Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Proteome Res ; 20(10): 4728-4745, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34469172

ABSTRACT

Chronic low-dose exposure to organophosphorus pesticides is associated with the risk of neurodegenerative disease. The mechanism of neurotoxicity is independent of acetylcholinesterase inhibition. Adducts on tyrosine, lysine, threonine, and serine can occur after exposure to organophosphorus pesticides, the most stable being adducts on tyrosine. Rabbit monoclonal 1C6 to diethoxyphosphate-modified tyrosine (depY) was created by single B cell cloning. The amino acid sequence and binding constant (Kd 3.2 × 10-8 M) were determined. Cultured human neuroblastoma SH-SY5Y and mouse neuroblastoma N2a cells incubated with a subcytotoxic dose of 10 µM chlorpyrifos oxon contained depY-modified proteins detected by monoclonal 1C6 on Western blots. depY-labeled peptides from tryptic digests of cell lysates were immunopurified by binding to immobilized 1C6. Peptides released with 50% acetonitrile and 1% formic acid were analyzed by liquid chromatography tandem mass spectrometry (LC-MS/MS) on an Orbitrap Fusion Lumos mass spectrometer. Protein Prospector database searches identified 51 peptides modified on tyrosine by diethoxyphosphate in SH-SY5Y cell lysate and 73 diethoxyphosphate-modified peptides in N2a cell lysate. Adducts appeared most frequently on the cytoskeleton proteins tubulin, actin, and vimentin. It was concluded that rabbit monoclonal 1C6 can be useful for studies that aim to understand the mechanism of neurotoxicity resulting from low-dose exposure to organophosphorus pesticides.


Subject(s)
Neurodegenerative Diseases , Pesticides , Acetylcholinesterase , Animals , B-Lymphocytes , Cells, Cultured , Chlorpyrifos/analogs & derivatives , Chromatography, Liquid , Cloning, Molecular , Mice , Organophosphorus Compounds , Peptides , Pesticides/toxicity , Tandem Mass Spectrometry
2.
Chem Res Toxicol ; 30(12): 2218-2228, 2017 12 18.
Article in English | MEDLINE | ID: mdl-29137457

ABSTRACT

Acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) are irreversibly inhibited by organophosphorus pesticides through formation of a covalent bond with the active site serine. Proteins that have no active site serine, for example albumin, are covalently modified on tyrosine and lysine. Chronic illness from pesticide exposure is not explained by inhibition of AChE and BChE. Our goal was to produce a monoclonal antibody that recognizes proteins diethoxyphosphorylated on tyrosine. Diethoxyphosphate-tyrosine adducts for 13 peptides were synthesized. The diethoxyphosphorylated (OP) peptides cross-linked to four different carrier proteins were used to immunize, boost, and screen mice. Monoclonal antibodies were produced with hybridoma technology. Monoclonal antibody depY was purified and characterized by ELISA, western blotting, Biacore, and Octet technology to determine binding affinity and binding specificity. DepY recognized diethoxyphosphotyrosine independent of the amino acid sequence around the modified tyrosine and independent of the identity of the carrier protein or peptide. It had an IC50 of 3 × 10-9 M in a competition assay with OP tubulin. Kd values measured by Biacore and OctetRED96 were 10-8 M for OP-peptides and 1 × 10-12 M for OP-proteins. The limit of detection measured on western blots hybridized with 0.14 µg/mL of depY was 0.025 µg of human albumin conjugated to YGGFL-OP. DepY was specific for diethoxyphosphotyrosine (chlorpyrifos oxon adduct) as it failed to recognize diethoxyphospholysine, phosphoserine, phosphotyrosine, phosphothreonine, dimethoxyphosphotyrosine (dichlorvos adduct), dimethoxyphosphoserine, monomethoxyphosphotyrosine (aged dichlorvos adduct), and cresylphosphoserine. In conclusion, a monoclonal antibody that specifically recognizes diethoxyphosphotyrosine adducts has been developed. The depY monoclonal antibody could be useful for identifying new biomarkers of OP exposure.


Subject(s)
Amino Acids/chemistry , Antibodies, Monoclonal/immunology , Peptides/chemistry , Peptides/immunology , Phosphotyrosine/analogs & derivatives , Phosphotyrosine/immunology , Amino Acids/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Carrier Proteins/chemistry , Carrier Proteins/immunology , Humans , Mice , Molecular Structure , Phosphotyrosine/chemistry
3.
Chem Res Toxicol ; 30(10): 1897-1910, 2017 10 16.
Article in English | MEDLINE | ID: mdl-28892361

ABSTRACT

Nerve agents and organophosphorus pesticides make a covalent bond with the active site serine of acetylcholinesterase (AChE), resulting in inhibition of AChE activity and toxic symptoms. AChE in red blood cells (RBCs) serves as a surrogate for AChE in the nervous system. Mass spectrometry analysis of adducts on RBC AChE could provide evidence of exposure. Our goal was to develop a method of immunopurifying human RBC AChE in quantities adequate for detecting exposure by mass spectrometry. For this purpose, we immobilized 3 commercially available anti-human acetylcholinesterase monoclonal antibodies (AE-1, AE-2, and HR2) plus 3 new monoclonal antibodies. The monoclonal antibodies were characterized for binding affinity, epitope mapping by pairing analysis, and nucleotide and amino acid sequences. AChE was solubilized from frozen RBCs with 1% (v/v) Triton X-100. A 16 mL sample containing 5.8 µg of RBC AChE was treated with a quantity of soman model compound that inhibited 50% of the AChE activity. Native and soman-inhibited RBC AChE samples were immunopurified on antibody-Sepharose beads. The immunopurified RBC AChE was digested with pepsin and analyzed by liquid chromatography tandem mass spectrometry on a 6600 Triple-TOF mass spectrometer. The aged soman-modified PheGlyGluSerAlaGlyAlaAlaSer (FGESAGAAS) peptide was detected using a targeted analysis method. It was concluded that all 6 monoclonal antibodies could be used to immunopurify RBC AChE and that exposure to nerve agents could be detected as adducts on the active site serine of RBC AChE.


Subject(s)
Acetylcholinesterase/isolation & purification , Erythrocytes/enzymology , Immunoprecipitation , Nerve Agents/analysis , Acetylcholinesterase/immunology , Acetylcholinesterase/metabolism , Humans , Mass Spectrometry
4.
Chem Biol Interact ; 266: 17-27, 2017 Mar 25.
Article in English | MEDLINE | ID: mdl-28189703

ABSTRACT

Human butyrylcholinesterase (HuBChE) protects from nerve agent toxicity. Our goal was to determine whether bovine serum could be used as a source of BChE. Bovine BChE (BoBChE) was immunopurified from 100 mL fetal bovine serum (FBS) or 380 mL adult bovine serum by binding to immobilized monoclonal mAb2. Bound proteins were digested with trypsin and analyzed by liquid chromatography-tandem mass spectrometry. The results proved that FBS and adult bovine serum contain BoBChE. The concentration of BoBChE was estimated to be 0.04 µg/mL in FBS, and 0.03 µg/mL in adult bovine serum, values lower than the 4 µg/mL BChE in human serum. Nondenaturing gel electrophoresis showed that monoclonal mAb2 bound BoBChE but not bovine acetylcholinesterase (BoAChE) and confirmed that FBS contains BoBChE and BoAChE. Recombinant bovine BChE (rBoBChE) expressed in serum-free culture medium spontaneously reactivated from inhibition by chlorpyrifos oxon at a rate of 0.0023 min-1 (t1/2 = 301 min-1) and aged at a rate of 0.0138 min-1 (t1/2 = 50 min-1). Both BoBChE and HuBChE have 574 amino acids per subunit and 90% sequence identity. However, the apparent size of serum BoBChE and rBoBChE tetramers was much greater than the 340,000 Da of HuBChE tetramers. Whereas HuBChE tetramers include short polyproline rich peptides derived from lamellipodin, no polyproline peptides have been identified in BoBChE. We hypothesize that BoBChE tetramers use a large polyproline-rich protein to organize subunits into a tetramer and that the low concentration of BoBChE in serum is explained by limited quantities of an unidentified polyproline-rich protein.


Subject(s)
Butyrylcholinesterase/blood , Amino Acid Sequence , Animals , Butyrylcholinesterase/chemistry , Cattle , Chromatography, Liquid , Humans , Kinetics , Mass Spectrometry , Molecular Dynamics Simulation , Sequence Homology, Amino Acid , Tandem Mass Spectrometry
5.
PLoS Pathog ; 7(8): e1002192, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21829370

ABSTRACT

A significant number of environmental microorganisms can cause serious, even fatal, acute and chronic infections in humans. The severity and outcome of each type of infection depends on the expression of specific bacterial phenotypes controlled by complex regulatory networks that sense and respond to the host environment. Although bacterial signals that contribute to a successful acute infection have been identified in a number of pathogens, the signals that mediate the onset and establishment of chronic infections have yet to be discovered. We identified a volatile, low molecular weight molecule, 2-amino acetophenone (2-AA), produced by the opportunistic human pathogen Pseudomonas aeruginosa that reduces bacterial virulence in vivo in flies and in an acute mouse infection model. 2-AA modulates the activity of the virulence regulator MvfR (multiple virulence factor regulator) via a negative feedback loop and it promotes the emergence of P. aeruginosa phenotypes that likely promote chronic lung infections, including accumulation of lasR mutants, long-term survival at stationary phase, and persistence in a Drosophila infection model. We report for the first time the existence of a quorum sensing (QS) regulated volatile molecule that induces bistability phenotype by stochastically silencing acute virulence functions in P. aeruginosa. We propose that 2-AA mediates changes in a subpopulation of cells that facilitate the exploitation of dynamic host environments and promote gene expression changes that favor chronic infections.


Subject(s)
Acetophenones/metabolism , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial/physiology , Pneumonia, Bacterial/metabolism , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/pathogenicity , Quorum Sensing/physiology , Trans-Activators/metabolism , Acute Disease , Animals , Bacterial Proteins/genetics , Chronic Disease , Disease Models, Animal , Drosophila melanogaster , Humans , Mice , Mutation , Pneumonia, Bacterial/genetics , Pseudomonas Infections/genetics , Pseudomonas aeruginosa/genetics , Trans-Activators/genetics
6.
RNA ; 16(11): 2218-25, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20817753

ABSTRACT

RNase P from E. coli will cleave a RNA at a site designated in a complex with an external guide sequence (EGS). The location of the site is determined by the Watson-Crick complementary sequence that can be formed between the RNA and the EGS. Morpholino oligonucleotides (PMOs) that have the same base sequences as any particular EGS will not direct cleavage by RNase P of the target RNA at the expected site in three mRNAs. Instead, cleavage occurs at a secondary site that does not correspond exactly to the expected Watson-Crick sequence in the PMO. This cleavage in the mRNA for a drug resistance gene, CAT mRNA, is at least second order in the concentration of the PMOs, but the mechanism is not understood yet and might be more complicated than a simple second-order reaction. EGSs and PMOs inhibit the reactions of each other effectively in a competitive fashion. A basic peptide attached to the PMO (PPMO) is more effective because of its binding properties to the mRNA as a substrate. However, a PMO is just as efficient as a PPMO on a mRNA that is mutated so that the canonical W-C site has been altered. The altered mRNA is not recognizable by effective extensive W-C pairing to an EGS or PMO. The complex of a PMO on a mutated mRNA as a substrate shows that the dimensions of the modified oligonucleotide cannot be the same as a naked piece of single-stranded RNA.


Subject(s)
Base Pairing , Oligonucleotides/chemistry , RNA, Messenger/chemistry , Kinetics , Mutation , Oligonucleotides/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Substrate Specificity
7.
PLoS Pathog ; 6(3): e1000810, 2010 Mar 12.
Article in English | MEDLINE | ID: mdl-20300606

ABSTRACT

Pathogenic bacteria use interconnected multi-layered regulatory networks, such as quorum sensing (QS) networks to sense and respond to environmental cues and external and internal bacterial cell signals, and thereby adapt to and exploit target hosts. Despite the many advances that have been made in understanding QS regulation, little is known regarding how these inputs are integrated and processed in the context of multi-layered QS regulatory networks. Here we report the examination of the Pseudomonas aeruginosa QS 4-hydroxy-2-alkylquinolines (HAQs) MvfR regulatory network and determination of its interaction with the QS acyl-homoserine-lactone (AHL) RhlR network. The aim of this work was to elucidate paradigmatically the complex relationships between multi-layered regulatory QS circuitries, their signaling molecules, and the environmental cues to which they respond. Our findings revealed positive and negative homeostatic regulatory loops that fine-tune the MvfR regulon via a multi-layered dependent homeostatic regulation of the cell-cell signaling molecules PQS and HHQ, and interplay between these molecules and iron. We discovered that the MvfR regulon component PqsE is a key mediator in orchestrating this homeostatic regulation, and in establishing a connection to the QS rhlR system in cooperation with RhlR. Our results show that P. aeruginosa modulates the intensity of its virulence response, at least in part, through this multi-layered interplay. Our findings underscore the importance of the homeostatic interplay that balances competition within and between QS systems via cell-cell signaling molecules and environmental cues in the control of virulence gene expression. Elucidation of the fine-tuning of this complex relationship offers novel insights into the regulation of these systems and may inform strategies designed to limit infections caused by P. aeruginosa and related human pathogens.


Subject(s)
Homeostasis/physiology , Iron/metabolism , Pseudomonas aeruginosa , Regulon/physiology , Signal Transduction/physiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Diptera , Feedback, Physiological/physiology , Female , Gene Expression Regulation, Bacterial , Genes, Reporter , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/pathogenicity , Quorum Sensing/physiology , Virulence
8.
Proc Natl Acad Sci U S A ; 106(20): 8163-8, 2009 May 19.
Article in English | MEDLINE | ID: mdl-19416872

ABSTRACT

The expression of gene products in bacteria can be inhibited by the use of RNA external guide sequences (EGSs) that hybridize to a target mRNA. Endogenous RNase P cleaves the mRNA in the complex, making it inactive. EGSs participate in this biochemical reaction as the data presented here show. They promote mRNA cleavage at the expected site and sometimes at other secondary sites. Higher-order structure must affect these reactions if the cleavage does not occur at the defined site, which has been determined by techniques based on their ability to find sites that are accessible to the EGS oligonucleotides. Sites defined by a random EGS technique occur as expected. Oligonucleotides made up primarily of defined or random nucleotides are extremely useful in inhibiting expression of the gyrA and rnpA genes from several different bacteria or the cat gene that determines resistance to chloramphenicol in Escherichia coli. An EGS made up of a peptide-phosphorodiamidate morpholino oligonucleotide (PPMO) does not cleave at the same site as an unmodified RNA EGS for reasons that are only partly understood. However, PPMO-EGSs are useful in inhibiting the expression of targeted genes from Gram-negative and Gram-positive organisms during ordinary growth in broth and may provide a basis for broad-spectrum antibiotics.


Subject(s)
RNA, Antisense/genetics , RNA, Bacterial/antagonists & inhibitors , Anti-Bacterial Agents , Binding Sites , Hydrolysis , Nucleic Acid Conformation , Oligonucleotides/chemistry , Oligonucleotides/pharmacology , RNA, Antisense/chemistry , RNA, Antisense/pharmacology , RNA, Bacterial/metabolism , RNA, Small Untranslated
9.
PLoS One ; 3(11): e3719, 2008.
Article in English | MEDLINE | ID: mdl-19005569

ABSTRACT

External guide sequences (EGSs) have successfully been used to inhibit expression of target genes at the post-transcriptional level in both prokaryotes and eukaryotes. We previously reported that EGS accessible and cleavable sites in the target RNAs can rapidly be identified by screening random EGS (rEGS) libraries. Here the method of screening rEGS libraries and a partial RNase T1 digestion assay were used to identify sites accessible to EGSs in the mRNA of a global virulence regulator MglB from Francisella tularensis, a Gram-negative pathogenic bacterium. Specific EGSs were subsequently designed and their activities in terms of the cleavage of mglB mRNA by RNase P were tested in vitro and in vivo. EGS73, EGS148, and EGS155 in both stem and M1 EGS constructs induced mglB mRNA cleavage in vitro. Expression of stem EGS73 and EGS155 in Escherichia coli resulted in significant reduction of the mglB mRNA level coded for the F. tularensis mglB gene inserted in those cells.


Subject(s)
Escherichia coli/genetics , Francisella tularensis/genetics , Gene Expression Regulation, Bacterial , Genes, Bacterial , Virulence Factors/genetics , Base Sequence , Electrophoresis, Polyacrylamide Gel , Molecular Sequence Data , Nucleotide Mapping , RNA, Bacterial/metabolism , RNA, Messenger/metabolism , Ribonuclease P , Virulence Factors/metabolism
10.
Proc Natl Acad Sci U S A ; 105(7): 2354-7, 2008 Feb 19.
Article in English | MEDLINE | ID: mdl-18263737

ABSTRACT

A method of inhibiting the expression of particular genes by using external guide sequences (EGSs) has been improved in its rapidity and specificity. Random EGSs that have 14-nt random sequences are used in the selection procedure for an EGS that attacks the mRNA for a gene in a particular location. A mixture of the random EGSs, the particular target RNA, and RNase P is used in the diagnostic procedure, which, after completion, is analyzed in a gel with suitable control lanes. Within a few hours, the procedure is complete. The action of EGSs designed by an older method is compared with EGSs designed by the random EGS method on mRNAs from two bacterial pathogens.


Subject(s)
Escherichia coli/enzymology , Ribonuclease P/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Databases, Nucleic Acid , RNA, Messenger/genetics , Ribonuclease P/genetics , Sensitivity and Specificity
11.
PLoS Pathog ; 3(3): e35, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17367209

ABSTRACT

There is now substantial evidence that compounds released during host stress directly activate the virulence of certain opportunistic pathogens. Here, we considered that endogenous opioids might function as such compounds, given that they are among the first signals to be released at multiple tissue sites during host stress. We tested the ability of various opioid compounds to enhance the virulence of Pseudomonas aeruginosa using pyocyanin production as a biological readout, and demonstrated enhanced virulence when P. aeruginosa was exposed to synthetic (U-50,488) and endogenous (dynorphin) kappa-agonists. Using various mutants and reporter strains of P. aeruginosa, we identified involvement of key elements of the quorum sensing circuitry such as the global transcriptional regulator MvfR and the quorum sensing-related quinolone signaling molecules PQS, HHQ, and HQNO that respond to kappa-opioids. The in vivo significance of kappa-opioid signaling of P. aeruginosa was demonstrated in mice by showing that dynorphin is released from the intestinal mucosa following ischemia/reperfusion injury, activates quinolone signaling in P. aeruginosa, and enhances the virulence of P. aeruginosa against Lactobacillus spp. and Caenorhabditis elegans. Taken together, these data demonstrate that P. aeruginosa can intercept opioid compounds released during host stress and integrate them into core elements of quorum sensing circuitry leading to enhanced virulence.


Subject(s)
Dynorphins/pharmacology , Pseudomonas aeruginosa/drug effects , Quinolones/metabolism , Quorum Sensing/drug effects , Signal Transduction/drug effects , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Animals , Dose-Response Relationship, Drug , Dynorphins/metabolism , Male , Mice , Mice, Inbred C57BL , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/pathogenicity , Pyocyanine/biosynthesis , Virulence
12.
Mol Microbiol ; 62(6): 1689-99, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17083468

ABSTRACT

MvfR (PqsR), a Pseudomonas aeruginosa LysR-type transcriptional regulator, plays a critical role in the virulence of this pathogen. MvfR modulates the expression of multiple quorum sensing (QS)-regulated virulence factors; and the expression of the phnAB and pqsA-E genes that encode functions mediating 4-hydroxy-2-alkylquinolines (HAQs) signalling compounds biosynthesis, including 3,4-dihydroxy-2heptylquinoline (PQS) and its precursor 4-hydroxy-2-heptylquinoline (HHQ). PQS enhances the in vitro DNA-binding affinity of MvfR to the pqsA-E promoter, to suggest it might function as the in vivo MvfR ligand. Here we identify a novel MvfR ligand, as we show that HHQ binds to the MvfR ligand-binding-domain and potentiates MvfR binding to the pqsA-E promoter leading to transcriptional activation of pqsA-E genes. We show that HHQ is highly produced in vivo, where it is not fully converted into PQS, and demonstrate that it is required for MvfR-dependent gene expression and pathogenicity; PQS is fully dispensable, as pqsH-mutant cells, which produce HHI but completely lack PQS, display normal MvfR-dependent gene expression and virulence. Conversely, PQS is required for full production of pyocyanin. These results uncover a novel biological role for HHQ; and provide novel insights on MvfR activation that may aid in the development of therapies that prevent or treat P. aeruginosa infections in humans.


Subject(s)
Bacterial Proteins/metabolism , Ligands , Pseudomonas aeruginosa/metabolism , Animals , Bacterial Proteins/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Electrophoretic Mobility Shift Assay , Gene Expression Regulation, Bacterial , Mice , Mutation , Promoter Regions, Genetic/genetics , Protein Binding , Protein Conformation , Pseudomonas Infections/microbiology , Pseudomonas Infections/mortality , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/pathogenicity , Quinolines/chemistry , Quinolines/metabolism , Quinolones/chemistry , Quinolones/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Survival Rate , Time Factors , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Virulence/genetics
13.
Microbiology (Reading) ; 152(Pt 6): 1679-1686, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16735731

ABSTRACT

The LysR-type transcriptional regulator MvfR (PqsR) (multiple virulence factor regulator) plays a critical role in Pseudomonas aeruginosa pathogenicity via the transcriptional regulation of multiple quorum-sensing (QS)-regulated virulence factors. LasR activates full mvfR transcription, and MvfR subsequently activates pqsA-E expression. This study identifies and characterizes the key cis-regulatory elements through which mvfR and pqsA-E transcription is regulated in the highly virulent P. aeruginosa strain PA14. Deletion and site-directed mutagenesis indicate that: (1) LasR activates mvfR transcription by binding to a las/rhl box, CTAACAAAAGACATAG, centred at -513 bp upstream of the MvfR translational start site; and (2) RhlR represses pqsA transcription by binding to a las/rhl box, CTGTGAGATTTGGGAG, centred at -311 bp upstream of the pqsA transcriptional initiation site. Furthermore, it is shown that MvfR activates pqsA-E transcription by binding to a LysR box, TTCGGACTCCGAA, centred at -45 bp relative to the pqsA transcriptional initiation site, demonstrating that this LysR box has a critical role in the physical interaction between the MvfR protein and the pqsA promoter. These results provide new insights into the regulatory relationships between LasR and mvfR, and between MvfR/RhlR and the pqs operon, and elucidate further the complex regulation of the P. aeruginosa QS circuitry.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Mutation , Promoter Regions, Genetic , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/physiology , Signal Transduction , Bacterial Proteins/genetics , Base Sequence , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Deletion , Molecular Sequence Data , Mutagenesis, Site-Directed , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/pathogenicity , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic , Virulence Factors/genetics , Virulence Factors/metabolism
14.
Mol Microbiol ; 55(4): 998-1014, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15686549

ABSTRACT

The transcriptional regulator MvfR is required for full Pseudomonas aeruginosa virulence, the function of multiple quorum sensing (QS)-regulated virulence factors and the synthesis of 4-hydroxy-2-alkylquinolines (HAQs), including the Pseudomonas quinolone signal (PQS). Here we investigate the role of MvfR in the QS circuitry and P. aeruginosa pathogenesis. We demonstrate using a combination of biochemical and molecular approaches, including transcription profiling, that MvfR is involved in the regulation of multiple P. aeruginosa QS-controlled genes without altering the expression of lasRI/rhlRI or the production of N-acyl-L-homoserine lactone (AHL) signals. Dissection of how mvfR is interwoven into the P. aeruginosa QS circuitry reveals that the MvfR system, through the essential contribution of PqsE, positively regulates a subset of genes dependant on both LasR and RhlR. Animal studies show that MvfR contributes to P. aeruginosa virulence by controlling the transcription of genes not under RhlR regulation, and that reduced virulence of a mvfR mutant is caused by the loss of pqsE expression and not only a deficiency in HAQs/PQS production. This study provides novel insights into the unique role of the MvfR system in AHL-mediated QS and further supports its importance in P. aeruginosa pathogenesis.


Subject(s)
Bacterial Proteins/metabolism , Homoserine/metabolism , Lactones/metabolism , Pseudomonas aeruginosa/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , DNA Primers , DNA-Binding Proteins , Genes, Bacterial/genetics , Mice , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Pseudomonas aeruginosa/pathogenicity , RNA, Bacterial/genetics , RNA, Bacterial/isolation & purification , Trans-Activators , Transcription, Genetic , Virulence
15.
Biochemistry ; 42(9): 2552-9, 2003 Mar 11.
Article in English | MEDLINE | ID: mdl-12614149

ABSTRACT

DNA binding proteins of two-component signal transduction systems in microorganisms are activated by phosphorylation through an unknown mechanism. NarL is an example from the nitrate/nitrite signal transduction system of Escherichia coli. NarL consists of N- and C-terminal domains, the latter of which contains the DNA binding elements. To explore the mechanism of activation, single nitroxide side chains were introduced, one at a time, at nine different sites throughout the C-terminal domain to monitor the tertiary structure and the status of the surface in contact with the N-terminal domain. In addition, three pairs of doubly labeled proteins were prepared to monitor the interdomain distance using the magnetic dipolar interaction. The results of these site-directed spin-labeling studies reveal that phosphorylation at a distant site in the N-terminal domain triggers domain separation, likely by a hinge-bending motion. This in turn presents key elements of the C-terminal domain for docking to the DNA target in the configuration described in the recent crystal structure. The data also imply that a single conformation of unphosphorylated NarL exists in solution, and there is no detectable equilibrium between the closed and open conformations.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , DNA/chemistry , DNA/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Amino Acid Substitution/genetics , Aspartic Acid/genetics , Crystallization , Cyclic N-Oxides/chemistry , DNA-Binding Proteins/genetics , Electron Spin Resonance Spectroscopy , Escherichia coli Proteins/genetics , Phosphorylation , Protein Binding/genetics , Protein Structure, Secondary/genetics , Protein Structure, Tertiary/genetics , Solutions , Spectrometry, Mass, Electrospray Ionization , Spin Labels , Surface Properties , Thermodynamics
16.
Protein Sci ; 11(10): 2427-36, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12237464

ABSTRACT

The NarL response regulatory protein of Escherichia coli has been engineered by covalent modification with 1,10-phenanthroline (OP) to create a set of site-specific DNA-cleaving agents. This was accomplished by introducing single cysteine amino acid replacements at selected locations within the carboxy-terminal DNA-binding domain in or nearby the helix 8 to helix 9 region of the NarL protein using site-directed mutagenesis. Of 18 modified NarL-OP proteins made, 13 retained the ability to bind DNA as evidenced by gel mobility assays, whereas 10 of the 1,10-phenanthroline-modified proteins also exhibited specific cleavage activity for a synthetic NarL recognition sequence. These DNA-cleaving agents were divided into two groups based on the location of the cleavage sites. The first class set cleaved the DNA nearby the center of a synthetic 7-2-7 sequence composed of two NarL heptamer sites separated by a 2-bp spacer element. The second class cut the DNA at the periphery of the 7-2-7 sequence. The cleavage data are consistent with the ability of two NarL monomers to recognize and bind to the DNA in a head-to-head orientation. A second set of DNA-cleaving agents was constructed using the carboxy-terminal domain of NarL called NarL(C). Similar cleavage patterns were observed whether full-length NarL or NarL(C) was used. The availability of 1,10-phenanthroline-modified NarL and NarL(C) proteins opens up the possibility to explore the position, orientation, and number of NarL recognition sites at E. coli promoters predicted to contain multiple and complex arrangements of NarL-binding sites.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Escherichia coli Proteins/metabolism , Phenanthrolines/metabolism , Base Sequence , Escherichia coli/metabolism , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...