Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.022
Filter
1.
Cell Rep ; 43(9): 114737, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39277862

ABSTRACT

Itaconate serves as an immune-specific metabolite that regulates gene transcription and metabolism in both host and pathogens. S-itaconation is a post-translational modification that regulates immune response; however, its antimicrobial mechanism under the physiological condition remains unclear. Here, we apply a bioorthogonal itaconate probe to perform global profiling of S-itaconation in living pathogens, including S. Typhimurium, S. aureus, and P. aeruginosa. Some functional enzymes are covalently modified by itaconate, including those involved in the de novo purine biosynthesis pathway. Further biochemical studies demonstrate that itaconate suppresses this specific pathway to limit Salmonella growth by inhibiting the initiator purF to lower de novo purine biosynthesis and simultaneously targeting the guaABC cluster to block the salvage route. Our chemoproteomic study provides a global portrait of S-itaconation in multiple pathogens and offers a valuable resource for finding susceptible targets to combat drug-resistant pathogens in the future.


Subject(s)
Proteomics , Purines , Succinates , Succinates/pharmacology , Succinates/metabolism , Purines/biosynthesis , Purines/pharmacology , Proteomics/methods , Staphylococcus aureus/drug effects , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/metabolism , Salmonella typhimurium/drug effects , Salmonella typhimurium/metabolism , Bacterial Proteins/metabolism
2.
Virulence ; 15(1): 2399792, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39239914

ABSTRACT

Human CD81 and CD9 are members of the tetraspanin family of proteins characterized by a canonical structure of four transmembrane domains and two extracellular loop domains. Tetraspanins are known as molecular facilitators, which assemble and organize cell surface receptors and partner molecules forming clusters known as tetraspanin-enriched microdomains. They have been implicated to play various biological roles including an involvement in infections with microbial pathogens. Here, we demonstrate an important role of CD81 for the invasion of epithelial cells by Salmonella enterica. We show that the overexpression of CD81 in HepG2 cells enhances invasion of various typhoidal and non-typhoidal Salmonella serovars. Deletion of CD81 by CRISPR/Cas9 in intestinal epithelial cells (C2BBe1 and HT29-MTX-E12) reduces S. Typhimurium invasion. In addition, the effect of human CD81 is species-specific as only human but not rat CD81 facilitates Salmonella invasion. Finally, immunofluorescence microscopy and proximity ligation assay revealed that both human tetraspanins CD81 and CD9 are recruited to the entry site of S. Typhimurium during invasion but not during adhesion to the host cell surface. Overall, we demonstrate that the human tetraspanin CD81 facilitates Salmonella invasion into epithelial host cells.


Subject(s)
Epithelial Cells , Salmonella enterica , Tetraspanin 28 , Tetraspanin 29 , Humans , Tetraspanin 28/metabolism , Tetraspanin 28/genetics , Epithelial Cells/microbiology , Tetraspanin 29/metabolism , Tetraspanin 29/genetics , Animals , Salmonella enterica/genetics , Salmonella enterica/physiology , Salmonella typhimurium/genetics , Salmonella typhimurium/pathogenicity , Salmonella typhimurium/metabolism , Salmonella typhimurium/physiology , Hep G2 Cells , Rats , Salmonella Infections/microbiology , HT29 Cells
3.
Anal Chim Acta ; 1320: 343002, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39142781

ABSTRACT

BACKGROUND: Rapid and sensitive detection of foodborne pathogens in food plays a crucial role in controlling outbreaks of foodborne diseases, of which Listeria monocytogenes and Salmonella typhimurium are representative and notable pathogens. Thus, it's of great importance to achieve the effective detection of these pathogens. However, the most common detection methods (culture-based technique, Polymerase Chain Reaction and immunological methods) have disadvantages that cannot be ignored, such as time-consuming, laborious, complex sample preparation process, and the possibility of cross-reaction. Hence, it is essential to develop a facile detection method for the pathogens with high sensitivity and specificity to avoid the above-mentioned disadvantages. RESULTS: We report a label-free visual platform for the simultaneous capture and detection of Listeria monocytogenes and Salmonella typhimurium. For the first time, we have prepared polydimethylsiloxane-Chromotrope 2R membrane which serves as the substrate for bacterial capture and enrichment through the formation of specific recognition sites. The positively charged Pt-covalent organic framework combines with the pathogens through surface charge interaction, thereby the label-free sandwich platform is formed. Remarkable peroxidase activity of Pt-covalent organic framework converts the conversion of bacterial quantity into amplified color signal by catalyzing 3,3',5,5'-Tetramethylbenzidine to oxidized 3,3',5,5'-Tetramethylbenzidine. The platform demonstrates the capability to identify two representative food-borne pathogens within a time frame of 100 min, exhibiting high sensitivity and excellent specificity without the interference from non-target bacteria. The limit of detection of the visual platform toward Listeria monocytogenes and Salmonella typhimurium was 1.61 CFU mL-1 and 1.31 CFU mL-1, respectively. And the limit of quantification toward Listeria monocytogenes and Salmonella typhimurium was 4.94 CFU mL-1 and 2.47 CFU mL-1, respectively. The relative standard derivations of the visual platform for both bacteria were lower than 4.9 %. Furthermore, our proposed platform has obtained reliable and satisfactory results on analyzing diverse food samples. SIGNIFICANCE: This research expands the application of a label-free platform combined with unlabeled nanocomponents in the rapid isolation and detection of diverse of food-borne pathogens. The platform possesses the advantages of simple operation and real-time monitoring, without complicated sample pretreatment process. The whole detection process can realize the simultaneous monitoring of Listeria monocytogenes and Salmonella typhimurium within 100 min. Furthermore, it is also of reference significance for the detection of other common pathogens.


Subject(s)
Food Microbiology , Listeria monocytogenes , Metal-Organic Frameworks , Molecular Imprinting , Salmonella typhimurium , Molecular Imprinting/methods , Metal-Organic Frameworks/chemistry , Food Microbiology/methods , Listeria monocytogenes/chemistry , Listeria monocytogenes/isolation & purification , Listeria monocytogenes/metabolism , Salmonella typhimurium/chemistry , Salmonella typhimurium/isolation & purification , Salmonella typhimurium/metabolism , Benzidines/chemistry , Benzidines/metabolism , Platinum/chemistry
4.
Nat Commun ; 15(1): 6958, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138228

ABSTRACT

Outer membrane vesicles (OMVs) produced by Gram-negative bacteria have key roles in cell envelope homeostasis, secretion, interbacterial communication, and pathogenesis. The facultative intracellular pathogen Salmonella Typhimurium increases OMV production inside the acidic vacuoles of host cells by changing expression of its outer membrane proteins and modifying the composition of lipid A. However, the molecular mechanisms that translate pH changes into OMV production are not completely understood. Here, we show that the outer membrane protein PagC promotes OMV production through pH-dependent interactions between its extracellular loops and surrounding lipopolysaccharide (LPS). Structural comparisons and mutational studies indicate that a pH-responsive amino acid motif in PagC extracellular loops, containing PagC-specific histidine residues, is crucial for OMV formation. Molecular dynamics simulations suggest that protonation of histidine residues leads to changes in the structure and flexibility of PagC extracellular loops and their interactions with the surrounding LPS, altering membrane curvature. Consistent with that hypothesis, mimicking acidic pH by mutating those histidine residues to lysine increases OMV production. Thus, our findings reveal a mechanism for sensing and responding to environmental pH and for control of membrane dynamics by outer membrane proteins.


Subject(s)
Bacterial Outer Membrane Proteins , Lipopolysaccharides , Molecular Dynamics Simulation , Salmonella typhimurium , Hydrogen-Ion Concentration , Bacterial Outer Membrane Proteins/metabolism , Bacterial Outer Membrane Proteins/genetics , Salmonella typhimurium/metabolism , Salmonella typhimurium/genetics , Lipopolysaccharides/metabolism , Bacterial Outer Membrane/metabolism , Amino Acid Motifs , Histidine/metabolism
5.
PLoS Biol ; 22(8): e3002731, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39102375

ABSTRACT

Bacterial pathogens utilize the factors of their hosts to infect them, but which factors they exploit remain poorly defined. Here, we show that a pathogenic Salmonella enterica serovar Typhimurium (STm) exploits host polyamines for the functional expression of virulence factors. An STm mutant strain lacking principal genes required for polyamine synthesis and transport exhibited impaired infectivity in mice. A polyamine uptake-impaired strain of STm was unable to inject effectors of the type 3 secretion system into host cells due to a failure of needle assembly. STm infection stimulated host polyamine production by increasing arginase expression. The decline in polyamine levels caused by difluoromethylornithine, which inhibits host polyamine production, attenuated STm colonization, whereas polyamine supplementation augmented STm pathogenesis. Our work reveals that host polyamines are a key factor promoting STm infection, and therefore a promising therapeutic target for bacterial infection.


Subject(s)
Polyamines , Salmonella typhimurium , Type III Secretion Systems , Virulence Factors , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , Salmonella typhimurium/genetics , Animals , Polyamines/metabolism , Mice , Type III Secretion Systems/metabolism , Type III Secretion Systems/genetics , Virulence Factors/metabolism , Virulence Factors/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Host-Pathogen Interactions , Humans , Salmonella Infections/metabolism , Salmonella Infections/microbiology , Female
6.
mSphere ; 9(8): e0036724, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39109886

ABSTRACT

Protein production strategies in bacteria are often limited due to the need for cell lysis and complicated purification schemes. To avoid these challenges, researchers have developed bacterial strains capable of secreting heterologous protein products outside the cell, but secretion titers often remain too low for commercial applicability. Improved understanding of the link between secretion system structure and its secretory abilities can help overcome the barrier to engineering higher secretion titers. Here, we investigated this link with the PrgI protein, the monomer of the secretory channel of the type 3 secretion system (T3SS) of Salmonella enterica. Despite detailed knowledge of the PrgI needle's assembly and structure, little is known about how its structure influences its secretory capabilities. To study this, we recently constructed a comprehensive codon mutagenesis library of the PrgI protein utilizing a novel one-pot recombineering approach. We then screened this library for functional T3SS assembly and secretion titer by measuring the secretion of alkaline phosphatase using a high-throughput activity assay. This allowed us to construct a first-of-its-kind secretion fitness landscape to characterize the PrgI needle's mutability at each position as well as the mutations which lead to enhanced T3SS secretion. We discovered new design rules for building a functional T3SS as well as identified hypersecreting mutants. This work can be used to increase understanding of the T3SS's assembly and identify further targets for engineering. This work also provides a blueprint for future efforts to engineer other complex protein assemblies through the construction of fitness landscapes.IMPORTANCEProtein secretion offers a simplified alternative method for protein purification from bacterial hosts. However, the current state-of-the-art methods for protein secretion in bacteria are still hindered by low yields relative to traditional protein purification strategies. Engineers are now seeking strategies to enhance protein secretion titers from bacterial hosts, often through genetic manipulations. In this study, we demonstrate that protein engineering strategies focused on altering the secretion apparatus can be a fruitful avenue toward this goal. Specifically, this study focuses on how changes to the PrgI needle protein from the type 3 secretion system from Salmonella enterica can impact secretion titer. We demonstrate that this complex is amenable to comprehensive mutagenesis studies and that this can yield both PrgI variants with increased secretory capabilities and insight into the normal functioning of the type 3 secretion system.


Subject(s)
Bacterial Proteins , Mutagenesis , Salmonella enterica , Type III Secretion Systems , Type III Secretion Systems/genetics , Type III Secretion Systems/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Salmonella enterica/genetics , Salmonella enterica/metabolism , Gene Library , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism
7.
Biochemistry ; 63(18): 2266-2279, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39189508

ABSTRACT

As part of its pathogenesis, Salmonella enterica serovar Typhimurium delivers effector proteins into host cells. One effector is SspH2, a member of the so-called novel E3 ubiquitin ligase family, that interacts with and enhances, NOD1 pro-inflammatory signaling, though the underlying mechanisms are unclear. Here, we report that SspH2 interacts with multiple members of the NLRC family to enhance pro-inflammatory signaling by targeted ubiquitination. We show that SspH2 modulates host innate immunity by interacting with both NOD1 and NOD2 in mammalian epithelial cell culture via the NF-κB pathway. Moreover, purified SspH2 and NOD1 directly interact, where NOD1 potentiates SspH2 E3 ubiquitin ligase activity. Mass spectrometry and mutational analyses identified four key lysine residues in NOD1 that are required for its enhanced activation by SspH2, but not its basal activity. These critical lysine residues are positioned in the same region of NOD1 and define a surface on the receptor that appears to be targeted by SspH2. Overall, this work provides evidence for post-translational modification of NOD1 by ubiquitin and uncovers a unique mechanism of spatially selective ubiquitination to enhance the activation of an archetypal NLR.


Subject(s)
Nod1 Signaling Adaptor Protein , Salmonella typhimurium , Signal Transduction , Ubiquitination , Nod1 Signaling Adaptor Protein/metabolism , Nod1 Signaling Adaptor Protein/genetics , Humans , Salmonella typhimurium/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Nod2 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/chemistry , HEK293 Cells , Immunity, Innate , Inflammation/metabolism , Inflammation/microbiology , NF-kappa B/metabolism , Salmonella Infections/metabolism , Salmonella Infections/microbiology , Salmonella Infections/immunology
8.
Nat Microbiol ; 9(10): 2653-2665, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39164392

ABSTRACT

Salmonella translocate to the gut epithelium via microfold cells lining the follicle-associated epithelium (FAE). How Salmonella localize to the FAE is not well characterized. Here we use live imaging and competitive assays between wild-type and chemotaxis-deficient mutants to show that Salmonella enterica serotype Typhimurium (S. Typhimurium) localize to the FAE independently of chemotaxis in an ex vivo mouse caecum infection model. Electrical recordings revealed polarized FAE with sustained outward current and small transepithelial potential, while the surrounding villus is depolarized with inward current and large transepithelial potential. The distinct electrical potentials attracted S. Typhimurium to the FAE while Escherichia coli (E. coli) localized to the villi, through a process called galvanotaxis. Chloride flux involving the cystic fibrosis transmembrane conductance regulator (CFTR) generated the ionic currents around the FAE. Pharmacological inhibition of CFTR decreased S. Typhimurium FAE localization but increased E. coli recruitment. Altogether, our findings demonstrate that bioelectric cues contribute to S. Typhimurium targeting of specific gut epithelial locations, with potential implications for other enteric bacterial infections.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator , Escherichia coli , Intestinal Mucosa , Salmonella typhimurium , Animals , Mice , Salmonella typhimurium/physiology , Salmonella typhimurium/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Escherichia coli/metabolism , Escherichia coli/physiology , Escherichia coli/genetics , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Chemotaxis , Cecum/microbiology , Mice, Inbred C57BL , Female , Disease Models, Animal , Humans
9.
J Bacteriol ; 206(8): e0001024, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39046248

ABSTRACT

Catabolite repression is a mechanism of selectively utilizing preferred nutrient sources by redirecting the metabolic pathways. Therefore, it prevents non-essential energy expenditure by repressing the genes and proteins involved in the metabolism of other less favored nutrient sources. Catabolite repressor protein (CRP) is a chief mediator of catabolite repression in microorganisms. In this context, we investigated the role of CRP in starvation tolerance, at both cell physiology and molecular level, by comparing the growth, survival, competitive fitness, maintenance rate, and gene and protein expression of wild type (WT) and ∆crp of Salmonella Typhimurium, under nutrient-rich and minimal medium condition. The ∆crp shows slow growth upon the arrival of nutrient-limiting conditions, poor survival under prolong-starvation, and inability to compete with its counterpart WT strain in nutrient-rich [Luria broth (LB)] and glucose-supplemented M9 minimal medium. Surprisingly, we observed that the survival and competitive fitness of ∆crp are influenced by the composition of the growth medium. Consequently, compared to the glucose-supplemented M9 medium, ∆crp shows faster death and a higher maintenance rate in the LB medium. The comparative gene and protein expression studies of WT and ∆crp in LB medium show that ∆crp has partial or complete loss of repression from CRP-controlled genes, resulting in a high abundance of hundreds of proteins in ∆crp compared to WT. Subsequently, the addition of metabolizable sugar or fresh nutrients to the competing culture showed extended survival of ∆crp. Therefore, our results suggest that CRP-mediated gene repression improves starvation tolerance and competitive fitness of Salmonella Typhimurium by adapting its cellular maintenance rate to environmental conditions.IMPORTANCESalmonella Typhimurium is a master at adapting to chronic starvation conditions. However, the molecular mechanisms to adapt to such conditions are still unknown. In this context, we have evaluated the role of catabolite repressor protein (CRP), a dual transcriptional regulator, in providing survival and competitive fitness under starvation conditions. Also, it showed an association between CRP and nutrient composition. We observed that Δcrp growing on alternate carbon sources has lower survival and competitive fitness than Δcrp growing on glucose as a carbon source. We observed that this is due to the loss of repression from the glucose and CRP-controlled genes, resulting in elevated cellular metabolism (a high maintenance rate) of the Δcrp during growth in a medium having a carbon source other than glucose (e.g., Luria broth medium).


Subject(s)
Bacterial Proteins , Culture Media , Cyclic AMP Receptor Protein , Gene Expression Regulation, Bacterial , Salmonella typhimurium , Salmonella typhimurium/metabolism , Salmonella typhimurium/genetics , Salmonella typhimurium/physiology , Salmonella typhimurium/growth & development , Cyclic AMP Receptor Protein/metabolism , Cyclic AMP Receptor Protein/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Culture Media/chemistry , Catabolite Repression , Microbial Viability , Glucose/metabolism
10.
J Bacteriol ; 206(8): e0017824, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39082861

ABSTRACT

Ciprofloxacin-resistant Salmonella Typhimurium (S. Typhimurium) causes a significant health burden worldwide. A wealth of studies has been published on the contributions of different mechanisms to ciprofloxacin resistance in Salmonella spp. But we still lack a deep understanding of the physiological responses and genetic changes that underlie ciprofloxacin exposure. This study aims to know how phenotypic and genotypic characteristics are impacted by ciprofloxacin exposure, from ciprofloxacin-susceptible to ciprofloxacin-resistant strains in vitro. Here, we investigated the multistep evolution of resistance in replicate populations of S. Typhimurium during 24 days of continuously increasing ciprofloxacin exposure and assessed how ciprofloxacin impacts physiology and genetics. Numerous studies have demonstrated that RamA is a global transcriptional regulator that prominently perturbs the transcriptional landscape of S. Typhimurium, resulting in a ciprofloxacin-resistant phenotype appearing first; the quinolone resistance-determining region mutation site can only be detected later. Comparing the microbial physiological changes and RNA sequencing (RNA-Seq) results of ancestral and selectable mutant strains, the selectable mutant strains had some fitness costs, such as decreased virulence, an increase of biofilm-forming ability, a change of "collateral" sensitivity to other drugs, and inability to utilize galactitol. Importantly, in the ciprofloxacin induced, RamA directly binds and activates the gatR gene responsible for the utilization of galactitol, but RamA deletion strains could not activate gatR. The elevated levels of RamA, which inhibit the galactitol metabolic pathway through the activation of gatR, can lead to a reduction in the growth rate, adhesion, and colonization resistance of S. Typhimurium. This finding is supported by studies conducted in M9 medium as well as in vivo infection models. IMPORTANCE: Treatment of antibiotic resistance can significantly benefit from a deeper understanding of the interactions between drugs and genetics. The physiological responses and genetic mechanisms in antibiotic-exposed bacteria are not well understood. Traditional resistance studies, often retrospective, fail to capture the entire resistance development process and typically exhibit unpredictable dynamics. To explore how clinical isolates of S. Typhimurium respond to ciprofloxacin, we analyzed their adaptive responses. We found that S. Typhimurium RamA-mediated regulation disrupts microbial metabolism under ciprofloxacin exposure, affecting genes in the galactitol metabolic pathways. This disruption facilitates adaptive responses to drug therapy and enhances the efficiency of intracellular survival. A more comprehensive and integrated understanding of these physiological and genetic changes is crucial for improving treatment outcomes.


Subject(s)
Anti-Bacterial Agents , Ciprofloxacin , Drug Resistance, Bacterial , Gene Expression Regulation, Bacterial , Salmonella typhimurium , Ciprofloxacin/pharmacology , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Virulence , Drug Resistance, Bacterial/genetics , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Animals , Mice , Biofilms/drug effects , Biofilms/growth & development , Microbial Sensitivity Tests , Mutation
11.
ACS Synth Biol ; 13(7): 2177-2187, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-38968698

ABSTRACT

Transcription factor (TF)-based biosensors have arisen as powerful tools in the advancement of metabolic engineering. However, with the emergence of numerous bioproduction targets, the variety of applicable TF-based biosensors remains severely limited. In this study, we investigated and engineered an 1,2-propanediol (1,2-PD)-responsive transcription activator, PocR, from Salmonella typhimurium to enrich the current biosensor repertoire. Heterologous characterization of PocR in E. coli revealed a significantly limited operational range and dynamic range, primarily attributed to the leaky binding between PocR and its corresponding promoters in the absence of the 1,2-PD inducer. Promiscuity characterization uncovered the minor responsiveness of PocR toward glycerol and 1,2-butanediol (1,2-BD). Using AlphaFold-predicted structure and protein mutagenesis, we preliminarily explored the underlying mechanism of PocR. Based on the investigated mechanism, we engineered a PcoR-F46R/G105D variant with an altered inducer specificity to glycerol, as well as a PocR-ARE (Q107A/S192R/A203E) variant with nearly a 4-fold higher dynamic range (6.7-fold activation) and a 20-fold wider operational range (0-20 mM 1,2-PD). Finally, we successfully converted PocR to a repressor through promoter engineering. Integrating the activation and repression functions established a versatile 1,2-PD-induced bifunctional regulation system based on PocR-ARE. Our work showcases the exploration and exploitation of an underexplored type of transcriptional activator capable of recruiting RNA polymerase. It also expands the biosensor toolbox by providing a 1,2-PD-responsive bifunctional regulator and glycerol-responsive activator.


Subject(s)
Biosensing Techniques , Escherichia coli , Metabolic Engineering , Propylene Glycol , Salmonella typhimurium , Transcription Factors , Biosensing Techniques/methods , Transcription Factors/genetics , Transcription Factors/metabolism , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Propylene Glycol/metabolism , Metabolic Engineering/methods , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Glycerol/metabolism , Promoter Regions, Genetic/genetics
12.
J Biol Chem ; 300(7): 107427, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38823641

ABSTRACT

Salmonella enterica serovar Typhimurium melibiose permease (MelBSt) is a prototype of the major facilitator superfamily (MFS) transporters, which play important roles in human health and diseases. MelBSt catalyzed the symport of galactosides with Na+, Li+, or H+ but prefers the coupling with Na+. Previously, we determined the structures of the inward- and outward-facing conformation of MelBSt and the molecular recognition for galactoside and Na+. However, the molecular mechanisms for H+- and Na+-coupled symport remain poorly understood. In this study, we solved two x-ray crystal structures of MelBSt, the cation-binding site mutants D59C at an unliganded apo-state and D55C at a ligand-bound state, and both structures display the outward-facing conformations virtually identical as published. We determined the energetic contributions of three major Na+-binding residues for the selection of Na+ and H+ by free energy simulations. Transport assays showed that the D55C mutant converted MelBSt to a solely H+-coupled symporter, and together with the free-energy perturbation calculation, Asp59 is affirmed to be the sole protonation site of MelBSt. Unexpectedly, the H+-coupled melibiose transport exhibited poor activities at greater bulky ΔpH and better activities at reversal ΔpH, supporting the novel theory of transmembrane-electrostatically localized protons and the associated membrane potential as the primary driving force for the H+-coupled symport mediated by MelBSt. This integrated study of crystal structure, bioenergetics, and free energy simulations, demonstrated the distinct roles of the major binding residues in the cation-binding pocket of MelBSt.


Subject(s)
Sodium , Symporters , Symporters/chemistry , Symporters/metabolism , Symporters/genetics , Binding Sites , Crystallography, X-Ray , Sodium/metabolism , Sodium/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Salmonella typhimurium/metabolism , Salmonella typhimurium/genetics , Salmonella typhimurium/chemistry , Melibiose/metabolism , Melibiose/chemistry , Cations/metabolism , Cations/chemistry , Protein Conformation
13.
J Biol Chem ; 300(7): 107424, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38823640

ABSTRACT

Lysozyme is a ß-1,4-glycosidase that hydrolyzes the polysaccharide backbone of bacterial cell walls. With an additional bactericidal function mediated by a separate protein domain, lysozyme is considered a uniquely important antimicrobial molecule contributing to the host's innate immune response to infection. Elevated lysozyme production is found in various inflammatory conditions while patients with genetic risks for inflammatory bowel diseases demonstrate abnormal lysozyme expression, granule packaging, and secretion in Paneth cells. However, it remains unclear how a gain- or loss-of-function in host lysozyme may impact the host inflammatory responses to pathogenic infection. We challenged Lyz1-/- and ectopic Lyz1-expressing (Villin-Lyz1TG) mice with S. Typhimurium and then comprehensively assessed the inflammatory disease progression. We conducted proteomics analysis to identify molecules derived from human lysozyme-mediated processing of live Salmonella. We examined the barrier-impairing effects of these identified molecules in human intestinal epithelial cell monolayer and enteroids. Lyz1-/- mice are protected from infection in terms of morbidity, mortality, and barrier integrity, whereas Villin-Lyz1TG mice demonstrate exacerbated infection and inflammation. The growth and invasion of Salmonella in vitro are not affected by human or chicken lysozyme, whereas lysozyme encountering of live Salmonella stimulates the release of barrier-disrupting factors, InvE-sipC and Lpp1, which directly or indirectly impair the tight junctions. The direct engagement of host intestinal lysozyme with an enteric pathogen such as Salmonella promotes the release of virulence factors that are barrier-impairing and pro-inflammatory. Controlling lysozyme function may help alleviate the inflammatory progression.


Subject(s)
Muramidase , Salmonella typhimurium , Muramidase/metabolism , Animals , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , Mice , Humans , Salmonella Infections/microbiology , Salmonella Infections/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice, Knockout , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Microfilament Proteins
14.
Nat Microbiol ; 9(7): 1792-1811, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38862602

ABSTRACT

The Klebsiella oxytoca species complex is part of the human microbiome, especially during infancy and childhood. K. oxytoca species complex strains can produce enterotoxins, namely, tilimycin and tilivalline, while also contributing to colonization resistance (CR). The relationship between these seemingly contradictory roles is not well understood. Here, by coupling ex vivo assays with CRISPR-mutagenesis and various mouse models, we show that K. oxytoca provides CR against Salmonella Typhimurium. In vitro, the antimicrobial activity against various Salmonella strains depended on tilimycin production and was induced by various simple carbohydrates. In vivo, CR against Salmonella depended on toxin production in germ-free mice, while it was largely toxin-independent in mice with residual microbiota. This was linked to the relative levels of toxin-inducing carbohydrates in vivo. Finally, dulcitol utilization was essential for toxin-independent CR in gnotobiotic mice. Together, this demonstrates that nutrient availability is key to both toxin-dependent and substrate-driven competition between K. oxytoca and Salmonella.


Subject(s)
Klebsiella oxytoca , Salmonella Infections , Salmonella typhimurium , Klebsiella oxytoca/genetics , Klebsiella oxytoca/metabolism , Animals , Mice , Salmonella Infections/microbiology , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Salmonella typhimurium/growth & development , Salmonella typhimurium/drug effects , Humans , Disease Models, Animal , Enterotoxins/metabolism , Enterotoxins/genetics , Female , Mice, Inbred C57BL , Klebsiella Infections/microbiology , Microbiota , Gastrointestinal Microbiome , Antibiosis , Benzodiazepinones
15.
PLoS Biol ; 22(6): e3002616, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38865418

ABSTRACT

The gastrointestinal tract is densely colonized by a polymicrobial community known as the microbiota which serves as primary line of defence against pathogen invasion. The microbiota can limit gut-luminal pathogen growth at different stages of infection. This can be traced to specific commensal strains exhibiting direct or indirect protective functions. Although these mechanisms hold the potential to develop new approaches to combat enteric pathogens, they remain far from being completely described. In this study, we investigated how a mouse commensal Escherichia coli can outcompete Salmonella enterica serovar Typhimurium (S. Tm). Using a salmonellosis mouse model, we found that the commensal E. coli 8178 strain relies on a trojan horse trap strategy to limit S. Tm expansion in the inflamed gut. Combining mutants and reporter tools, we demonstrated that inflammation triggers the expression of the E. coli 8178 antimicrobial microcin H47 toxin which, when fused to salmochelin siderophores, can specifically alter S. Tm growth. This protective function was compromised upon disruption of the E. coli 8178 tonB-dependent catecholate siderophore uptake system, highlighting a previously unappreciated crosstalk between iron intake and microcin H47 activity. By identifying the genetic determinants mediating S. Tm competition, our work not only provides a better mechanistic understanding of the protective function displayed by members of the gut microbiota but also further expands the general contribution of microcins in bacterial antagonistic relationships. Ultimately, such insights can open new avenues for developing microbiota-based approaches to better control intestinal infections.


Subject(s)
Escherichia coli , Inflammation , Salmonella typhimurium , Siderophores , Animals , Escherichia coli/metabolism , Escherichia coli/genetics , Siderophores/metabolism , Mice , Salmonella typhimurium/pathogenicity , Salmonella typhimurium/metabolism , Inflammation/metabolism , Inflammation/microbiology , Mice, Inbred C57BL , Bacteriocins/metabolism , Bacteriocins/pharmacology , Gastrointestinal Microbiome , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Salmonella Infections/microbiology , Salmonella Infections/metabolism , Female , Iron/metabolism , Symbiosis , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/metabolism
16.
Methods Mol Biol ; 2813: 107-115, 2024.
Article in English | MEDLINE | ID: mdl-38888773

ABSTRACT

Mass spectrometry-based proteomics provides a wealth of information about changes in protein production and abundance under diverse conditions, as well as mechanisms of regulation, signaling cascades, interaction partners, and communication patterns across biological systems. For profiling of intracellular pathogens, proteomic profiling can be performed in the absence of a host to singularly define the pathogenic proteome or during an infection-like setting to identify dual perspectives of infection. In this chapter, we present techniques to extract proteins from the human bacterial intracellular pathogen, Salmonella enterica serovar Typhimurium, in the presence of macrophages, an important innate immune cell in host defense. We outline sample preparation, including protein extraction, digestion, and purification, as well as mass spectrometry measurements and bioinformatics analysis. The data generated from our dual perspective profiling approach provides new insight into pathogen and host protein modulation under infection-like conditions.


Subject(s)
Bacterial Proteins , Macrophages , Proteomics , Salmonella typhimurium , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , Proteomics/methods , Humans , Bacterial Proteins/metabolism , Macrophages/microbiology , Macrophages/metabolism , Host-Pathogen Interactions , Proteome/metabolism , Salmonella Infections/microbiology , Salmonella Infections/metabolism , Salmonella Infections/immunology , Computational Biology/methods , Mass Spectrometry/methods
17.
mBio ; 15(7): e0112824, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38904384

ABSTRACT

The injectisome encoded by Salmonella pathogenicity island 2 (SPI-2) had been thought to translocate 28 effectors. Here, we used a proteomic approach to characterize the secretome of a clinical strain of invasive non-typhoidal Salmonella enterica serovar Enteritidis that had been mutated to cause hyper-secretion of the SPI-2 injectisome effectors. Along with many known effectors, we discovered the novel SseM protein. sseM is widely distributed among the five subspecies of Salmonella enterica, is found in many clinically relevant serovars, and is co-transcribed with pipB2, a SPI-2 effector gene. The translocation of SseM required a functional SPI-2 injectisome. Following expression in human cells, SseM interacted with five components of the dystrophin-associated protein complex (DAPC), namely, ß-2-syntrophin, utrophin/dystrophin, α-catulin, α-dystrobrevin, and ß-dystrobrevin. The interaction between SseM and ß-2-syntrophin and α-dystrobrevin was verified in Salmonella Typhimurium-infected cells and relied on the postsynaptic density-95/discs large/zonula occludens-1 (PDZ) domain of ß-2-syntrophin and a sequence corresponding to a PDZ-binding motif (PBM) in SseM. A ΔsseM mutant strain had a small competitive advantage over the wild-type strain in the S. Typhimurium/mouse model of systemic disease. This phenotype was complemented by a plasmid expressing wild-type SseM from S. Typhimurium or S. Enteritidis and was dependent on the PBM of SseM. Therefore, a PBM within a Salmonella effector mediates interactions with the DAPC and modulates the systemic growth of bacteria in mice. Furthermore, the ΔsseM mutant strain displayed enhanced replication in bone marrow-derived macrophages, demonstrating that SseM restrains intracellular bacterial growth to modulate Salmonella virulence. IMPORTANCE: In Salmonella enterica, the injectisome machinery encoded by Salmonella pathogenicity island 2 (SPI-2) is conserved among the five subspecies and delivers proteins (effectors) into host cells, which are required for Salmonella virulence. The identification and functional characterization of SPI-2 injectisome effectors advance our understanding of the interplay between Salmonella and its host(s). Using an optimized method for preparing secreted proteins and a clinical isolate of the invasive non-typhoidal Salmonella enterica serovar Enteritidis strain D24359, we identified 22 known SPI-2 injectisome effectors and one new effector-SseM. SseM modulates bacterial growth during murine infection and has a sequence corresponding to a postsynaptic density-95/discs large/zonula occludens-1 (PDZ)-binding motif that is essential for interaction with the PDZ-containing host protein ß-2-syntrophin and other components of the dystrophin-associated protein complex (DAPC). To our knowledge, SseM is unique among Salmonella effectors in containing a functional PDZ-binding motif and is the first bacterial protein to target the DAPC.


Subject(s)
Bacterial Proteins , Salmonella enteritidis , Animals , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Humans , Mice , Virulence , Salmonella enteritidis/genetics , Salmonella enteritidis/metabolism , Salmonella enteritidis/pathogenicity , Virulence Factors/metabolism , Virulence Factors/genetics , Salmonella Infections/microbiology , Dystrophin-Associated Proteins/metabolism , Dystrophin-Associated Proteins/genetics , Genomic Islands , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , Proteomics , Disease Models, Animal , Membrane Proteins/metabolism , Membrane Proteins/genetics
18.
Int J Food Microbiol ; 419: 110753, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38797020

ABSTRACT

Salmonella enterica subsp. enterica serovar Typhimurium variant 4,[5],12:i:- (so called S. 4,[5],12:i:-) has rapidly become one of the most prevalent serovars in humans in Europe, with clinical cases associated with foodborne from pork products. The mechanisms, genetic basis and biofilms relevance by which S. 4,[5],12:i:- maintains and spreads its presence in pigs remain unclear. In this study, we examined the genetic basis of biofilm production in 78 strains of S. 4,[5],12:i:- (n = 57) and S. Typhimurium (n = 21), from human gastroenteritis, food products and asymptomatic pigs. The former showed a lower Specific Biofilm Formation index (SBF) and distant phylogenetic clades, suggesting that the ability to form biofilms is not a crucial adaptation for the S. 4,[5],12:i:- emerging success in pigs. However, using a pan-Genome-Wide Association Study (pan-GWAS) we identified genetic determinants of biofilm formation, revealing 167 common orthologous groups and genes associated with the SBF. The analysis of annotated sequences highlighted specific genetic deletions in three chromosomal regions of S. 4,[5],12:i:- correlating with SBF values: i) the complete fimbrial operon stbABCDE widely recognized as the most critical factor involved in Salmonella adherence; ii) the hxlA, hlxB, and pgiA genes, which expression in S. Typhimurium is induced in the tonsils during swine infection, and iii) the entire iroA locus related to the characteristic deletion of the second-phase flagellar genomic region in S. 4,[5],12:i:-. Consequently, we further investigated the role of the iro-genes on biofilm by constructing S. Typhimurium deletion mutants in iroBCDE and iroN. While iroBCDE showed no significant impact, iroN clearly contributed to S. Typhimurium biofilm formation. In conclusion, the pan-GWAS approach allowed us to uncover complex interactions between genetic and phenotypic factors influencing biofilm formation in S. 4,[5],12:i:- and S. Typhimurium.


Subject(s)
Bacterial Proteins , Biofilms , Genome-Wide Association Study , Salmonella typhimurium , Biofilms/growth & development , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Animals , Swine , Humans , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Iron/metabolism , Phylogeny , Salmonella Infections, Animal/microbiology , Salmonella Infections/microbiology , Gastroenteritis/microbiology , Serogroup
19.
PLoS Genet ; 20(5): e1011287, 2024 May.
Article in English | MEDLINE | ID: mdl-38768229

ABSTRACT

In many organisms, stress responses to adverse environments can trigger secondary functions of certain proteins by altering protein levels, localization, activity, or interaction partners. Escherichia coli cells respond to the presence of specific cationic antimicrobial peptides by strongly activating the PhoQ/PhoP two-component signaling system, which regulates genes important for growth under this stress. As part of this pathway, a biosynthetic enzyme called QueE, which catalyzes a step in the formation of queuosine (Q) tRNA modification is upregulated. When cellular QueE levels are high, it co-localizes with the central cell division protein FtsZ at the septal site, blocking division and resulting in filamentous growth. Here we show that QueE affects cell size in a dose-dependent manner. Using alanine scanning mutagenesis of amino acids in the catalytic active site, we pinpoint residues in QueE that contribute distinctly to each of its functions-Q biosynthesis or regulation of cell division, establishing QueE as a moonlighting protein. We further show that QueE orthologs from enterobacteria like Salmonella typhimurium and Klebsiella pneumoniae also cause filamentation in these organisms, but the more distant counterparts from Pseudomonas aeruginosa and Bacillus subtilis lack this ability. By comparative analysis of E. coli QueE with distant orthologs, we elucidate a unique region in this protein that is responsible for QueE's secondary function as a cell division regulator. A dual-function protein like QueE is an exception to the conventional model of "one gene, one enzyme, one function", which has divergent roles across a range of fundamental cellular processes including RNA modification and translation to cell division and stress response.


Subject(s)
Cell Division , Escherichia coli Proteins , Escherichia coli , Escherichia coli/genetics , Escherichia coli/metabolism , Cell Division/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Nucleoside Q/metabolism , Nucleoside Q/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Klebsiella pneumoniae/genetics , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Gene Expression Regulation, Bacterial , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/genetics , RNA, Transfer/genetics , RNA, Transfer/metabolism
20.
Nat Commun ; 15(1): 3120, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38600106

ABSTRACT

Salmonella utilizes a type 3 secretion system to translocate virulence proteins (effectors) into host cells during infection1. The effectors modulate host cell machinery to drive uptake of the bacteria into vacuoles, where they can establish an intracellular replicative niche. A remarkable feature of Salmonella invasion is the formation of actin-rich protuberances (ruffles) on the host cell surface that contribute to bacterial uptake. However, the membrane source for ruffle formation and how these bacteria regulate membrane mobilization within host cells remains unclear. Here, we show that Salmonella exploits membrane reservoirs for the generation of invasion ruffles. The reservoirs are pre-existing tubular compartments associated with the plasma membrane (PM) and are formed through the activity of RAB10 GTPase. Under normal growth conditions, membrane reservoirs contribute to PM homeostasis and are preloaded with the exocyst subunit EXOC2. During Salmonella invasion, the bacterial effectors SipC, SopE2, and SopB recruit exocyst subunits from membrane reservoirs and other cellular compartments, thereby allowing exocyst complex assembly and membrane delivery required for bacterial uptake. Our findings reveal an important role for RAB10 in the establishment of membrane reservoirs and the mechanisms by which Salmonella can exploit these compartments during host cell invasion.


Subject(s)
Salmonella Infections , Salmonella typhimurium , Humans , Salmonella typhimurium/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Salmonella Infections/microbiology , Cell Membrane/metabolism , Membranes/metabolism , HeLa Cells
SELECTION OF CITATIONS
SEARCH DETAIL