Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Biochem Biophys Res Commun ; 722: 150150, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-38805787

ABSTRACT

Two component system bacterial response regulators are typically DNA-binding proteins which enable the genetic regulation of many adaptive bacterial behaviors. Despite structural similarity across response regulator families, there is a diverse array of DNA-binding mechanisms. Bacteria usually encode several dozen two-component system response regulators, but Francisella tularensis only encodes three. Due to their simplified response regulatory network, Francisella species are a model for studying the role of response regulator proteins in virulence. Here, we show that Francisella response regulators QseB, KdpE, and BfpR all utilize different DNA-binding mechanisms. Our evidence suggests that QseB follows a simple mechanism whereby it binds a single inverted repeat sequence with a higher affinity upon phosphorylation. This behavior is independent of whether QseB is a positive or negative regulator of the gene as demonstrated by qseB and priM promoter sequences, respectively. Similarly, KdpE binds DNA more tightly upon phosphorylation, but also exhibits a cooperative binding isotherm. While we propose a KdpE binding site, it is possible that KdpE has a complex DNA-binding mechanism potentially involving multiple copies of KdpE being recruited to a promoter region. Finally, we show that BfpR appears to bind a region of its own promoter sequence with a lower affinity upon phosphorylation. Further structural and enzymatic work will need to be performed to deconvolute the KdpE and BfpR binding mechanisms.


Subject(s)
Bacterial Proteins , DNA-Binding Proteins , Protein Binding , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , Phosphorylation , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/chemistry , Gene Expression Regulation, Bacterial , DNA, Bacterial/metabolism , DNA, Bacterial/genetics , Francisella tularensis/metabolism , Francisella tularensis/genetics , Binding Sites , Promoter Regions, Genetic , Francisella
2.
J Mol Biol ; 435(3): 167923, 2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36535428

ABSTRACT

Bacterial biofilms are notorious for their ability to protect bacteria from environmental challenges, most importantly the action of antibiotics. Bacillus subtilis is an extensively studied model organism used to understand the process of biofilm formation. A complex network of principal regulatory proteins including Spo0A, AbrB, AbbA, Abh, SinR, SinI, SlrR, and RemA, work in concert to transition B. subtilis from the free-swimming planktonic state to the biofilm state. In this review, we explore, connect, and summarize decades worth of structural and biochemical studies that have elucidated this protein signaling network. Since structure dictates function, unraveling aspects of protein molecular mechanisms will allow us to devise ways to exploit critical features of the biofilm regulatory pathway, such as possible therapeutic intervention. This review pools our current knowledge base of B. subtilis biofilm regulatory proteins and highlights potential therapeutic intervention points.


Subject(s)
Bacillus subtilis , Bacterial Proteins , Biofilms , Bacillus subtilis/genetics , Bacillus subtilis/physiology , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Transcription Factors/metabolism
3.
Biochemistry ; 61(24): 2948-2960, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36454711

ABSTRACT

Multidrug-resistant bacteria cause immense public health concerns as once effective antibiotics no longer work against even common infections. Concomitantly, there has been a decline in the discovery of new antibiotics, and the current global clinical pipeline is woefully inadequate, especially against resistant Gram-negative bacteria. One major contribution to Gram-negative resistance is the presence of a protective outer membrane. Consequently, an appealing option for tackling resistance is to adversely affect that outer membrane. With that in mind, we define the response regulator PhoP as a target for new 2-aminoimidazole compounds and show that they affect the integrity of the outer membrane in resistant strains of Escherichia coli and Klebsiella pneumoniae. We also provide empirical evidence for the 2-aminoimidazole mechanism of action.


Subject(s)
Anti-Bacterial Agents , Escherichia coli Proteins , Anti-Bacterial Agents/pharmacology , Gram-Negative Bacteria , Imidazoles/pharmacology , Drug Resistance, Multiple, Bacterial , Escherichia coli/metabolism , DNA , Microbial Sensitivity Tests , Escherichia coli Proteins/pharmacology
4.
J Bacteriol ; 204(2): e0049421, 2022 02 15.
Article in English | MEDLINE | ID: mdl-34871031

ABSTRACT

Acinetobacter baumannii is a common nosocomial pathogen that utilizes numerous mechanisms to aid its survival in both the environment and the host. Coordination of such mechanisms requires an intricate regulatory network. We report here that A. baumannii can directly regulate several stress-related pathways via the two-component regulatory system BfmRS. Similar to previous studies, results from transcriptomic analysis showed that mutation of the BfmR response regulator causes dysregulation of genes required for the oxidative stress response, the osmotic stress response, the misfolded protein/heat shock response, Csu pilus/fimbria production, and capsular polysaccharide biosynthesis. We also found that the BfmRS system is involved in controlling siderophore biosynthesis and transport, and type IV pili production. We provide evidence that BfmR binds to various stress-related promoter regions and show that BfmR alone can directly activate transcription of some stress-related genes. Additionally, we show that the BfmS sensor kinase acts as a BfmR phosphatase to negatively regulate BfmR activity. This work highlights the importance of the BfmRS system in promoting survival of A. baumannii. IMPORTANCE Acinetobacter baumannii is a nosocomial pathogen that has extremely high rates of multidrug resistance. This organism's ability to endure stressful conditions is a key part of its ability to spread in the hospital environment and cause infections. Unlike other members of the gammaproteobacteria, A. baumannii does not encode a homolog of the RpoS sigma factor to coordinate its stress response. Here, we demonstrate that the BfmRS two-component system directly controls the expression of multiple stress resistance genes. Our findings suggest that BfmRS is central to a unique scheme of general stress response regulation by A. baumannii.


Subject(s)
Acinetobacter baumannii/genetics , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Stress, Physiological/genetics , Acinetobacter baumannii/metabolism , Acinetobacter baumannii/pathogenicity , Bacterial Proteins/metabolism , Biofilms/growth & development , Mutation , Promoter Regions, Genetic , Virulence/genetics
5.
J Biochem ; 170(6): 787-800, 2022 Jan 07.
Article in English | MEDLINE | ID: mdl-34585233

ABSTRACT

Acinetobacter baumannii is an insidious emerging nosocomial pathogen that has developed resistance to all available antimicrobials, including the last resort antibiotic, colistin. Colistin resistance often occurs due to mutations in the PmrAB two-component regulatory system. To better understand the regulatory mechanisms contributing to colistin resistance, we have biochemically characterized the A. baumannii PmrA response regulator. Initial DNA-binding analysis shows that A. baumannii PmrA bound to the Klebsiella pneumoniae PmrA box motif. This prompted analysis of the putative A. baumannii PmrAB regulon that indicated that the A. baumannii PmrA consensus box is 5'-HTTAAD N5 HTTAAD. Additionally, we provide the first structural information for the A. baumannii PmrA N-terminal domain through X-ray crystallography and we present a full-length model using molecular modelling. From these studies, we were able to infer the effects of two critical PmrA mutations, PmrA::I13M and PmrA::P102R, both of which confer increased colistin resistance. Based on these data, we suggest structural and dynamic reasons for how these mutations can affect PmrA function and hence encourage resistive traits. Understanding these mechanisms will aid in the development of new targeted antimicrobial therapies. Graphical Abstract.


Subject(s)
Acinetobacter baumannii/chemistry , Bacterial Proteins/chemistry , Colistin , DNA, Bacterial/chemistry , Drug Resistance, Bacterial , Mutation , Acinetobacter baumannii/genetics , Acinetobacter baumannii/metabolism , Amino Acid Motifs , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Crystallography, X-Ray , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Protein Domains
6.
Article in English | MEDLINE | ID: mdl-32232010

ABSTRACT

Response regulators are a critical part of the two-component system of gene expression regulation in bacteria, transferring a signal from a sensor kinase into DNA binding activity resulting in alteration of gene expression. In this study, we investigated a previously uncharacterized response regulator in Francisella novicida, FTN_1452 that we have named BfpR (Biofilm-regulating Francisella protein Regulator, FTN_1452). In contrast to another Francisella response regulator, QseB/PmrA, BfpR appears to be a negative regulator of biofilm production, and also a positive regulator of antimicrobial peptide resistance in this bacterium. The protein was crystallized and X-ray crystallography studies produced a 1.8 Å structure of the BfpR N-terminal receiver domain revealing interesting insight into its potential interaction with the sensor kinase. Structural analysis of BfpR places it in the OmpR/PhoP family of bacterial response regulators along with WalR and ResD. Proteomic and transcriptomic analyses suggest that BfpR overexpression affects expression of the critical Francisella virulence factor iglC, as well as other proteins in the bacterium. We demonstrate that mutation of bfpR is associated with an antimicrobial peptide resistance phenotype, a phenotype also associated with other response regulators, for the human cathelicidin peptide LL-37 and a sheep antimicrobial peptide SMAP-29. F. novicida with mutated bfpR replicated better than WT in intracellular infection assays in human-derived macrophages suggesting that the down-regulation of iglC expression in bfpR mutant may enable this intracellular replication to occur. Response regulators have been shown to play important roles in the regulation of bacterial biofilm production. We demonstrate that F. novicida biofilm formation was highly increased in the bfpR mutant, corresponding to altered glycogen synthesis. Waxworm infection experiments suggest a role of BfpR as a negative modulator of iglC expression with de-repression by Mg2+. In this study, we find that the response regulator BfpR may be a negative regulator of biofilm formation, and a positive regulator of antimicrobial peptide resistance in F. novicida.


Subject(s)
Bacterial Proteins/metabolism , Biofilms/growth & development , Francisella/physiology , Pore Forming Cytotoxic Proteins/pharmacology , Virulence Factors/genetics , Animals , Antimicrobial Cationic Peptides/pharmacology , Bacterial Proteins/genetics , Blood Proteins/pharmacology , Cathelicidins/pharmacology , Drug Resistance, Bacterial , Francisella/drug effects , Francisella/genetics , Gene Expression Regulation, Bacterial , Macrophages/microbiology , Moths/microbiology , Mutation , Virulence/genetics , Virulence Factors/metabolism
7.
J Mol Biol ; 432(2): 343-357, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31493408

ABSTRACT

Bacteria have developed numerous protection strategies to ensure survival in harsh environments, with perhaps the most robust method being the formation of a protective biofilm. In biofilms, bacterial cells are embedded within a matrix that is composed of a complex mixture of polysaccharides, proteins, and DNA. The gram-positive bacterium Bacillus subtilis has become a model organism for studying regulatory networks directing biofilm formation. The phenotypic transition from a planktonic to biofilm state is regulated by the activity of the transcriptional repressor, SinR, and its inactivation by its primary antagonist, SinI. In this work, we present the first full-length structural model of tetrameric SinR using a hybrid approach combining high-resolution solution nuclear magnetic resonance (NMR), chemical cross-linking, mass spectrometry, and molecular docking. We also present the solution NMR structure of the antagonist SinI dimer and probe the mechanism behind the SinR-SinI interaction using a combination of biochemical and biophysical techniques. As a result of these findings, we propose that SinI utilizes a residue replacement mechanism to block SinR multimerization, resulting in diminished DNA binding and concomitant decreased repressor activity. Finally, we provide an evidence-based mechanism that confirms how disruption of the SinR tetramer by SinI regulates gene expression.


Subject(s)
Bacillus subtilis/ultrastructure , Bacterial Proteins/ultrastructure , DNA-Binding Proteins/ultrastructure , Amino Acid Sequence/genetics , Bacillus subtilis/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Biofilms/growth & development , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Gene Expression Regulation, Bacterial/genetics , Molecular Docking Simulation , Mutation/genetics , Protein Binding/genetics , Protein Conformation
8.
Front Mol Biosci ; 5: 15, 2018.
Article in English | MEDLINE | ID: mdl-29487854

ABSTRACT

2-aminoimidazole (2-AI) compounds inhibit the formation of bacterial biofilms, disperse preformed biofilms, and re-sensitize multidrug resistant bacteria to antibiotics. 2-AIs have previously been shown to interact with bacterial response regulators, but the mechanism of interaction is still unknown. Response regulators are one part of two-component systems (TCS). TCSs allow cells to respond to changes in their environment, and are used to trigger quorum sensing, virulence factors, and antibiotic resistance. Drugs that target the TCS signaling process can inhibit pathogenic behavior, making this a potent new therapeutic approach that has not yet been fully exploited. We previously laid the groundwork for the interaction of the Acinetobacter baumannii response regulator BfmR with an early 2-AI derivative. Here, we further investigate the response regulator/2-AI interaction and look at a wider library of 2-AI compounds. By combining molecular modeling with biochemical and cellular studies, we expand on a potential mechanism for interaction between response regulators and 2-AIs. We also establish that Francisella tularensis/novicida, encoding for only three known response regulators, can be a model system to study the interaction between 2-AIs and response regulators. We show that knowledge gained from studying Francisella can be applied to the more complex A. baumannii system, which contains over 50 response regulators. Understanding the impact of 2-AIs on response regulators and their mechanism of interaction will lead to the development of more potent compounds that will serve as adjuvant therapies to broad-range antibiotics.

9.
J Mol Biol ; 430(6): 806-821, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29438671

ABSTRACT

The rise of drug-resistant bacterial infections coupled with decreasing antibiotic efficacy poses a significant challenge to global health care. Acinetobacter baumannii is an insidious, emerging bacterial pathogen responsible for severe nosocomial infections aided by its ability to form biofilms. The response regulator BfmR, from the BfmR/S two-component system, is the master regulator of biofilm initiation in A. baumannii and is a tractable therapeutic target. Here we present the structure of A. baumannii BfmR using a hybrid approach combining X-ray crystallography, nuclear magnetic resonance spectroscopy, chemical crosslinking mass spectrometry, and molecular modeling. We also show that BfmR binds the previously proposed bfmRS promoter sequence with moderate affinity. While BfmR shares many traits with other OmpR/PhoB family response regulators, some unusual properties were observed. Most importantly, we observe that when phosphorylated, BfmR binds this promoter sequence with a lower affinity than when not phosphorylated. All other OmpR/PhoB family members studied to date show an increase in DNA-binding affinity upon phosphorylation. Understanding the structural and biochemical mechanisms of BfmR will aid in the development of new antimicrobial therapies.


Subject(s)
Acinetobacter baumannii/metabolism , Bacterial Proteins/chemistry , Biofilms/growth & development , DNA-Binding Proteins/metabolism , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/genetics , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Biofilms/drug effects , Cloning, Molecular , Crystallography, X-Ray , DNA-Binding Proteins/genetics , Gene Expression Regulation, Bacterial , Models, Molecular , Phosphorylation , Promoter Regions, Genetic , Protein Conformation
10.
Mol Microbiol ; 106(2): 223-235, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28755524

ABSTRACT

With antibiotic resistance increasing at alarming rates, targets for new antimicrobial therapies must be identified. A particularly promising target is the bacterial two-component system. Two-component systems allow bacteria to detect, evaluate and protect themselves against changes in the environment, such as exposure to antibiotics and also to trigger production of virulence factors. Drugs that target the response regulator portion of two-component systems represent a potent new approach so far unexploited. Here, we focus efforts on the highly virulent bacterium Francisella tularensis tularensis. Francisella contains only three response regulators, making it an ideal system to study. In this study, we initially present the structure of the N-terminal domain of QseB, the response regulator responsible for biofilm formation. Subsequently, using binding assays, computational docking and cellular studies, we show that QseB interacts with2-aminoimidazole based compounds that impede its function. This information will assist in tailoring compounds to act as adjuvants that will enhance the effect of antibiotics.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Francisella tularensis/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/ultrastructure , Biofilms/drug effects , Gene Expression Regulation, Bacterial/genetics , Imidazoles/metabolism , Imidazoles/pharmacology , Protein Binding , Virulence/drug effects , Virulence Factors/genetics , Virulence Factors/metabolism
11.
Drug Des Devel Ther ; 11: 153-162, 2017.
Article in English | MEDLINE | ID: mdl-28138218

ABSTRACT

2-Aminoimidazole (2-AI)-based compounds have been shown to efficiently disrupt biofilm formation, disperse existing biofilms, and resensitize numerous multidrug-resistant bacteria to antibiotics. Using Pseudomonas aeruginosa and Staphylococcus aureus, we provide initial pharmacological studies regarding the application of a 2-AI as a topical adjuvant for persistent dermal infections. In vitro assays indicated that the 2-AI H10 is nonbactericidal, resensitizes bacteria to antibiotics, does not harm the integument, and promotes wound healing. Furthermore, in vivo application of H10 on swine skin caused no gross abnormalities or immune reactions. Taken together, these results indicate that H10 represents a promising lead dermal adjuvant compound.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Imidazoles/therapeutic use , Skin Diseases, Bacterial/drug therapy , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Chemotherapy, Adjuvant , Dose-Response Relationship, Drug , Humans , Imidazoles/administration & dosage , Imidazoles/pharmacology , Microbial Sensitivity Tests , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/growth & development , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Structure-Activity Relationship
12.
Medchemcomm ; 7(1): 128-131, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26877862

ABSTRACT

A screen of 20 compounds identified small molecule adjuvants capable of potentiating anitbiotic activty against Francisella philomiragia. Analogue synthesis of an initial hit compound led to the discovery of a potentially new class of small molecule adjuvants containg an indole core. The lead compound was able to lower the MIC of colistin by 32-fold against intrinsically resistant F. philomiragia.

14.
Curr Drug Deliv ; 12(2): 223-30, 2015.
Article in English | MEDLINE | ID: mdl-25348099

ABSTRACT

Acinetobacter baumannii has quickly become one of the most insidious and prevalent nosocomial infections. Recently, the reverse-amide class of 2-aminoimidazole compounds (RA-2AI) was found both to prevent A. baumannii biofilm formation and also to disperse preexisting formations, putatively through interactions with cytosolic response regulators. Here we focus on how this class of antibiofilm agent traverses cellular membranes. Following the discovery of dosage-dependent growth rate changes, the cellular effects of RA-2AI were investigated using a combination of molecular assays and microscopic techniques. It was found that RA-2AI exposure has measureable effects on the bacterial membranes, resulting in a period of increased permeability and visible structural aberrations. Based on these results, we propose a model that describes how the structure of RA-2AI allows it to insert itself into and disrupt the fluidity of the membrane, creating an opportunity for increased molecular permeability.


Subject(s)
Acinetobacter baumannii/drug effects , Amides/chemistry , Biofilms/drug effects , Cell Membrane Permeability/drug effects , Imidazoles/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Cells, Cultured , Cross Infection , Imidazoles/pharmacology , Microbial Sensitivity Tests
15.
Mar Drugs ; 13(1): 65-75, 2014 Dec 26.
Article in English | MEDLINE | ID: mdl-25548974

ABSTRACT

Aquatic microbes produce diverse secondary metabolites with interesting biological activities. Cytotoxic metabolites have the potential to become lead compounds or drugs for cancer treatment. Many cytotoxic compounds, however, show undesirable toxicity at higher concentrations. Such undesirable activity may be reduced or eliminated by using lower doses of the cytotoxic compound in combination with another compound that modulates its activity. Here, we have examined the cytotoxicity of four microbial metabolites [ethyl N-(2-phenethyl) carbamate (NP-1), Euglenophycin, Anabaenopeptin, and Glycolipid 652] using three in vitro cell lines [human breast cancer cells (MCF-7), mouse neuroblastoma cells (N2a), and rat pituitary epithelial cells (GH4C1)]. The compounds showed variable cytotoxicity, with Euglenophycin displaying specificity for N2a cells. We have also examined the modulatory power of NP-1 on the cytotoxicity of the other three compounds and found that at a permissible concentration (125 µg/mL), NP-1 sensitized N2a and MCF-7 cells to Euglenophycin and Glycolipid 652 induced cytotoxicity.


Subject(s)
Adjuvants, Pharmaceutic/therapeutic use , Antineoplastic Agents/therapeutic use , Biological Products/therapeutic use , Glycolipids/therapeutic use , Animals , Antineoplastic Agents/administration & dosage , Biological Products/administration & dosage , Biological Products/pharmacology , Brain Neoplasms/drug therapy , Cell Line, Tumor/drug effects , Glycolipids/administration & dosage , Humans , MCF-7 Cells/drug effects , Marine Toxins/administration & dosage , Marine Toxins/therapeutic use , Mice , Neuroblastoma/drug therapy , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/therapeutic use , Piperidines/administration & dosage , Piperidines/therapeutic use , Rats , Seawater/microbiology , Thiazoles/administration & dosage , Thiazoles/therapeutic use
16.
Structure ; 22(11): 1650-6, 2014 Nov 04.
Article in English | MEDLINE | ID: mdl-25308864

ABSTRACT

The AbrB protein from Bacillus subtilis is a DNA-binding global regulator controlling the onset of a vast array of protective functions under stressful conditions. Such functions include biofilm formation, antibiotic production, competence development, extracellular enzyme production, motility, and sporulation. AbrB orthologs are known in a variety of prokaryotic organisms, most notably in all infectious strains of Clostridia, Listeria, and Bacilli. Despite its central role in bacterial response and defense, its structure has been elusive because of its highly dynamic character. Orienting its N- and C-terminal domains with respect to one another has been especially problematic. Here, we have generated a structure of full-length, tetrameric AbrB using nuclear magnetic resonance, chemical crosslinking, and mass spectrometry. We note that AbrB possesses a strip of positive electrostatic potential encompassing its DNA-binding region and that its C-terminal domain aids in DNA binding.


Subject(s)
Bacillus subtilis/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , DNA, Bacterial/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Binding Sites , Magnetic Resonance Spectroscopy , Mass Spectrometry , Molecular Docking Simulation , Protein Multimerization , Protein Structure, Secondary
17.
J Med Chem ; 57(17): 7450-8, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25137478

ABSTRACT

Recent efforts toward combating antibiotic resistance in bacteria have focused on Gram-positive bacteria; however, multidrug-resistant Gram-negative bacteria pose a significant risk to public health. An orthogonal approach to the development of new antibiotics is to develop adjuvant compounds that enhance the susceptibility of drug-resistant strains of bacteria to currently approved antibiotics. This paper describes the synthesis and biological activity of a library of aryl amide 2-aminoimidazoles based on a lead structure from an initial screen. A small molecule was identified from this library that is capable of lowering the minimum inhibitory concentration of ß-lactam antibiotics by up to 64-fold.


Subject(s)
Drug Resistance, Multiple, Bacterial/drug effects , Gram-Negative Bacteria/drug effects , Small Molecule Libraries/pharmacology , beta-Lactam Resistance/drug effects , Animals , Cell Line , Cell Survival/drug effects , Gram-Negative Bacteria/classification , Hemolysis/drug effects , Humans , Keratinocytes/cytology , Keratinocytes/drug effects , Microbial Sensitivity Tests , Sheep , Small Molecule Libraries/chemistry
18.
J Mol Biol ; 426(9): 1911-24, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24534728

ABSTRACT

Bacteria respond to adverse environmental conditions by switching on the expression of large numbers of genes that enable them to adapt to unfavorable circumstances. In Bacillus subtilis, many adaptive genes are under the negative control of the global transition state regulator, the repressor protein AbrB. Stressful conditions lead to the de-repression of genes under AbrB control. Contributing to this de-repression is AbbA, an anti-repressor that binds to and blocks AbrB from binding to DNA. Here, we have determined the NMR structure of the functional AbbA dimer, confirmed that it binds to the N-terminal DNA-binding domain of AbrB, and have provided an initial description for the interaction using computational docking procedures. Interestingly, we show that AbbA has structural and surface characteristics that closely mimic the DNA phosphate backbone, enabling it to readily carry out its physiological function.


Subject(s)
Bacillus subtilis/chemistry , Bacterial Proteins/chemistry , Bacillus subtilis/metabolism , Bacterial Proteins/metabolism , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Docking Simulation , Protein Conformation , Protein Multimerization
19.
Biomol NMR Assign ; 8(1): 155-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23475644

ABSTRACT

Bacillus subtilis is a soil-dwelling Gram-positive bacterial species that has been extensively studied as a model of biofilm formation and stress-induced cellular differentiation. The tetrameric protein, SinR, has been identified as a master regulator for biofilm formation and linked to the regulation of the early transition states during cellular stress response, such as motility and biofilm-linked biosynthetic genes. SinR is a 111-residue protein that is active as a dimer of dimers, composed of two distinct domains, a DNA-binding helix-turn-helix N-terminus domain and a C-terminal multimerization domain. In order for biofilm formation to proceed, the antagonist, SinI, must inactivate SinR. This interaction results in a dramatic structural rearrangement of both proteins. Here we report the full-length backbone and side chain chemical shift values in addition to the experimentally derived secondary structure predictions as the first step towards directly studying the complex interaction dynamics between SinR and SinI.


Subject(s)
Bacillus subtilis/metabolism , Bacterial Proteins/chemistry , Biofilms , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Secondary
20.
Biomol NMR Assign ; 8(1): 67-70, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23264007

ABSTRACT

Acinetobacter baumannii is a Gram-negative pathogen responsible for severe nocosomial infections by forming biofilms in healthcare environments. The two-domain response regulator BfmR has been shown to be the master controller for biofilm formation. Inactivation of BfmR resulted in an abolition of pili production and consequently biofilm creation. Here we report backbone and sidechain resonance assignments and secondary structure prediction for the C-terminal domain of BfmR (residues 130-238) from A. baumannii.


Subject(s)
Acinetobacter baumannii/metabolism , Bacterial Proteins/chemistry , Nuclear Magnetic Resonance, Biomolecular , Protein Structure, Secondary , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL
...