Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.051
Filter
Add more filters

Publication year range
1.
Cell ; 186(23): 5098-5113.e19, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37918395

ABSTRACT

Drug-resistant Pseudomonas aeruginosa (PA) poses an emerging threat to human health with urgent need for alternative therapeutic approaches. Here, we deciphered the B cell and antibody response to the virulence-associated type III secretion system (T3SS) in a cohort of patients chronically infected with PA. Single-cell analytics revealed a diverse B cell receptor repertoire directed against the T3SS needle-tip protein PcrV, enabling the production of monoclonal antibodies (mAbs) abrogating T3SS-mediated cytotoxicity. Mechanistic studies involving cryoelectron microscopy identified a surface-exposed C-terminal PcrV epitope as the target of highly neutralizing mAbs with broad activity against drug-resistant PA isolates. These anti-PcrV mAbs were as effective as treatment with conventional antibiotics in vivo. Our study reveals that chronically infected patients represent a source of neutralizing antibodies, which can be exploited as therapeutics against PA.


Subject(s)
Antibodies, Bacterial , Antibodies, Neutralizing , Pseudomonas Infections , Pseudomonas aeruginosa , Humans , Antibodies, Bacterial/pharmacology , Cryoelectron Microscopy , Immunoglobulins/metabolism , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Pseudomonas Infections/drug therapy
2.
Cell ; 186(4): 864-876.e21, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36750095

ABSTRACT

A fundamental strategy of eukaryotic antiviral immunity involves the cGAS enzyme, which synthesizes 2',3'-cGAMP and activates the effector STING. Diverse bacteria contain cGAS-like enzymes that produce cyclic oligonucleotides and induce anti-phage activity, known as CBASS. However, this activity has only been demonstrated through heterologous expression. Whether bacteria harboring CBASS antagonize and co-evolve with phages is unknown. Here, we identified an endogenous cGAS-like enzyme in Pseudomonas aeruginosa that generates 3',3'-cGAMP during phage infection, signals to a phospholipase effector, and limits phage replication. In response, phages express an anti-CBASS protein ("Acb2") that forms a hexamer with three 3',3'-cGAMP molecules and reduces phospholipase activity. Acb2 also binds to molecules produced by other bacterial cGAS-like enzymes (3',3'-cUU/UA/UG/AA) and mammalian cGAS (2',3'-cGAMP), suggesting broad inhibition of cGAS-based immunity. Upon Acb2 deletion, CBASS blocks lytic phage replication and lysogenic induction, but rare phages evade CBASS through major capsid gene mutations. Altogether, we demonstrate endogenous CBASS anti-phage function and strategies of CBASS inhibition and evasion.


Subject(s)
Bacteria , Bacteriophages , Animals , Bacteria/immunology , Bacteria/virology , Bacteriophages/physiology , Immunity , Nucleotidyltransferases/metabolism
3.
Cell ; 182(4): 919-932.e19, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32763156

ABSTRACT

Redox cycling of extracellular electron shuttles can enable the metabolic activity of subpopulations within multicellular bacterial biofilms that lack direct access to electron acceptors or donors. How these shuttles catalyze extracellular electron transfer (EET) within biofilms without being lost to the environment has been a long-standing question. Here, we show that phenazines mediate efficient EET through interactions with extracellular DNA (eDNA) in Pseudomonas aeruginosa biofilms. Retention of pyocyanin (PYO) and phenazine carboxamide in the biofilm matrix is facilitated by eDNA binding. In vitro, different phenazines can exchange electrons in the presence or absence of DNA and can participate directly in redox reactions through DNA. In vivo, biofilm eDNA can also support rapid electron transfer between redox active intercalators. Together, these results establish that PYO:eDNA interactions support an efficient redox cycle with rapid EET that is faster than the rate of PYO loss from the biofilm.


Subject(s)
Biofilms/growth & development , DNA/chemistry , Pseudomonas aeruginosa/physiology , Pyocyanine/chemistry , DNA/metabolism , Electrochemical Techniques , Electrodes , Electron Transport/drug effects , Fluorescent Dyes/chemistry , Hydrogen-Ion Concentration , Oxidation-Reduction , Phenazines/chemistry , Phenazines/metabolism , Phenazines/pharmacology , Pyocyanine/metabolism
4.
Cell ; 178(6): 1452-1464.e13, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31474367

ABSTRACT

Phages express anti-CRISPR (Acr) proteins to inhibit CRISPR-Cas systems that would otherwise destroy their genomes. Most acr genes are located adjacent to anti-CRISPR-associated (aca) genes, which encode proteins with a helix-turn-helix DNA-binding motif. The conservation of aca genes has served as a signpost for the identification of acr genes, but the function of the proteins encoded by these genes has not been investigated. Here we reveal that an acr-associated promoter drives high levels of acr transcription immediately after phage DNA injection and that Aca proteins subsequently repress this transcription. Without Aca activity, this strong transcription is lethal to a phage. Our results demonstrate how sufficient levels of Acr proteins accumulate early in the infection process to inhibit existing CRISPR-Cas complexes in the host cell. They also imply that the conserved role of Aca proteins is to mitigate the deleterious effects of strong constitutive transcription from acr promoters.


Subject(s)
Bacteriophages/genetics , CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Viral Proteins/genetics , CRISPR-Associated Proteins/genetics , Escherichia coli/virology , Promoter Regions, Genetic/genetics , Pseudomonas aeruginosa/virology , Transcription Factors/genetics , Transcription, Genetic
5.
Immunity ; 57(10): 2280-2295.e6, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39299238

ABSTRACT

Toll/interleukin-1/resistance (TIR)-domain proteins with enzymatic activity are essential for immunity in plants, animals, and bacteria. However, it is not known how these proteins function in pathogen sensing in animals. We discovered that the lone enzymatic TIR-domain protein in the nematode C. elegans (TIR-1, homolog of mammalian sterile alpha and TIR motif-containing 1 [SARM1]) was strategically expressed on the membranes of a specific intracellular compartment called lysosome-related organelles. The positioning of TIR-1 on lysosome-related organelles enables intestinal epithelial cells in the nematode C. elegans to survey for pathogen effector-triggered host damage. A virulence effector secreted by the bacterial pathogen Pseudomonas aeruginosa alkalinized and condensed lysosome-related organelles. This pathogen-induced morphological change in lysosome-related organelles triggered TIR-1 multimerization, which engaged its intrinsic NAD+ hydrolase (NADase) activity to activate the p38 innate immune pathway and protect the host against microbial intoxication. Thus, TIR-1 is a guard protein in an effector-triggered immune response, which enables intestinal epithelial cells to survey for pathogen-induced host damage.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Immunity, Innate , Lysosomes , Pseudomonas aeruginosa , Animals , Caenorhabditis elegans/immunology , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/immunology , Caenorhabditis elegans Proteins/genetics , Pseudomonas aeruginosa/immunology , Lysosomes/metabolism , Lysosomes/immunology , Immunity, Innate/immunology , Intestines/immunology , Pseudomonas Infections/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Host-Pathogen Interactions/immunology , p38 Mitogen-Activated Protein Kinases/metabolism , Receptors, G-Protein-Coupled
6.
Immunity ; 57(4): 859-875.e11, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38513665

ABSTRACT

At mucosal surfaces, epithelial cells provide a structural barrier and an immune defense system. However, dysregulated epithelial responses can contribute to disease states. Here, we demonstrated that epithelial cell-intrinsic production of interleukin-23 (IL-23) triggers an inflammatory loop in the prevalent oral disease periodontitis. Epithelial IL-23 expression localized to areas proximal to the disease-associated microbiome and was evident in experimental models and patients with common and genetic forms of disease. Mechanistically, flagellated microbial species of the periodontitis microbiome triggered epithelial IL-23 induction in a TLR5 receptor-dependent manner. Therefore, unlike other Th17-driven diseases, non-hematopoietic-cell-derived IL-23 served as an initiator of pathogenic inflammation in periodontitis. Beyond periodontitis, analysis of publicly available datasets revealed the expression of epithelial IL-23 in settings of infection, malignancy, and autoimmunity, suggesting a broader role for epithelial-intrinsic IL-23 in human disease. Collectively, this work highlights an important role for the barrier epithelium in the induction of IL-23-mediated inflammation.


Subject(s)
Interleukin-23 , Periodontitis , Humans , Epithelial Cells , Inflammation , Toll-Like Receptor 5/metabolism
7.
Cell ; 174(4): 917-925.e10, 2018 08 09.
Article in English | MEDLINE | ID: mdl-30033364

ABSTRACT

Bacteria utilize CRISPR-Cas adaptive immune systems for protection from bacteriophages (phages), and some phages produce anti-CRISPR (Acr) proteins that inhibit immune function. Despite thorough mechanistic and structural information for some Acr proteins, how they are deployed and utilized by a phage during infection is unknown. Here, we show that Acr production does not guarantee phage replication when faced with CRISPR-Cas immunity, but instead, infections fail when phage population numbers fall below a critical threshold. Infections succeed only if a sufficient Acr dose is contributed to a single cell by multiple phage genomes. The production of Acr proteins by phage genomes that fail to replicate leave the cell immunosuppressed, which predisposes the cell for successful infection by other phages in the population. This altruistic mechanism for CRISPR-Cas inhibition demonstrates inter-virus cooperation that may also manifest in other host-parasite interactions.


Subject(s)
Bacteriophages/immunology , CRISPR-Cas Systems/immunology , Host-Pathogen Interactions/immunology , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/virology , Viral Proteins/immunology , Evolution, Molecular , Pseudomonas aeruginosa/genetics , Viral Proteins/metabolism
8.
Cell ; 172(1-2): 121-134.e14, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29307490

ABSTRACT

Chronic Pseudomonas aeruginosa infections evade antibiotic therapy and are associated with mortality in cystic fibrosis (CF) patients. We find that in vitro resistance evolution of P. aeruginosa toward clinically relevant antibiotics leads to phenotypic convergence toward distinct states. These states are associated with collateral sensitivity toward several antibiotic classes and encoded by mutations in antibiotic resistance genes, including transcriptional regulator nfxB. Longitudinal analysis of isolates from CF patients reveals similar and defined phenotypic states, which are associated with extinction of specific sub-lineages in patients. In-depth investigation of chronic P. aeruginosa populations in a CF patient during antibiotic therapy revealed dramatic genotypic and phenotypic convergence. Notably, fluoroquinolone-resistant subpopulations harboring nfxB mutations were eradicated by antibiotic therapy as predicted by our in vitro data. This study supports the hypothesis that antibiotic treatment of chronic infections can be optimized by targeting phenotypic states associated with specific mutations to improve treatment success in chronic infections.


Subject(s)
Cystic Fibrosis/microbiology , Drug Resistance, Bacterial , Evolution, Molecular , Phenotype , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/genetics , Cystic Fibrosis/complications , DNA-Binding Proteins/genetics , Humans , Male , Middle Aged , Mutation , Pseudomonas Infections/complications , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/pathogenicity , Selection, Genetic , Transcription Factors/genetics
9.
Immunity ; 56(4): 768-782.e9, 2023 04 11.
Article in English | MEDLINE | ID: mdl-36804958

ABSTRACT

Distinguishing infectious pathogens from harmless microorganisms is essential for animal health. The mechanisms used to identify infectious microbes are not fully understood, particularly in metazoan hosts that eat bacteria as their food source. Here, we characterized a non-canonical pattern-recognition system in Caenorhabditis elegans (C. elegans) that assesses the relative threat of virulent Pseudomonas aeruginosa (P. aeruginosa) to activate innate immunity. We discovered that the innate immune response in C. elegans was triggered by phenazine-1-carboxamide (PCN), a toxic metabolite produced by pathogenic strains of P. aeruginosa. We identified the nuclear hormone receptor NHR-86/HNF4 as the PCN sensor in C. elegans and validated that PCN bound to the ligand-binding domain of NHR-86/HNF4. Activation of NHR-86/HNF4 by PCN directly engaged a transcriptional program in intestinal epithelial cells that protected against P. aeruginosa. Thus, a bacterial metabolite is a pattern of pathogenesis surveilled by nematodes to identify a pathogen in its bacterial diet.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Gene Expression Regulation , Receptors, Cytoplasmic and Nuclear/metabolism , Immunity, Innate , Bacteria , Pseudomonas aeruginosa/metabolism
10.
Mol Cell ; 82(18): 3484-3498.e11, 2022 09 15.
Article in English | MEDLINE | ID: mdl-36070765

ABSTRACT

ADP-ribosyltransferases (ARTs) were among the first identified bacterial virulence factors. Canonical ART toxins are delivered into host cells where they modify essential proteins, thereby inactivating cellular processes and promoting pathogenesis. Our understanding of ARTs has since expanded beyond protein-targeting toxins to include antibiotic inactivation and DNA damage repair. Here, we report the discovery of RhsP2 as an ART toxin delivered between competing bacteria by a type VI secretion system of Pseudomonas aeruginosa. A structure of RhsP2 reveals that it resembles protein-targeting ARTs such as diphtheria toxin. Remarkably, however, RhsP2 ADP-ribosylates 2'-hydroxyl groups of double-stranded RNA, and thus, its activity is highly promiscuous with identified cellular targets including the tRNA pool and the RNA-processing ribozyme, ribonuclease P. Consequently, cell death arises from the inhibition of translation and disruption of tRNA processing. Overall, our data demonstrate a previously undescribed mechanism of bacterial antagonism and uncover an unprecedented activity catalyzed by ART enzymes.


Subject(s)
RNA, Catalytic , Type VI Secretion Systems , ADP Ribose Transferases/chemistry , Adenosine Diphosphate/metabolism , Anti-Bacterial Agents/metabolism , Bacteria/genetics , Diphtheria Toxin/genetics , Diphtheria Toxin/metabolism , RNA, Catalytic/genetics , RNA, Catalytic/metabolism , RNA, Double-Stranded/metabolism , Ribonuclease P/genetics , Type VI Secretion Systems/metabolism , Virulence Factors/metabolism
11.
Mol Cell ; 81(3): 571-583.e6, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33412111

ABSTRACT

The arms race between bacteria and phages has led to the evolution of diverse anti-phage defenses, several of which are controlled by quorum-sensing pathways. In this work, we characterize a quorum-sensing anti-activator protein, Aqs1, found in Pseudomonas phage DMS3. We show that Aqs1 inhibits LasR, the master regulator of quorum sensing, and present the crystal structure of the Aqs1-LasR complex. The 69-residue Aqs1 protein also inhibits PilB, the type IV pilus assembly ATPase protein, which blocks superinfection by phages that require the pilus for infection. This study highlights the remarkable ability of small phage proteins to bind multiple host proteins and disrupt key biological pathways. As quorum sensing influences various anti-phage defenses, Aqs1 provides a mechanism by which infecting phages might simultaneously dampen multiple defenses. Because quorum-sensing systems are broadly distributed across bacteria, this mechanism of phage counter-defense may play an important role in phage-host evolutionary dynamics.


Subject(s)
Bacterial Proteins/metabolism , Bacteriophages/metabolism , Pseudomonas aeruginosa/metabolism , Quorum Sensing , Trans-Activators/metabolism , Viral Proteins/metabolism , Bacterial Proteins/genetics , Bacteriophages/genetics , Bacteriophages/pathogenicity , Fimbriae, Bacterial/metabolism , Host-Pathogen Interactions , Oxidoreductases/genetics , Oxidoreductases/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/growth & development , Pyocyanine/metabolism , Trans-Activators/genetics , Viral Proteins/genetics
12.
EMBO J ; 43(19): 4384-4405, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39143239

ABSTRACT

Bacteriophages are the most abundant biological entities on Earth, but our understanding of many aspects of their lifecycles is still incomplete. Here, we have structurally analysed the infection cycle of the siphophage Casadabanvirus JBD30. Using its baseplate, JBD30 attaches to Pseudomonas aeruginosa via the bacterial type IV pilus, whose subsequent retraction brings the phage to the bacterial cell surface. Cryo-electron microscopy structures of the baseplate-pilus complex show that the tripod of baseplate receptor-binding proteins attaches to the outer bacterial membrane. The tripod and baseplate then open to release three copies of the tape-measure protein, an event that is followed by DNA ejection. JBD30 major capsid proteins assemble into procapsids, which expand by 7% in diameter upon filling with phage dsDNA. The DNA-filled heads are finally joined with 180-nm-long tails, which bend easily because flexible loops mediate contacts between the successive discs of major tail proteins. It is likely that the structural features and replication mechanisms described here are conserved among siphophages that utilize the type IV pili for initial cell attachment.


Subject(s)
Cryoelectron Microscopy , Pseudomonas Phages , Pseudomonas aeruginosa , Virus Replication , Pseudomonas aeruginosa/virology , Pseudomonas aeruginosa/metabolism , Pseudomonas Phages/ultrastructure , Pseudomonas Phages/genetics , Pseudomonas Phages/metabolism , Pseudomonas Phages/physiology , Fimbriae, Bacterial/metabolism , Fimbriae, Bacterial/ultrastructure , Fimbriae, Bacterial/virology , Capsid Proteins/metabolism , Capsid Proteins/chemistry , Capsid Proteins/genetics , DNA, Viral/metabolism , DNA, Viral/genetics , Siphoviridae/genetics , Siphoviridae/ultrastructure , Siphoviridae/physiology , Siphoviridae/metabolism
13.
EMBO J ; 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39322758

ABSTRACT

Toxin-antitoxin (TA) systems are widespread in bacteria and implicated in genome stability, virulence, phage defense, and persistence. TA systems have diverse activities and cellular targets, but their physiological roles and regulatory mechanisms are often unclear. Here, we show that the NatR-NatT TA system, which is part of the core genome of the human pathogen Pseudomonas aeruginosa, generates drug-tolerant persisters by specifically depleting nicotinamide dinucleotides. While actively growing P. aeruginosa cells compensate for NatT-mediated NAD+ deficiency by inducing the NAD+ salvage pathway, NAD depletion generates drug-tolerant persisters under nutrient-limited conditions. Our structural and biochemical analyses propose a model for NatT toxin activation and autoregulation and indicate that NatT activity is subject to powerful metabolic feedback control by the NAD+ precursor nicotinamide. Based on the identification of natT gain-of-function alleles in patient isolates and on the observation that NatT increases P. aeruginosa virulence, we postulate that NatT modulates pathogen fitness during infections. These findings pave the way for detailed investigations into how a toxin-antitoxin system can promote pathogen persistence by disrupting essential metabolic pathways.

14.
EMBO J ; 43(8): 1634-1652, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38467832

ABSTRACT

During bacterial cell growth, hydrolases cleave peptide cross-links between strands of the peptidoglycan sacculus to allow new strand insertion. The Pseudomonas aeruginosa carboxyl-terminal processing protease (CTP) CtpA regulates some of these hydrolases by degrading them. CtpA assembles as an inactive hexamer composed of a trimer-of-dimers, but its lipoprotein binding partner LbcA activates CtpA by an unknown mechanism. Here, we report the cryo-EM structures of the CtpA-LbcA complex. LbcA has an N-terminal adaptor domain that binds to CtpA, and a C-terminal superhelical tetratricopeptide repeat domain. One LbcA molecule attaches to each of the three vertices of a CtpA hexamer. LbcA triggers relocation of the CtpA PDZ domain, remodeling of the substrate binding pocket, and realignment of the catalytic residues. Surprisingly, only one CtpA molecule in a CtpA dimer is activated upon LbcA binding. Also, a long loop from one CtpA dimer inserts into a neighboring dimer to facilitate the proteolytic activity. This work has revealed an activation mechanism for a bacterial CTP that is strikingly different from other CTPs that have been characterized structurally.


Subject(s)
Endopeptidases , Pseudomonas aeruginosa , Pseudomonas aeruginosa/metabolism , Endopeptidases/metabolism , Proteolysis
15.
Annu Rev Microbiol ; 76: 413-433, 2022 09 08.
Article in English | MEDLINE | ID: mdl-35655342

ABSTRACT

Microbial communities enmeshed in a matrix of macromolecules, termed as biofilms, are the natural setting of bacteria. Exopolysaccharide is a critical matrix component of biofilms. Here, we focus on biofilm matrix exopolysaccharides in Pseudomonas aeruginosa. This opportunistic pathogen can adapt to a wide range of environments and can form biofilms or aggregates in a variety of surfaces or environments, such as the lungs of people with cystic fibrosis, catheters, wounds, and contact lenses. The ability to synthesize multiple exopolysaccharides is one of the advantages that facilitate bacterial survival in different environments. P. aeruginosa can produce several exopolysaccharides, including alginate, Psl, Pel, and lipopolysaccharide. In this review, we highlight the roles of each exopolysaccharide in P. aeruginosa biofilm development and how bacteria coordinate the biosynthesis of multiple exopolysaccharides and bacterial motility. In addition, we present advances in antibiofilm strategies targeting matrix exopolysaccharides, with a focus on glycoside hydrolases.


Subject(s)
Polysaccharides, Bacterial , Pseudomonas aeruginosa , Biofilms , Humans , Pseudomonas aeruginosa/metabolism
16.
Proc Natl Acad Sci U S A ; 121(32): e2304382121, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39088389

ABSTRACT

Microbes rarely exist in isolation and instead form complex polymicrobial communities. As a result, microbes have developed intricate offensive and defensive strategies that enhance their fitness in these complex communities. Thus, identifying and understanding the molecular mechanisms controlling polymicrobial interactions is critical for understanding the function of microbial communities. In this study, we show that the gram-negative opportunistic human pathogen Pseudomonas aeruginosa, which frequently causes infection alongside a plethora of other microbes including fungi, encodes a genetic network which can detect and defend against gliotoxin, a potent, disulfide-containing antimicrobial produced by the ubiquitous filamentous fungus Aspergillus fumigatus. We show that gliotoxin exposure disrupts P. aeruginosa zinc homeostasis, leading to transcriptional activation of a gene encoding a previously uncharacterized dithiol oxidase (herein named as DnoP), which detoxifies gliotoxin and structurally related toxins. Despite sharing little homology to the A. fumigatus gliotoxin resistance protein (GliT), the enzymatic mechanism of DnoP from P. aeruginosa appears to be identical that used by A. fumigatus. Thus, DnoP and its transcriptional induction by low zinc represent a rare example of both convergent evolution of toxin defense and environmental cue sensing across kingdoms. Collectively, these data provide compelling evidence that P. aeruginosa has evolved to survive exposure to an A. fumigatus disulfide-containing toxin in the natural environment.


Subject(s)
Aspergillus fumigatus , Gliotoxin , Pseudomonas aeruginosa , Gliotoxin/metabolism , Pseudomonas aeruginosa/metabolism , Pseudomonas aeruginosa/genetics , Aspergillus fumigatus/metabolism , Aspergillus fumigatus/genetics , Zinc/metabolism , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Microbial Interactions , Humans , Fungal Proteins/metabolism , Fungal Proteins/genetics
17.
Proc Natl Acad Sci U S A ; 121(2): e2312334121, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38170744

ABSTRACT

Bacterial infections are often polymicrobial. Pseudomonas aeruginosa and Staphylococcus aureus cause chronic co-infections, which are more problematic than mono-species infections. Understanding the mechanisms of their interactions is crucial for treating co-infections. Staphyloxanthin (STX), a yellow pigment synthesized by the S. aureus crt operon, promotes S. aureus resistance to oxidative stress and neutrophil-mediated killing. We found that STX production by S. aureus, either as surface-grown macrocolonies or planktonic cultures, was elevated when exposed to the P. aeruginosa exoproduct, 2-heptyl-4-hydroxyquinoline N-oxide (HQNO). This was observed with both mucoid and non-mucoid P. aeruginosa strains. The induction phenotype was found in a majority of P. aeruginosa and S. aureus clinical isolates examined. When subjected to hydrogen peroxide or human neutrophils, P. aeruginosa survival was significantly higher when mixed with wild-type (WT) S. aureus, compared to P. aeruginosa alone or with an S. aureus crt mutant deficient in STX production. In a murine wound model, co-infection with WT S. aureus, but not the STX-deficient mutant, enhanced P. aeruginosa burden and disease compared to mono-infection. In conclusion, we identified a role for P. aeruginosa HQNO mediating polymicrobial interactions with S. aureus by inducing STX production, which consequently promotes resistance to the innate immune effectors H2O2 and neutrophils. These results further our understanding of how different bacterial species cooperatively cause co-infections.


Subject(s)
Coinfection , Staphylococcal Infections , Humans , Animals , Mice , Staphylococcus aureus/genetics , Hydrogen Peroxide/pharmacology , Neutrophils , Staphylococcal Infections/microbiology , Pseudomonas aeruginosa/genetics , Biological Factors , Biofilms
18.
Proc Natl Acad Sci U S A ; 121(33): e2406234121, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39102545

ABSTRACT

Laboratory models are central to microbiology research, advancing the understanding of bacterial physiology by mimicking natural environments, from soil to the human microbiome. When studying host-bacteria interactions, animal models enable investigators to examine bacterial dynamics associated with a host, and in the case of human infections, animal models are necessary to translate basic research into clinical treatments. Efforts toward improving animal infection models are typically based on reproducing host genotypes/phenotypes and disease manifestations, leaving a gap in how well the physiology of microbes reflects their behavior in a human host. Understanding bacterial physiology is vital because it dictates host response and bacterial interactions with antimicrobials. Thus, our goal was to develop an animal model that accurately recapitulates bacterial physiology in human infection. The system we chose to model was a chronic Pseudomonas aeruginosa respiratory infection in cystic fibrosis (CF). To accomplish this goal, we leveraged a framework that we recently developed to evaluate model accuracy by calculating the percentage of bacterial genes that are expressed similarly in a model to how they are expressed in their infection environment. We combined two complementary models of P. aeruginosa infection-an in vitro synthetic CF sputum model (SCFM2) and a mouse acute pneumonia model. This combined model captured the chronic physiology of P. aeruginosa in CF better than the standard mouse infection model, showing the power of a data-driven approach to refining animal models. In addition, the results of this work challenge the assumption that a chronic infection model requires long-term colonization.


Subject(s)
Cystic Fibrosis , Disease Models, Animal , Pseudomonas Infections , Pseudomonas aeruginosa , Cystic Fibrosis/microbiology , Cystic Fibrosis/complications , Pseudomonas aeruginosa/physiology , Pseudomonas aeruginosa/pathogenicity , Animals , Pseudomonas Infections/microbiology , Mice , Humans , Respiratory Tract Infections/microbiology , Host-Pathogen Interactions , Sputum/microbiology
19.
Brief Bioinform ; 25(5)2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39133098

ABSTRACT

Pseudomonas aeruginosa is a complex nosocomial infectious agent responsible for numerous illnesses, with its growing resistance variations complicating treatment development. Studies have emphasized the importance of virulence factors OprE and OprF in pathogenesis, highlighting their potential as vaccine candidates. In this study, B-cell, MHC-I, and MHC-II epitopes were identified, and molecular linkers were active to join these epitopes with an appropriate adjuvant to construct a vaccine. Computational tools were employed to forecast the tertiary framework, characteristics, and also to confirm the vaccine's composition. The potency was weighed through population coverage analysis and immune simulation. This project aims to create a multi-epitope vaccine to reduce P. aeruginosa-related illness and mortality using immunoinformatics resources. The ultimate complex has been determined to be stable, soluble, antigenic, and non-allergenic upon inspection of its physicochemical and immunological properties. Additionally, the protein exhibited acidic and hydrophilic characteristics. The Ramachandran plot, ProSA-web, ERRAT, and Verify3D were employed to ensure the final model's authenticity once the protein's three-dimensional structure had been established and refined. The vaccine model showed a significant binding score and stability when interacting with MHC receptors. Population coverage analysis indicated a global coverage rate of 83.40%, with the USA having the highest coverage rate, exceeding 90%. Moreover, the vaccine sequence underwent codon optimization before being cloned into the Escherichia coli plasmid vector pET-28a (+) at the EcoRI and EcoRV restriction sites. Our research has developed a vaccine against P. aeruginosa that has strong binding affinity and worldwide coverage, offering an acceptable way to mitigate nosocomial infections.


Subject(s)
Computational Biology , Pseudomonas Infections , Pseudomonas aeruginosa , Sepsis , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/genetics , Humans , Pseudomonas Infections/prevention & control , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Sepsis/prevention & control , Sepsis/immunology , Sepsis/microbiology , Computational Biology/methods , Epitopes/immunology , Epitopes/chemistry , Pneumonia/prevention & control , Pneumonia/immunology , Pneumonia/microbiology , Pseudomonas Vaccines/immunology , Bacterial Vaccines/immunology , Bacterial Proteins/immunology , Bacterial Proteins/genetics
20.
Immunity ; 47(6): 1169-1181.e7, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29246444

ABSTRACT

The tumor suppressor PTEN controls cell proliferation by regulating phosphatidylinositol-3-kinase (PI3K) activity, but the participation of PTEN in host defense against bacterial infection is less well understood. Anti-inflammatory PI3K-Akt signaling is suppressed in patients with cystic fibrosis (CF), a disease characterized by hyper-inflammatory responses to airway infection. We found that Ptenl-/- mice, which lack the NH2-amino terminal splice variant of PTEN, were unable to eradicate Pseudomonas aeruginosa from the airways and could not generate sufficient anti-inflammatory PI3K activity, similar to what is observed in CF. PTEN and the CF transmembrane conductance regulator (CFTR) interacted directly and this interaction was necessary to position PTEN at the membrane. CF patients under corrector-potentiator therapy, which enhances CFTR transport to the membrane, have increased PTEN amounts. These findings suggest that improved CFTR trafficking could enhance P. aeruginosa clearance from the CF airway by activating PTEN-mediated anti-bacterial responses and might represent a therapeutic strategy.


Subject(s)
Cell Membrane/immunology , Cystic Fibrosis Transmembrane Conductance Regulator/immunology , Cystic Fibrosis/immunology , PTEN Phosphohydrolase/immunology , Pseudomonas Infections/immunology , Aminophenols/pharmacology , Aminopyridines/pharmacology , Animals , Benzodioxoles/pharmacology , Cell Membrane/drug effects , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Cystic Fibrosis/microbiology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Gene Expression Regulation , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Monocytes/drug effects , Monocytes/immunology , Monocytes/microbiology , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/immunology , Protein Binding , Protein Conformation , Protein Transport , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Pseudomonas Infections/genetics , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/immunology , Quinolones/pharmacology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL