest that conserved residues are not the primary determinants of σ/anti-σ interactions-a finding that suggests a potential route to set tolerance limits in interaction specificity. Non-specific σ/anti-σ interactions are likely to be biologically significant as it can contribute to heterogeneity in cellular responses in a bacterial population under less stringent requirements. This finding is relevant for synthetic biology approaches to engineer bacteria using σ/anti-σ transcription initiation modules for diverse applications in biotechnology.


Subject(s)
Bacterial Proteins/chemistry , Mycobacterium tuberculosis/metabolism , Sigma Factor/chemistry , Sigma Factor/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Circular Dichroism , Computer Simulation , Mycobacterium tuberculosis/genetics , Protein Interaction Domains and Motifs , Surface Plasmon Resonance
8.
J Bacteriol ; 201(6)2019 03 15.
Article in English | MEDLINE | ID: mdl-30617240

ABSTRACT

Extracytoplasmic function σ factors that are stress inducible are often sequestered in an inactive complex with a membrane-associated anti-σ factor. Mycobacterium tuberculosis membrane-associated anti-σ factors have a small, stable RNA gene A (ssrA)-like degron for targeted proteolysis. Interaction between the unfoldase, ClpX, and a substrate with an accessible degron initiates energy-dependent proteolysis. Four anti-σ factors with a mutation in the degron provided a set of natural substrates to evaluate the influence of the degron on degradation strength in ClpX-substrate processivity. We note that a point mutation in the degron (X-Ala-Ala) leads to an order-of-magnitude difference in the dwell time of the substrate on ClpX. Differences in ClpX/anti-σ interactions were correlated with changes in unfoldase activities. Green fluorescent protein (GFP) chimeras or polypeptides with a length identical to that of the anti-σ factor degron also demonstrate degron-dependent variation in ClpX activities. We show that degron-dependent ClpX activity leads to differences in anti-σ degradation, thereby regulating the release of free σ from the σ/anti-σ complex. M. tuberculosis ClpX activity thus influences changes in gene expression by modulating the cellular abundance of ECF σ factors.IMPORTANCE The ability of Mycobacterium tuberculosis to quickly adapt to changing environmental stimuli occurs by maintaining protein homeostasis. Extracytoplasmic function (ECF) σ factors play a significant role in coordinating the transcription profile to changes in environmental conditions. Release of the σ factor from the anti-σ is governed by the ClpXP2P1 assembly. M. tuberculosis ECF anti-σ factors have an ssrA-like degron for targeted degradation. A point mutation in the degron leads to differences in ClpX-mediated proteolysis and affects the cellular abundance of ECF σ factors. ClpX activity thus synchronizes changes in gene expression with environmental stimuli affecting M. tuberculosis physiology.


Subject(s)
Endopeptidase Clp/metabolism , Gene Expression Regulation, Bacterial , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/metabolism , Repressor Proteins/metabolism , Sigma Factor/metabolism , DNA Mutational Analysis , Point Mutation , Proteolysis , Repressor Proteins/genetics
9.
Nucleic Acids Res ; 45(16): 9760-9772, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28934483

ABSTRACT

Extra-cytoplasmic function (ECF) σ-factors are widespread in bacteria, linking environmental stimuli with changes in gene expression. These transcription factors span several phylogenetically distinct groups and are remarkably diverse in their activation and regulatory mechanisms. Here, we describe the structural and biochemical features of a Mycobacterium tuberculosis ECF factor σJ that suggests that the SnoaL_2 domain at the C-terminus can modulate the activity of this initiation factor in the absence of a cognate regulatory anti-σ factor. M. tuberculosis σJ can bind promoter DNA in vitro; this interaction is substantially impaired by the removal of the SnoaL_2 domain. This finding is consistent with assays to evaluate σJ-mediated gene expression. Structural similarity of the SnoaL_2 domain with epoxide hydrolases also suggests a novel functional role for this domain. The conserved sequence features between M. tuberculosis σJ and other members of the ECF41 family of σ-factors suggest that the regulatory mechanism involving the C-terminal SnoaL_2 domain is likely to be retained in this family of proteins. These studies suggest that the ECF41 family of σ-factors incorporate features of both-the σ70 family and bacterial one-component systems thereby providing a direct mechanism to implement environment-mediated transcription changes.


Subject(s)
Mycobacterium tuberculosis/genetics , Promoter Regions, Genetic , Sigma Factor/chemistry , Sigma Factor/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Crystallography, X-Ray , Molecular Dynamics Simulation , Protein Conformation , Protein Domains , Sigma Factor/genetics , Surface Plasmon Resonance , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
10.
Biochem Biophys Res Commun ; 495(2): 2078-2084, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29242153

ABSTRACT

Polynucleotide phosphorylase catalyzes both 3'-5' exoribonuclease and polyadenylation reactions. The crystal structure of Staphylococcus epidermidis PNPase revealed a bound phosphate in the PH2 domain of each protomer coordinated by three adjacent serine residues. Mutational analysis suggests that phosphate coordination by these serine residues is essential to maintain the catalytic center in an active conformation. We note that PNPase forms a complex with RNase J1 and RNase J2 without substantially altering either exo-ribonuclease or polyadenylation activity of this enzyme. This decoupling of catalytic activity from protein-protein interactions suggests that association of these endo- or exo-ribonucleases with PNPase could be more relevant for cellular localization or concerted targeting of structured RNA for recycling.


Subject(s)
Molecular Docking Simulation , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/ultrastructure , Ribonucleases/chemistry , Ribonucleases/ultrastructure , Staphylococcus epidermidis/enzymology , Binding Sites , Enzyme Activation , Enzyme Stability , Models, Chemical , Multienzyme Complexes , Protein Binding , Protein Conformation , Structure-Activity Relationship , Substrate Specificity
11.
Biochemistry ; 56(16): 2209-2218, 2017 Apr 25.
Article in English | MEDLINE | ID: mdl-28355052

ABSTRACT

Transcription in prokaryotes is a multistep process and is primarily regulated at the initiation stage. σ factors are involved in promoter recognition and thus govern prokaryotic gene expression. Mycobacterium tuberculosis (Mtb) σ factors have been previously suggested as important drug targets through large-scale genome analyses. Here we demonstrate the feasibility of specific targeting of Mtb σ factors using designed peptides. A peptide library was generated using three-dimensional structural features corresponding to the interface regions of σ factors and the RNA polymerase. In silico optimization of the peptides, employing structural as well as sequence features, aided specific targeting of σA and σB. We synthesized and characterized the best hit peptide from the peptide library along with other control peptides and studied the interaction of these peptides with σB using biolayer interferometry. The experimental data validate the design strategy. These studies suggest the feasibility of designing specific peptides via in silico methods that bind σB with nanomolar affinity. We note that this strategy can be broadly applied to modulate prokaryotic transcription by designed peptides, thereby providing a tool for studying bacterial adaptation as well as host-pathogen interactions in infectious bacteria.


Subject(s)
Mycobacterium tuberculosis/metabolism , Peptide Fragments/metabolism , Sigma Factor/metabolism , Amino Acid Sequence , Circular Dichroism , DNA-Directed RNA Polymerases/chemistry , Kinetics , Ligands , Models, Molecular , Peptide Fragments/chemistry , Protein Binding , Sequence Homology, Amino Acid , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
12.
J Bacteriol ; 198(23): 3186-3199, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27645388

ABSTRACT

ArgO and LysE are members of the LysE family of exporter proteins and ordinarily mediate the export of l-arginine (Arg) in Escherichia coli and l-lysine (Lys) and Arg in Corynebacterium glutamicum, respectively. Under certain conditions, ArgO also mediates Lys export. To delineate the arrangement of ArgO in the cytoplasmic membrane of E. coli, we have employed a combination of cysteine accessibility in situ, alkaline phosphatase fusion reporters, and protein modeling to arrive at a topological model of ArgO. Our studies indicate that ArgO assumes an Nin-Cout configuration, potentially forming a five-transmembrane helix bundle flanked by a cytoplasmic N-terminal domain (NTD) comprising roughly its first 38 to 43 amino acyl residues and a short periplasmic C-terminal region (CTR). Mutagenesis studies indicate that the CTR, but not the NTD, is dispensable for ArgO function in vivo and that a pair of conserved aspartate residues, located near the opposing edges of the cytoplasmic membrane, may play a pivotal role in facilitating transmembrane Arg flux. Additional studies on amino acid substitutions that impair ArgO function in vivo and their derivatives bearing compensatory amino acid alterations indicate a role for intramolecular interactions in the Arg export mechanism, and some interactions are corroborated by normal-mode analyses. Lastly, our studies suggest that ArgO may exist as a monomer in vivo, thus highlighting the requirement for intramolecular interactions in ArgO, as opposed to interactions across multiple ArgO monomers, in the formation of an Arg-translocating conduit. IMPORTANCE: The orthologous proteins LysE of C. glutamicum and ArgO of E. coli function as exporters of the basic amino acids l-arginine and l-lysine and the basic amino acid l-arginine, respectively, and LysE can functionally substitute for ArgO when expressed in E. coli Notwithstanding this functional equivalence, studies reported here show that ArgO possesses a membrane topology that is distinct from that reported for LysE, with substantial variation in the topological arrangement of the proximal one-third portions of the two exporters. Additional genetic and in silico studies reveal the importance of (i) the cytoplasmic N-terminal domain, (ii) a pair of conserved aspartate residues, and (iii) potential intramolecular interactions in ArgO function and indicate that an Arg-translocating conduit is formed by a monomer of ArgO.


Subject(s)
Arginine/metabolism , Aspartic Acid/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Amino Acid Motifs , Biological Transport , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial , Protein Conformation , Protein Domains
13.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 5): 1216-25, 2015 May.
Article in English | MEDLINE | ID: mdl-25945586

ABSTRACT

Homoserine dehydrogenase (HSD) is an oxidoreductase in the aspartic acid pathway. This enzyme coordinates a critical branch point of the metabolic pathway that leads to the synthesis of bacterial cell-wall components such as L-lysine and m-DAP in addition to other amino acids such as L-threonine, L-methionine and L-isoleucine. Here, a structural rationale for the hydride-transfer step in the reaction mechanism of HSD is reported. The structure of Staphylococcus aureus HSD was determined at different pH conditions to understand the basis for the enhanced enzymatic activity at basic pH. An analysis of the crystal structure revealed that Lys105, which is located at the interface of the catalytic and cofactor-binding sites, could mediate the hydride-transfer step of the reaction mechanism. The role of Lys105 was subsequently confirmed by mutational analysis. Put together, these studies reveal the role of conserved water molecules and a lysine residue in hydride transfer between the substrate and the cofactor.


Subject(s)
Homoserine Dehydrogenase/chemistry , Homoserine Dehydrogenase/metabolism , Lysine/chemistry , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Staphylococcus aureus/enzymology , Binding Sites , Catalysis , Crystallography, X-Ray , Homoserine Dehydrogenase/genetics , Kinetics , Lysine/genetics , Lysine/metabolism , Models, Molecular , Mutant Proteins/genetics , Mutation/genetics , Protein Binding , Protein Conformation
14.
Nucleic Acids Res ; 41(5): 3414-23, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23314154

ABSTRACT

The relative levels of different σ factors dictate the expression profile of a bacterium. Extracytoplasmic function σ factors synchronize the transcriptional profile with environmental conditions. The cellular concentration of free extracytoplasmic function σ factors is regulated by the localization of this protein in a σ/anti-σ complex. Anti-σ factors are multi-domain proteins with a receptor to sense environmental stimuli and a conserved anti-σ domain (ASD) that binds a σ factor. Here we describe the structure of Mycobacterium tuberculosis anti-σ(D) (RsdA) in complex with the -35 promoter binding domain of σ(D) (σ(D)4). We note distinct conformational features that enable the release of σ(D) by the selective proteolysis of the ASD in RsdA. The structural and biochemical features of the σ(D)/RsdA complex provide a basis to reconcile diverse regulatory mechanisms that govern σ/anti-σ interactions despite high overall structural similarity. Multiple regulatory mechanisms embedded in an ASD scaffold thus provide an elegant route to rapidly re-engineer the expression profile of a bacterium in response to an environmental stimulus.


Subject(s)
Bacterial Proteins/chemistry , Mycobacterium tuberculosis , Sigma Factor/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , DNA, Bacterial/chemistry , Molecular Dynamics Simulation , Molecular Sequence Data , Promoter Regions, Genetic , Proteasome Endopeptidase Complex/chemistry , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Proteolysis
15.
J Bacteriol ; 196(15): 2876-88, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24858185

ABSTRACT

The Staphylococcus aureus agr quorum-sensing system plays a major role in the transition from the persistent to the virulent phenotype. S. aureus agr type I to IV strains are characterized by mutations in the sensor domain of the histidine kinase AgrC and differences in the sequences of the secreted autoinducing peptides (AIP). Here we demonstrate that interactions between the cytosolic domain of AgrC (AgrCCyto) and the response regulator domain of AgrA (AgrARR) dictate the spontaneity of the cellular response to AIP stimuli. The crystal structure of AgrCCyto provided a basis for a mechanistic model of AgrC-AgrA interactions. This model enabled an analysis of the biochemical and biophysical parameters of AgrC-AgrA interactions in the context of the conformational features of the AgrC-AgrA complex. This analysis revealed distinct sequence and conformational features that determine the affinity, specificity, and kinetics of the phosphotransfer reaction. This step, which governs the response time for transcriptional reengineering triggered by an AIP stimulus, is independent of the agr type and similar for agonist and antagonist stimuli. These experimental data could serve as a basis on which to validate simulations of the quorum-sensing response and for strategies that employ the agr quorum-sensing system to combat biofilm formation in S. aureus infections.


Subject(s)
Bacterial Proteins/metabolism , Quorum Sensing/physiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/physiology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Biofilms , Crystallography , Cytosol/metabolism , Dimerization , Humans , Kinetics , Models, Biological , Models, Molecular , Peptides, Cyclic/chemistry , Peptides, Cyclic/genetics , Peptides, Cyclic/metabolism , Phenotype , Phosphorylation , Protein Binding , Protein Conformation , Protein Kinases/chemistry , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Structure, Tertiary , Recombinant Proteins , Staphylococcus aureus/genetics , Time Factors
16.
Article in English | MEDLINE | ID: mdl-24192352

ABSTRACT

Staphylococcus aureus is a Gram-positive nosocomial pathogen. The prevalence of multidrug-resistant S. aureus strains in both hospital and community settings makes it imperative to characterize new drug targets to combat S. aureus infections. In this context, enzymes involved in cell-wall maintenance and essential amino-acid biosynthesis are significant drug targets. Homoserine dehydrogenase (HSD) is an oxidoreductase that is involved in the reversible conversion of L-aspartate semialdehyde to L-homoserine in a dinucleotide cofactor-dependent reduction reaction. HSD is thus a crucial intermediate enzyme linked to the biosynthesis of several essential amino acids such as lysine, methionine, isoleucine and threonine.


Subject(s)
Bacterial Proteins/chemistry , Homoserine Dehydrogenase/chemistry , Staphylococcus aureus/enzymology , Bacterial Proteins/isolation & purification , Crystallization , Homoserine Dehydrogenase/isolation & purification , Hydrogen-Ion Concentration , Temperature , X-Ray Diffraction
17.
BMJ Case Rep ; 16(1)2023 Jan 12.
Article in English | MEDLINE | ID: mdl-36634990

ABSTRACT

The paraneoplastic leukemoid reaction is a rare haematological paraneoplastic syndrome, which is typically seen with solid tumours and squamous cell carcinomas. As an indication of bone marrow infiltration and malignancy involvement, it indicates a poor outcome and a grave prognosis. We report a woman in her 50s, who presented with an ulcer over the right forearm. Biopsy revealed squamous cell carcinoma. The patient underwent radiological investigations, which showed the presence of metastatic squamous cell carcinoma. Incidentally, the patient was found to have leucocytosis, which was attributed to a paraneoplastic leukemoid reaction, after ruling out all other causes of leukemoid reaction. Due to metastatic disease, the patient was planned for palliative radiotherapy and the best supportive care.


Subject(s)
Carcinoma, Squamous Cell , Leukemoid Reaction , Paraneoplastic Syndromes , Female , Humans , Leukemoid Reaction/diagnosis , Leukemoid Reaction/etiology , Forearm , Carcinoma, Squamous Cell/complications , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/radiotherapy , Leukocytosis/complications , Paraneoplastic Syndromes/etiology , Paraneoplastic Syndromes/complications
18.
Methods Enzymol ; 692: 177-215, 2023.
Article in English | MEDLINE | ID: mdl-37925179

ABSTRACT

RNase J is involved in RNA maturation as well as degradation of RNA to the level of mononucleotides. This enzyme plays a vital role in maintaining intracellular RNA levels and governs different steps of the cellular metabolism in bacteria. RNase J is the first ribonuclease that was shown to have both endonuclease and 5'-3' exonuclease activity. RNase J enzymes can be identified by their characteristic sequence features and domain architecture. The quaternary structure of RNase J plays a role in regulating enzyme activity. The structure of RNase J has been characterized from several homologs. These reveal extensive overall structural similarity alongside a distinct active site topology that coordinates a metal cofactor. The metal cofactor is essential for catalytic activity. The catalytic activity of RNase J is influenced by oligomerization, the choice and stoichiometry of metal cofactors, and the 5' phosphorylation state of the RNA substrate. Here we describe the sequence and structural features of RNase J alongside phylogenetic analysis and reported functional roles in diverse organisms. We also provide a detailed purification strategy to obtain an RNase J enzyme sample with or without a metal cofactor. Different methods to identify the nature of the bound metal cofactor, the binding affinity and stoichiometry are presented. Finally, we describe enzyme assays to characterize RNase J using radioactive and fluorescence-based strategies with diverse RNA substrates.


Subject(s)
Endoribonucleases , Ribonucleases , Ribonucleases/metabolism , Phylogeny , Endoribonucleases/metabolism , RNA/chemistry , Ribonuclease, Pancreatic , Metals
19.
Int J Low Extrem Wounds ; 22(1): 163-167, 2023 Mar.
Article in English | MEDLINE | ID: mdl-33527864

ABSTRACT

Opportunistic fungal infections are known to occur in immunocompromised patients. Mucormycosis is one of the most common opportunistic fungal infections with significant mortality rates. In this article, we present a case of an adult female, a known diabetic who presented with fever and pus discharge from the amputation site of toes in the left foot with blackening of the foot. Examination revealed gangrenous changes of the left foot with no distal pulses palpable. Computed tomography angiogram revealed no flow of blood in distal vessels of the left lower limb. Left below knee guillotine amputation was done. Intraoperative biopsy of the neurovascular bundle revealed invasive neuromucormycosis. She was started on liposomal amphotericin B. The wound started granulating after a few days with serial dressings and the patient was planned for split skin grafting.


Subject(s)
Diabetes Mellitus , Mucormycosis , Adult , Humans , Female , Mucormycosis/complications , Mucormycosis/diagnosis , Mucormycosis/pathology , Gangrene , Foot , Tibial Nerve/pathology
20.
ACS Chem Biol ; 18(7): 1487-1499, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37319329

ABSTRACT

High temperature requirement A (HtrA) are allosterically regulated enzymes wherein effector binding to the PDZ domain triggers proteolytic activity. Yet, it remains unclear if the inter-residue network governing allostery is conserved across HtrA enzymes. Here, we investigated and identified the inter-residue interaction networks by molecular dynamics simulations on representative HtrA proteases, Escherichia coli DegS and Mycobacterium tuberculosis PepD, in effector-bound and free forms. This information was used to engineer mutations that could potentially perturb allostery and conformational sampling in a different homologue, M. tuberculosis HtrA. Mutations in HtrA perturbed allosteric regulation─a finding consistent with the hypothesis that the inter-residue interaction network is conserved across HtrA enzymes. Electron density from data collected on cryo-protected HtrA crystals revealed that mutations altered the topology of the active site. Ensemble models fitted into electron density calculated from room-temperature diffraction data showed that only a fraction of these models had a catalytically competent active site conformation alongside a functional oxyanion hole thus providing experimental evidence that these mutations influenced conformational sampling. Mutations at analogous positions in the catalytic domain of DegS perturbed the coupling between effector binding and proteolytic activity, thus confirming the role of these residues in the allosteric response. The finding that a perturbation in the conserved inter-residue network alters conformational sampling and the allosteric response suggests that an ensemble allosteric model best describes regulated proteolysis in HtrA enzymes.


Subject(s)
Endopeptidases , Escherichia coli , Temperature , Endopeptidases/metabolism , Escherichia coli/metabolism , Molecular Dynamics Simulation , Allosteric Regulation , Catalytic Domain
SELECTION OF CITATIONS
SEARCH DETAIL