Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 1840, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38418447

ABSTRACT

The cytoskeletal protein actin plays a critical role in the pathogenicity of the intracellular parasite, Toxoplasma gondii, mediating invasion and egress, cargo transport, and organelle inheritance. Advances in live cell imaging have revealed extensive filamentous actin networks in the Apicomplexan parasite, but there are conflicting data regarding the biochemical and biophysical properties of Toxoplasma actin. Here, we imaged the in vitro assembly of individual Toxoplasma actin filaments in real time, showing that native, unstabilized filaments grow tens of microns in length. Unlike skeletal muscle actin, Toxoplasma filaments intrinsically undergo rapid treadmilling due to a high critical concentration, fast monomer dissociation, and rapid nucleotide exchange. Cryo-EM structures of jasplakinolide-stabilized and native (i.e. unstabilized) filaments show an architecture like skeletal actin, with differences in assembly contacts in the D-loop that explain the dynamic nature of the filament, likely a conserved feature of Apicomplexan actin. This work demonstrates that evolutionary changes at assembly interfaces can tune the dynamic properties of actin filaments without disrupting their conserved structure.


Subject(s)
Parasites , Toxoplasma , Animals , Actins/metabolism , Toxoplasma/metabolism , Actin Cytoskeleton/metabolism , Cytoskeleton/metabolism , Parasites/metabolism
2.
bioRxiv ; 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38293063

ABSTRACT

The opportunistic pathogen Pseudomonas aeruginosa infects cystic fibrosis (CF) patient airways and produces a virulence factor Cif that is associated with worse outcomes. Cif is an epoxide hydrolase that reduces cell-surface abundance of the cystic fibrosis transmembrane conductance regulator (CFTR) and sabotages pro-resolving signals. Its expression is regulated by a divergently transcribed TetR family transcriptional repressor. CifR represents the first reported epoxide-sensing bacterial transcriptional regulator, but neither its interaction with cognate operator sequences nor the mechanism of activation has been investigated. Using biochemical and structural approaches, we uncovered the molecular mechanisms controlling this complex virulence operon. We present here the first molecular structures of CifR alone and in complex with operator DNA, resolved in a single crystal lattice. Significant conformational changes between these two structures suggest how CifR regulates the expression of the virulence gene cif. Interactions between the N-terminal extension of CifR with the DNA minor groove of the operator play a significant role in the operator recognition of CifR. We also determined that cysteine residue Cys107 is critical for epoxide sensing and DNA release. These results offer new insights into the stereochemical regulation of an epoxide-based virulence circuit in a critically important clinical pathogen.

3.
bioRxiv ; 2023 Aug 30.
Article in English | MEDLINE | ID: mdl-37693530

ABSTRACT

The cytoskeletal protein actin plays a critical role in the pathogenicity of Toxoplasma gondii, mediating invasion and egress, cargo transport, and organelle inheritance. Advances in live cell imaging have revealed extensive filamentous actin networks in the Apicomplexan parasite, but there is conflicting data regarding the biochemical and biophysical properties of Toxoplasma actin. Here, we imaged the in vitro assembly of individual Toxoplasma actin filaments in real time, showing that native, unstabilized filaments grow tens of microns in length. Unlike skeletal muscle actin, Toxoplasma filaments intrinsically undergo rapid treadmilling due to a high critical concentration, fast monomer dissociation, and rapid nucleotide exchange. Cryo-EM structures of stabilized and unstabilized filaments show an architecture like skeletal actin, with differences in assembly contacts in the D-loop that explain the dynamic nature of the filament, likely a conserved feature of Apicomplexan actin. This work demonstrates that evolutionary changes at assembly interfaces can tune dynamic properties of actin filaments without disrupting their conserved structure.

4.
Nat Struct Mol Biol ; 30(3): 391-402, 2023 03.
Article in English | MEDLINE | ID: mdl-36747094

ABSTRACT

The universally conserved enzyme phosphoribosyl pyrophosphate synthetase (PRPS) assembles filaments in evolutionarily diverse organisms. PRPS is a key regulator of nucleotide metabolism, and mutations in the human enzyme PRPS1 lead to a spectrum of diseases. Here we determine structures of human PRPS1 filaments in active and inhibited states, with fixed assembly contacts accommodating both conformations. The conserved assembly interface stabilizes the binding site for the essential activator phosphate, increasing activity in the filament. Some disease mutations alter assembly, supporting the link between filament stability and activity. Structures of active PRPS1 filaments turning over substrate also reveal coupling of catalysis in one active site with product release in an adjacent site. PRPS1 filaments therefore provide an additional layer of allosteric control, conserved throughout evolution, with likely impact on metabolic homeostasis. Stabilization of allosteric binding sites by polymerization adds to the growing diversity of assembly-based enzyme regulatory mechanisms.


Subject(s)
Ribose-Phosphate Pyrophosphokinase , Humans , Allosteric Site , Mutation , Ribose-Phosphate Pyrophosphokinase/genetics , Ribose-Phosphate Pyrophosphokinase/metabolism
5.
Curr Opin Struct Biol ; 79: 102530, 2023 04.
Article in English | MEDLINE | ID: mdl-36709625

ABSTRACT

Recent work in structural biology is shedding light on how many of the enzymes of intermediary metabolism are self- and co-assembling into large, filamentous polymers or agglomerates to organize and regulate the complex and essential biochemical pathways in cells. Filament assembly provides an additional layer of regulation by modulating the intrinsic allostery of the enzyme protomers which tunes activity in response to a variety of environmental cues. Enzyme filaments dynamically assemble and disassemble in response to changes in metabolite levels and environmental cues, shifting metabolic flux on a more rapid timescale than transcriptional or translational reprogramming. Here we present recent examples of high-resolution structures of filaments from proteins in intermediary metabolism and we discuss how filament assembly modulates the activities of these and other proteins.


Subject(s)
Cytoskeleton , Proteins
6.
PLoS Pathog ; 18(4): e1010496, 2022 04.
Article in English | MEDLINE | ID: mdl-35482847

ABSTRACT

Attachment to the intestinal epithelium is critical to the lifestyle of the ubiquitous parasite Giardia lamblia. The ventrolateral flange is a sheet-like membrane protrusion at the interface between parasites and attached surfaces. This structure has been implicated in attachment, but its role has been poorly defined. Here, we identified a novel actin associated protein with putative WH2-like actin binding domains we named Flangin. Flangin complexes with Giardia actin (GlActin) and is enriched in the ventrolateral flange making it a valuable marker for studying the flanges' role in Giardia biology. Live imaging revealed that the flange grows to around 1 µm in width after cytokinesis, then remains uniform in size during interphase, grows in mitosis, and is resorbed during cytokinesis. A flangin truncation mutant stabilizes the flange and blocks cytokinesis, indicating that flange disassembly is necessary for rapid myosin-independent cytokinesis in Giardia. Rho family GTPases are important regulators of membrane protrusions and GlRac, the sole Rho family GTPase in Giardia, was localized to the flange. Knockdown of Flangin, GlActin, and GlRac result in flange formation defects. This indicates a conserved role for GlRac and GlActin in forming membrane protrusions, despite the absence of canonical actin binding proteins that link Rho GTPase signaling to lamellipodia formation. Flangin-depleted parasites had reduced surface contact and when challenged with fluid shear force in flow chambers they had a reduced ability to remain attached, confirming a role for the flange in attachment. This secondary attachment mechanism complements the microtubule based adhesive ventral disc, a feature that may be particularly important during mitosis when the parental ventral disc disassembles in preparation for cytokinesis. This work supports the emerging view that Giardia's unconventional actin cytoskeleton has an important role in supporting parasite attachment.


Subject(s)
Giardia lamblia , Giardiasis , Parasites , Actins/metabolism , Animals , Giardia/metabolism , Giardia lamblia/genetics , Giardia lamblia/metabolism , Giardiasis/parasitology , Parasites/metabolism , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
7.
Curr Res Struct Biol ; 3: 72-84, 2021.
Article in English | MEDLINE | ID: mdl-34235487

ABSTRACT

Epoxide hydrolases catalyze the conversion of epoxides to vicinal diols in a range of cellular processes such as signaling, detoxification, and virulence. These enzymes typically utilize a pair of tyrosine residues to orient the substrate epoxide ring in the active site and stabilize the hydrolysis intermediate. A new subclass of epoxide hydrolases that utilize a histidine in place of one of the tyrosines was established with the discovery of the CFTR Inhibitory Factor (Cif) from Pseudomonas aeruginosa. Although the presence of such Cif-like epoxide hydrolases was predicted in other opportunistic pathogens based on sequence analyses, only Cif and its homolog aCif from Acinetobacter nosocomialis have been characterized. Here we report the biochemical and structural characteristics of Cfl1 and Cfl2, two Cif-like epoxide hydrolases from Burkholderia cenocepacia. Cfl1 is able to hydrolyze xenobiotic as well as biological epoxides that might be encountered in the environment or during infection. In contrast, Cfl2 shows very low activity against a diverse set of epoxides. The crystal structures of the two proteins reveal quaternary structures that build on the well-known dimeric assembly of the α/ß hydrolase domain, but broaden our understanding of the structural diversity encoded in novel oligomer interfaces. Analysis of the interfaces reveals both similarities and key differences in sequence conservation between the two assemblies, and between the canonical dimer and the novel oligomer interfaces of each assembly. Finally, we discuss the effects of these higher-order assemblies on the intra-monomer flexibility of Cfl1 and Cfl2 and their possible roles in regulating enzymatic activity.

8.
J Biol Chem ; 297(2): 100981, 2021 08.
Article in English | MEDLINE | ID: mdl-34302812

ABSTRACT

Gram-positive bacteria contain sortase enzymes on their cell surfaces that catalyze transpeptidation reactions critical for proper cellular function. In vitro, sortases are used in sortase-mediated ligation (SML) reactions for a variety of protein engineering applications. Historically, sortase A from Staphylococcus aureus (saSrtA) has been the enzyme of choice to catalyze SML reactions. However, the stringent specificity of saSrtA for the LPXTG sequence motif limits its uses. Here, we describe the impact on substrate selectivity of a structurally conserved loop with a high degree of sequence variability in all classes of sortases. We investigate the contribution of this ß7-ß8 loop by designing and testing chimeric sortase enzymes. Our chimeras utilize natural sequence variation of class A sortases from eight species engineered into the SrtA sequence from Streptococcus pneumoniae. While some of these chimeric enzymes mimic the activity and selectivity of the WT protein from which the loop sequence was derived (e.g., that of saSrtA), others results in chimeric Streptococcus pneumoniae SrtA enzymes that are able to accommodate a range of residues in the final position of the substrate motif (LPXTX). Using mutagenesis, structural comparisons, and sequence analyses, we identify three interactions facilitated by ß7-ß8 loop residues that appear to be broadly conserved or converged upon in class A sortase enzymes. These studies provide the foundation for a deeper understanding of sortase target selectivity and can expand the sortase toolbox for future SML applications.


Subject(s)
Aminoacyltransferases/chemistry , Bacterial Proteins/chemistry , Catalytic Domain , Cysteine Endopeptidases/chemistry , Mutation , Protein Engineering/methods , Staphylococcal Infections/enzymology , Staphylococcus aureus/enzymology , Amino Acid Sequence , Aminoacyltransferases/genetics , Aminoacyltransferases/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Crystallography, X-Ray , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Models, Molecular , Mutagenesis, Site-Directed , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/chemistry , Staphylococcus aureus/isolation & purification , Substrate Specificity
9.
Nat Microbiol ; 5(8): 1011-1015, 2020 08.
Article in English | MEDLINE | ID: mdl-32424337

ABSTRACT

Host-virus interactions structure microbial communities, drive biogeochemical cycles and enhance genetic diversity in nature1,2. Hypotheses proposed to explain the range of interactions that mediate these processes often invoke lysogeny3-6, a latent infection strategy used by temperate bacterial viruses to replicate in host cells until an induction event triggers the production and lytic release of free viruses. Most cultured bacteria harbour temperate viruses in their genomes (prophage)7. The absence of prophages in cultures of the dominant lineages of marine bacteria has contributed to an ongoing debate over the ecological significance of lysogeny and other viral life strategies in nature6,8-15. Here, we report the discovery of prophages in cultured SAR11, the ocean's most abundant clade of heterotrophic bacteria16,17. We show the concurrent production of cells and viruses, with enhanced virus production under carbon-limiting growth conditions. Evidence that related prophages are broadly distributed in the oceans suggests that similar interactions have contributed to the evolutionary success of SAR11 in nutrient-limited systems.


Subject(s)
Alphaproteobacteria/virology , Bacteriophages/metabolism , Genome, Viral , Host Microbial Interactions , Lysogeny , Prophages/metabolism , Viral Proteins/genetics , Heterotrophic Processes , Metagenomics , Microbiota , Oceans and Seas , Seawater/microbiology , Seawater/virology , Sequence Alignment , Sequence Analysis, DNA , Viral Proteins/metabolism
10.
Anal Chim Acta ; 1057: 106-113, 2019 May 30.
Article in English | MEDLINE | ID: mdl-30832908

ABSTRACT

Lead identification and optimization are essential steps in the development of a new drug. It requires cost-effective, selective and sensitive chemical tools. Here, we report a novel method using nanobodies that allows the efficient screening for potent ligands. The method is illustrated with the cystic fibrosis transmembrane conductance regulator inhibitory factor (Cif), a virulence factor secreted by the opportunistic pathogen Pseudomonas aeruginosa. 18 nanobodies selective to Cif were isolated by bio-panning from nanobody-phage library constructed from immunized llama. 8 out of 18 nanobodies were identified as potent inhibitors of Cif enzymatic activity with IC50s in the range of 0.3-6.4 µM. A nanobody VHH219 showed high affinity (KD = 0.08 nM) to Cif and the highest inhibitory potency, IC50 = 0.3 µM. A displacement sandwich ELISA (dsELISA) with VHH219 was then developed for classification of synthetic small molecule inhibitors according their inhibitory potency. The developed assay allowed identification of new inhibitor with highest potency reported so far (0.16 ±â€¯0.02 µM). The results from dsELISA assay correlates strongly with a conventional fluorogenic assay (R = 0.9998) in predicting the inhibitory potency of the tested compounds. However, the novel dsELISA is an order of magnitude more sensitive and allows the identification and ranking of potent inhibitors missed by the classic fluorogenic assay method. These data were supported with Octet biolayer interferometry measurements. The novel method described herein relies solely on the binding properties of the specific neutralizing nanobody, and thus is applicable to any pharmacological target for which such a nanobody can be found, independent of any requirement for catalytic activity.


Subject(s)
Bacterial Proteins/immunology , Single-Domain Antibodies/immunology , Virulence Factors/immunology , Amino Acid Sequence , Animals , Camelids, New World , Catalytic Domain , Immunization , Inhibitory Concentration 50 , Single-Domain Antibodies/chemistry
11.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L150-L156, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28982736

ABSTRACT

The opportunistic pathogen Pseudomonas aeruginosa colonizes the lungs of susceptible individuals by deploying virulence factors targeting host defenses. The secreted factor Cif (cystic fibrosis transmembrane conductance regulator inhibitory factor) dysregulates the endocytic recycling of CFTR and thus reduces CFTR abundance in host epithelial membranes. We have postulated that the decrease in ion secretion mediated by Cif would slow mucociliary transport and decrease bacterial clearance from the lungs. To test this hypothesis, we explored the effects of Cif in cultured epithelia and in the lungs of mice. We developed a strategy to interpret the "hurricane-like" motions observed in reconstituted cultures and identified a Cif-mediated decrease in the velocity of mucus transport in vitro. Presence of Cif also increased the number of bacteria recovered at two time points in an acute mouse model of pneumonia caused by P. aeruginosa. Furthermore, recent work has demonstrated an inverse correlation between the airway concentrations of Cif and 15-epi-lipoxin A4, a proresolving lipid mediator important in host defense and the resolution of pathogen-initiated inflammation. Here, we observe elevated levels of 15-epi-lipoxin A4 in the lungs of mice infected with a strain of P. aeruginosa that expresses only an inactive form of cif compared with those mice infected with wild-type P. aeruginosa. Together these data support the inclusion of Cif on the list of virulence factors that assist P. aeruginosa in colonizing and damaging the airways of compromised patients. Furthermore, this study establishes techniques that enable our groups to explore the underlying mechanisms of Cif effects during respiratory infection.


Subject(s)
Bacterial Proteins/metabolism , Bronchi/pathology , Epithelial Cells/pathology , Pneumonia/etiology , Pseudomonas Infections/complications , Pseudomonas aeruginosa/pathogenicity , Virulence Factors/metabolism , Animals , Biological Transport , Bronchi/enzymology , Bronchi/microbiology , Cells, Cultured , Disease Models, Animal , Epithelial Cells/enzymology , Epithelial Cells/microbiology , Humans , Lipoxins/metabolism , Male , Mice , Mice, Inbred C57BL , Mucociliary Clearance , Pneumonia/metabolism , Pneumonia/pathology , Pseudomonas Infections/microbiology
12.
Structure ; 25(5): 697-707.e4, 2017 05 02.
Article in English | MEDLINE | ID: mdl-28392259

ABSTRACT

Pseudomonas aeruginosa secretes an epoxide hydrolase with catalytic activity that triggers degradation of the cystic fibrosis transmembrane conductance regulator (CFTR) and perturbs other host defense networks. Targets of this CFTR inhibitory factor (Cif) are largely unknown, but include an epoxy-fatty acid. In this class of signaling molecules, chirality can be an important determinant of physiological output and potency. Here we explore the active-site chemistry of this two-step α/ß-hydrolase and its implications for an emerging class of virulence enzymes. In combination with hydrolysis data, crystal structures of 15 trapped hydroxyalkyl-enzyme intermediates reveal the stereochemical basis of Cif's substrate specificity, as well as its regioisomeric and enantiomeric preferences. The structures also reveal distinct sets of conformational changes that enable the active site to expand dramatically in two directions, accommodating a surprising array of potential physiological epoxide targets. These new substrates may contribute to Cif's diverse effects in vivo, and thus to the success of P. aeruginosa and other pathogens during infection.


Subject(s)
Epoxide Hydrolases/chemistry , Virulence Factors/chemistry , Binding Sites , Crystallography, X-Ray , Epoxide Hydrolases/metabolism , Molecular Dynamics Simulation , Protein Binding , Pseudomonas aeruginosa/enzymology , Substrate Specificity , Virulence Factors/metabolism
13.
Proc Natl Acad Sci U S A ; 114(1): 136-141, 2017 01 03.
Article in English | MEDLINE | ID: mdl-27980032

ABSTRACT

Recurrent Pseudomonas aeruginosa infections coupled with robust, damaging neutrophilic inflammation characterize the chronic lung disease cystic fibrosis (CF). The proresolving lipid mediator, 15-epi lipoxin A4 (15-epi LXA4), plays a critical role in limiting neutrophil activation and tissue inflammation, thus promoting the return to tissue homeostasis. Here, we show that a secreted P. aeruginosa epoxide hydrolase, cystic fibrosis transmembrane conductance regulator inhibitory factor (Cif), can disrupt 15-epi LXA4 transcellular biosynthesis and function. In the airway, 15-epi LXA4 production is stimulated by the epithelial-derived eicosanoid 14,15-epoxyeicosatrienoic acid (14,15-EET). Cif sabotages the production of 15-epi LXA4 by rapidly hydrolyzing 14,15-EET into its cognate diol, eliminating a proresolving signal that potently suppresses IL-8-driven neutrophil transepithelial migration in vitro. Retrospective analyses of samples from patients with CF supported the translational relevance of these preclinical findings. Elevated levels of Cif in bronchoalveolar lavage fluid were correlated with lower levels of 15-epi LXA4, increased IL-8 concentrations, and impaired lung function. Together, these findings provide structural, biochemical, and immunological evidence that the bacterial epoxide hydrolase Cif disrupts resolution pathways during bacterial lung infections. The data also suggest that Cif contributes to sustained pulmonary inflammation and associated loss of lung function in patients with CF.


Subject(s)
8,11,14-Eicosatrienoic Acid/analogs & derivatives , Bacterial Proteins/metabolism , Lipoxins/metabolism , Neutrophil Activation/immunology , Neutrophils/immunology , Pseudomonas aeruginosa/metabolism , Virulence Factors/metabolism , 8,11,14-Eicosatrienoic Acid/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Cell Line , Crystallography, X-Ray , Cystic Fibrosis/microbiology , Cystic Fibrosis/pathology , Humans , Inflammation/chemically induced , Lung Diseases/microbiology , Lung Diseases/pathology , Pseudomonas Infections/microbiology , Pseudomonas Infections/pathology , Pseudomonas aeruginosa/pathogenicity , Retrospective Studies
14.
J Med Chem ; 59(10): 4790-9, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27120257

ABSTRACT

The virulence factor cystic fibrosis transmembrane conductance regulator (CFTR) inhibitory factor (Cif) is secreted by Pseudomonas aeruginosa and is the founding member of a distinct class of epoxide hydrolases (EHs) that triggers the catalysis-dependent degradation of the CFTR. We describe here the development of a series of potent and selective Cif inhibitors by structure-based drug design. Initial screening revealed 1a (KB2115), a thyroid hormone analog, as a lead compound with low micromolar potency. Structural requirements for potency were systematically probed, and interactions between Cif and 1a were characterized by X-ray crystallography. On the basis of these data, new compounds were designed to yield additional hydrogen bonding with residues of the Cif active site. From this effort, three compounds were identified that are 10-fold more potent toward Cif than our first-generation inhibitors and have no detectable thyroid hormone-like activity. These inhibitors will be useful tools to study the pathological role of Cif and have the potential for clinical application.


Subject(s)
Drug Design , Epoxide Hydrolases/antagonists & inhibitors , Pseudomonas aeruginosa/enzymology , Triiodothyronine/analogs & derivatives , Virulence Factors/antagonists & inhibitors , Crystallography, X-Ray , Dose-Response Relationship, Drug , Epoxide Hydrolases/metabolism , Models, Molecular , Structure-Activity Relationship , Triiodothyronine/chemical synthesis , Triiodothyronine/chemistry , Triiodothyronine/pharmacology , Virulence Factors/metabolism
15.
Biochemistry ; 55(5): 788-97, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26752215

ABSTRACT

The CFTR inhibitory factor (Cif) is an epoxide hydrolase (EH) virulence factor secreted by the bacterium Pseudomonas aeruginosa. Sequence alignments reveal a pattern of Cif-like substitutions that proved to be characteristic of a new subfamily of bacterial EHs. At the same time, crystallographic and mutagenetic data suggest that EH activity is required for virulence and that Cif's active site remains generally compatible with a canonical two-step EH mechanism. A hallmark of this mechanism is the formation of a covalent hydroxyalkyl-enzyme intermediate by nucleophilic attack. In several well-studied EHs, this intermediate has been captured at near stoichiometric levels, presumably reflecting rate-limiting hydrolysis. Here we show by mass spectrometry that only minimal levels of the expected intermediate can be trapped with WT Cif. In contrast, substantial amounts of intermediate are recovered from an active-site mutant (Cif-E153Q) that selectively targets the second, hydrolytic release step. Utilizing Cif-E153Q and a previously reported nucleophile mutant (Cif-D129S), we then captured Cif in the substrate-bound, hydroxyalkyl-intermediate, and product-bound states for 1,2-epoxyhexane, yielding the first crystallographic snapshots of an EH at these key stages along the reaction coordinate. Taken together, our data illuminate the proposed two-step hydrolytic mechanism of a new class of bacterial virulence factor. They also suggest that the failure of WT Cif to accumulate a covalent hydroxyalkyl-enzyme intermediate reflects an active-site chemistry in which hydrolysis is no longer the rate-limiting step, a noncanonical kinetic regime that may explain similar observations with a number of other EHs.


Subject(s)
Bacterial Proteins/metabolism , Epoxy Compounds/metabolism , Pseudomonas aeruginosa/metabolism , Virulence Factors/metabolism , Bacterial Proteins/chemistry , Catalysis , Crystallography, X-Ray , Hydrolysis , Models, Molecular , Protein Conformation , Virulence Factors/chemistry
16.
Angew Chem Int Ed Engl ; 54(34): 9881-5, 2015 Aug 17.
Article in English | MEDLINE | ID: mdl-26136396

ABSTRACT

Opportunistic pathogens exploit diverse strategies to sabotage host defenses. Pseudomonas aeruginosa secretes the CFTR inhibitory factor Cif and thus triggers loss of CFTR, an ion channel required for airway mucociliary defense. However, the mechanism of action of Cif has remained unclear. It catalyzes epoxide hydrolysis, but there is no known role for natural epoxides in CFTR regulation. It was demonstrated that the hydrolase activity of Cif is strictly required for its effects on CFTR. A small-molecule inhibitor that protects this key component of the mucociliary defense system was also uncovered. These results provide a basis for targeting the distinctive virulence chemistry of Cif and suggest an unanticipated role of physiological epoxides in intracellular protein trafficking.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Enzyme Inhibitors/pharmacology , Epoxide Hydrolases/antagonists & inhibitors , Pseudomonas aeruginosa/chemistry , Small Molecule Libraries/pharmacology , Virulence Factors/antagonists & inhibitors , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Epoxide Hydrolases/metabolism , Models, Molecular , Molecular Structure , Pseudomonas aeruginosa/metabolism , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Virulence Factors/metabolism
17.
J Biol Chem ; 289(11): 7460-9, 2014 Mar 14.
Article in English | MEDLINE | ID: mdl-24474692

ABSTRACT

Endocytic recycling of the cystic fibrosis transmembrane conductance regulator (CFTR) is blocked by the CFTR inhibitory factor (Cif). Originally discovered in Pseudomonas aeruginosa, Cif is a secreted epoxide hydrolase that is transcriptionally regulated by CifR, an epoxide-sensitive repressor. In this report, we investigate a homologous protein found in strains of the emerging nosocomial pathogens Acinetobacter nosocomialis and Acinetobacter baumannii ("aCif"). Like Cif, aCif is an epoxide hydrolase that carries an N-terminal secretion signal and can be purified from culture supernatants. When applied directly to polarized airway epithelial cells, mature aCif triggers a reduction in CFTR abundance at the apical membrane. Biochemical and crystallographic studies reveal a dimeric assembly with a stereochemically conserved active site, confirming our motif-based identification of candidate Cif-like pathogenic EH sequences. Furthermore, cif expression is transcriptionally repressed by a CifR homolog ("aCifR") and is induced in the presence of epoxides. Overall, this Acinetobacter protein recapitulates the essential attributes of the Pseudomonas Cif system and thus may facilitate airway colonization in nosocomial lung infections.


Subject(s)
Acinetobacter baumannii/metabolism , Acinetobacter/enzymology , Bacterial Proteins/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epoxide Hydrolases/metabolism , Acinetobacter/genetics , Amino Acid Motifs , Amino Acid Sequence , Bacterial Proteins/genetics , Crystallography, X-Ray , Endocytosis , Epoxide Hydrolases/genetics , Gene Deletion , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Plasmids/metabolism , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Substrate Specificity , Transcription, Genetic , Virulence Factors/genetics , Virulence Factors/metabolism
18.
J Bacteriol ; 193(18): 4685-98, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21764921

ABSTRACT

Cyclic di-GMP (c-di-GMP) is a broadly conserved, intracellular second-messenger molecule that regulates biofilm formation by many bacteria. The synthesis of c-di-GMP is catalyzed by diguanylate cyclases (DGCs) containing the GGDEF domain, while its degradation is achieved through the phosphodiesterase activities of EAL and HD-GYP domains. c-di-GMP controls biofilm formation by Pseudomonas fluorescens Pf0-1 by promoting the cell surface localization of a large adhesive protein, LapA. LapA localization is regulated posttranslationally by a c-di-GMP effector system consisting of LapD and LapG, which senses cytoplasmic c-di-GMP and modifies the LapA protein in the outer membrane. Despite the apparent requirement for c-di-GMP for biofilm formation by P. fluorescens Pf0-1, no DGCs from this strain have been characterized to date. In this study, we undertook a systematic mutagenesis of 30 predicted DGCs and found that mutations in just 4 cause reductions in biofilm formation by P. fluorescens Pf0-1 under the conditions tested. These DGCs were characterized genetically and biochemically to corroborate the hypothesis that they function to produce c-di-GMP in vivo. The effects of DGC gene mutations on phenotypes associated with biofilm formation were analyzed. One DGC preferentially affects LapA localization, another DGC mainly controls swimming motility, while a third DGC affects both LapA and motility. Our data support the conclusion that different c-di-GMP-regulated outputs can be specifically controlled by distinct DGCs.


Subject(s)
Biofilms/growth & development , Escherichia coli Proteins/metabolism , Phosphorus-Oxygen Lyases/metabolism , Pseudomonas fluorescens/enzymology , Pseudomonas fluorescens/physiology , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , DNA Transposable Elements , Escherichia coli Proteins/genetics , Gene Knockout Techniques , Mutagenesis, Insertional , Phosphorus-Oxygen Lyases/genetics , Pseudomonas fluorescens/growth & development , Pseudomonas fluorescens/metabolism
19.
Yeast ; 27(10): 861-5, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20641023

ABSTRACT

Progress in modern genetics is greatly facilitated by systematic resources that enable rapid and comprehensive analysis. Here we report the creation of a nearly complete systematic low-copy (CEN URA3) library of the Saccharomyces cerevisiae genome that complements existing systematic high-copy libraries.


Subject(s)
Centromere/genetics , Gene Library , Genome, Fungal , Saccharomyces cerevisiae/genetics , Classification , Genes, Fungal
SELECTION OF CITATIONS
SEARCH DETAIL
...