Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.580
Filter
1.
Vaccine ; 42(19S1): S101-S124, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39003017

ABSTRACT

Invasive non-typhoidal Salmonella (iNTS) disease is an under-recognized high-burden disease causing major health and socioeconomic issues in sub-Saharan Africa (sSA), predominantly among immune-naïve infants and young children, including those with recognized comorbidities such as HIV infection. iNTS disease is primarily caused by Salmonella enterica serovar Typhimurium sequence type (ST) 313 and 'African-restricted clades' of Salmonella Enteritidis ST11 that have emerged across the African continent as a series of epidemics associated with acquisition of new antimicrobial resistance. Due to genotypes with a high prevalence of antimicrobial resistance and scarcity of therapeutic options, these NTS serovars are designated by the World Health Organization as a priority pathogen for research and development of interventions, including vaccines, to address and reduce NTS associated bacteremia and meningitis in sSA. Novel and traditional vaccine technologies are being applied to develop vaccines against iNTS disease, and the results of the first clinical trials in the infant target population should become available in the near future. The "Vaccine Value Profile" (VVP) addresses information related predominantly to invasive disease caused by Salmonella Enteritidis and Salmonella Typhimurium prevalent in sSA. Information is included on stand-alone iNTS disease candidate vaccines and candidate vaccines targeting iNTS disease combined with another invasive serotype, Salmonella Typhi, that is also common across sSA. Out of scope for the first version of this VVP is a wider discussion on either diarrheagenic NTS disease (dNTS) also associated with Salmonella Enteritidis and Salmonella Typhimurium or the development of a multivalent Salmonella vaccines targeting key serovars for use globally. This VVP for vaccines to prevent iNTS disease is intended to provide a high-level, holistic assessment of the information and data that are currently available to inform the potential public health, economic, and societal value of pipeline vaccines and vaccine-like products. Future versions of this VVP will be updated to reflect ongoing activities such as vaccine development strategies and a "Full Vaccine Value Assessment" that will inform the value proposition of an iNTS disease vaccine. This VVP was developed by a working group of subject matter experts from academia, non-profit organizations, public private partnerships, and multi-lateral organizations, and in collaboration with stakeholders from the World Health Organization African Region. All contributors have extensive expertise on various elements of the iNTS disease VVP and collectively aimed to identify current research and knowledge gaps. The VVP was developed using only existing and publicly available information.


Subject(s)
Salmonella Infections , Salmonella Vaccines , Salmonella enteritidis , Humans , Africa South of the Sahara/epidemiology , Salmonella enteritidis/immunology , Salmonella enteritidis/genetics , Salmonella enteritidis/pathogenicity , Salmonella Infections/prevention & control , Salmonella Infections/epidemiology , Salmonella Infections/microbiology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Salmonella typhimurium/genetics , Salmonella Vaccines/immunology , Salmonella Vaccines/administration & dosage
2.
Sci Rep ; 14(1): 15160, 2024 07 02.
Article in English | MEDLINE | ID: mdl-38956132

ABSTRACT

In order to survive and replicate, Salmonella has evolved mechanisms to gain access to intestinal epithelial cells of the crypt. However, the impact of Salmonella Typhimurium on stem cells and progenitors, which are responsible for the ability of the intestinal epithelium to renew and protect itself, remains unclear. Given that intestinal organoids growth is sustained by stem cells and progenitors activity, we have used this model to document the effects of Salmonella Typhimurium infection on epithelial proliferation and differentiation, and compared it to an in vivo model of Salmonella infection in mice. Among gut segments, the caecum was preferentially targeted by Salmonella. Analysis of infected crypts and organoids demonstrated increased length and size, respectively. mRNA transcription profiles of infected crypts and organoids pointed to upregulated EGFR-dependent signals, associated with a decrease in secretory cell lineage differentiation. To conclude, we show that organoids are suited to mimic the impact of Salmonella on stem cells and progenitors cells, carrying a great potential to drastically reduce the use of animals for scientific studies on that topic. In both models, the EGFR pathway, crucial to stem cells and progenitors proliferation and differentiation, is dysregulated by Salmonella, suggesting that repeated infections might have consequences on crypt integrity and further oncogenesis.


Subject(s)
Cell Differentiation , ErbB Receptors , Organoids , Salmonella Infections , Salmonella typhimurium , Stem Cells , Animals , Organoids/microbiology , Stem Cells/metabolism , Mice , Salmonella typhimurium/pathogenicity , Salmonella typhimurium/physiology , Salmonella Infections/microbiology , Salmonella Infections/pathology , ErbB Receptors/metabolism , ErbB Receptors/genetics , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Cell Proliferation , Disease Models, Animal , Mice, Inbred C57BL
3.
Front Cell Infect Microbiol ; 14: 1416537, 2024.
Article in English | MEDLINE | ID: mdl-39040600

ABSTRACT

Infection of ruminants such as cattle with Mycobacterium avium subsp. paratuberculosis (MAP) causes Johne's disease, a disease characterized by chronic inflammation of the small intestine and diarrhoea. Infection with MAP is acquired via the faecal-to-oral route and the pathogen initially invades the epithelial lining of the small intestine. In this study we used an in vitro 3D mouse enteroid model to determine the influence of M cells in infection of the gut epithelia by MAP, in comparison with another bacterial intestinal pathogen of veterinary importance, Salmonella enterica serovar Typhimurium. The differentiation of M cells in the enteroid cultures was induced by stimulation with the cytokine receptor activator of nuclear factor-κB ligand (RANKL), and the effects on MAP and Salmonella uptake and intracellular survival were determined. The presence of M cells in the cultures correlated with increased uptake and intracellular survival of Salmonella, but had no effect on MAP. Interestingly neither pathogen was observed to preferentially accumulate within GP2-positive M cells.


Subject(s)
Mycobacterium avium subsp. paratuberculosis , Salmonella typhimurium , Animals , Mycobacterium avium subsp. paratuberculosis/physiology , Salmonella typhimurium/physiology , Salmonella typhimurium/pathogenicity , Mice , Paratuberculosis/microbiology , Microbial Viability , Intestinal Mucosa/microbiology , Cattle , M Cells
4.
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
5.
Arch Microbiol ; 206(7): 302, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38874634

ABSTRACT

Host-generated antimicrobial peptides (AMPs) play a pivotal role in defense against bacterial pathogens. AMPs kill invading bacteria majorly by disrupting the bacterial cell walls. AMPs are actively synthesized and released into the lumen of the gastrointestinal tract to limit colonization of enteric pathogens like Salmonella typhimurium (S. typhimurium). However, S. typhimurium has evolved several resistance mechanisms to defend AMPs. The multicomponent SapABCDF uptake transporter is one such system that helps in resisting AMPs. In the current study, we analyzed the role of S. typhimurium SapA against stress survival and virulence of this bacterium. ∆sapA mutant strain showed hypersensitivity to AMPs, like melittin and mastoparan. Further, ∆sapA mutant showed more than 22 folds (p = 0.019) hypersensitivity to neutrophils as compared to the WT strain of S. typhimurium. In addition, ∆sapA strain showed defective survival in mice. In conclusion, the results of the current study suggest that the SapA is essential for survival against AMPs and virulence of S. typhimurium.


Subject(s)
Neutrophils , Salmonella typhimurium , Animals , Salmonella typhimurium/drug effects , Salmonella typhimurium/pathogenicity , Salmonella typhimurium/genetics , Mice , Neutrophils/immunology , Neutrophils/microbiology , Virulence , Antimicrobial Peptides/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Female , Mice, Inbred BALB C , Salmonella Infections/microbiology , Antimicrobial Cationic Peptides/pharmacology
6.
Sci Rep ; 14(1): 14586, 2024 06 25.
Article in English | MEDLINE | ID: mdl-38918457

ABSTRACT

Natural killer (NK) cells play a key role in defense against Salmonella infections during the early phase of infection. Our previous work showed that the excretory/secretory products of Ascaris suum repressed NK activity in vitro. Here, we asked if NK cell functionality was influenced in domestic pigs during coinfection with Ascaris and Salmonella enterica serotype Typhimurium. Ascaris coinfection completely abolished the IL-12 and IL-18 driven elevation of IFN-γ production seen in CD16 + CD8α + perforin + NK cells of Salmonella single-infected pigs. Furthermore, Ascaris coinfection prohibited the Salmonella-driven rise in NK perforin levels and CD107a surface expression. In line with impaired effector functions, NK cells from Ascaris-single and coinfected pigs displayed elevated expression of the inhibitory KLRA1 and NKG2A receptors genes, contrasting with the higher expression of the activating NKp46 and NKp30 receptors in NK cells during Salmonella single infection. These differences were accompanied by the highly significant upregulation of T-bet protein expression in NK cells from Ascaris-single and Ascaris/Salmonella coinfected pigs. Together, our data strongly indicate a profound repression of NK functionality by an Ascaris infection which may hinder infected individuals from adequately responding to a concurrent bacterial infection.


Subject(s)
Ascariasis , Coinfection , Killer Cells, Natural , Swine Diseases , Animals , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Ascariasis/immunology , Ascariasis/veterinary , Ascariasis/parasitology , Coinfection/immunology , Coinfection/microbiology , Coinfection/parasitology , Swine , Swine Diseases/parasitology , Swine Diseases/immunology , Swine Diseases/microbiology , Salmonella Infections, Animal/immunology , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Ascaris suum/immunology , Interferon-gamma/metabolism , Perforin/metabolism , Interleukin-12/metabolism , T-Box Domain Proteins/metabolism , T-Box Domain Proteins/genetics , Interleukin-18/metabolism
7.
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
8.
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
9.
Int J Food Microbiol ; 422: 110810, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-38945050

ABSTRACT

To study potential ramifications of antimicrobial resistance, we carried out adaptive laboratory evolution assays (ALE) to isolate three resistant variants (RVs) of Salmonella enterica Typhimurium, employing three different types of food preservation methods: 1) an emergent technology, plasma-activated water (PAW), leading to variant RV-PAW; a traditional method, heat, leading to variant RV-HT, and a natural antimicrobial compound, carvacrol, leading to variant RV-CAR. The variant resistant to plasma-activated water, RV-PAW, had mutations in rpoA and rpoD; it showed increased tolerance to heat in orange juice but ultimately did not pose a significant threat, as it exhibited a fitness cost at refrigeration temperature (8 °C), whereas its virulence against Caenorhabditis elegans decreased. The variant resistant to heat, RV-HT, had mutations in flhC, dnaJ: it exhibited a fitness cost at high growth temperatures (43 °C) and induced morphofunctional alterations in C. elegans. The variant resistant to carvacrol, RV-CAR, had mutations in sseG, flhA, wbaV, lon; this variant not only exhibited significantly higher thermotolerance in both laboratory media and food models but also effectively increased its growth fitness at refrigeration temperatures while retaining its virulence, evidenced by the highest percentage of Smurf phenotype in C. elegans. To address these challenges, we applied a process combining thermal treatment with citral, with the aim of leveraging the sublethal damage caused in RVs by heat treatments in orange juice. This approach achieves enhanced microbial inactivation without having to escalate the intensity of the thermal treatment. The result was particularly encouraging in the case of RV-CAR, the most challenging strain, for which we improved lethality by up to 3 log10 inactivation cycles.


Subject(s)
Caenorhabditis elegans , Food Preservation , Hot Temperature , Salmonella typhimurium , Salmonella typhimurium/genetics , Salmonella typhimurium/growth & development , Salmonella typhimurium/drug effects , Salmonella typhimurium/pathogenicity , Virulence , Caenorhabditis elegans/microbiology , Animals , Food Preservation/methods , Thermotolerance , Mutation , Food Microbiology , Drug Resistance, Bacterial/genetics , Cymenes/pharmacology
10.
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
11.
Virulence ; 15(1): 2357670, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38804638

ABSTRACT

Salmonella enterica subspecies enterica serovar Typhimurium is an intracellular pathogen that invades and colonizes the intestinal epithelium. Following bacterial invasion, Salmonella is enclosed within a membrane-bound vacuole known as a Salmonella-containing vacuole (SCV). However, a subset of Salmonella has the capability to prematurely rupture the SCV and escape, resulting in Salmonella hyper-replication within the cytosol of epithelial cells. A recently published RNA-seq study provides an overview of cytosolic and vacuolar upregulated genes and highlights pagN vacuolar upregulation. Here, using transcription kinetics, protein production profile, and immunofluorescence microscopy, we showed that PagN is exclusively produced by Salmonella in SCV. Gentamicin protection and chloroquine resistance assays were performed to demonstrate that deletion of pagN affects Salmonella replication by affecting the cytosolic bacterial population. This study presents the first example of a Salmonella virulence factor expressed within the endocytic compartment, which has a significant impact on the dynamics of Salmonella cytosolic hyper-replication.


Subject(s)
Bacterial Proteins , Cytosol , Salmonella typhimurium , Vacuoles , Virulence Factors , Salmonella typhimurium/genetics , Salmonella typhimurium/pathogenicity , Cytosol/microbiology , Vacuoles/microbiology , Vacuoles/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism , Humans , Virulence , Salmonella Infections/microbiology , HeLa Cells , Epithelial Cells/microbiology , Gene Expression Regulation, Bacterial
12.
PLoS Biol ; 22(4): e3002597, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38684033

ABSTRACT

Intestinal epithelial cells (IECs) play pivotal roles in nutrient uptake and in the protection against gut microorganisms. However, certain enteric pathogens, such as Salmonella enterica serovar Typhimurium (S. Tm), can invade IECs by employing flagella and type III secretion systems (T3SSs) with cognate effector proteins and exploit IECs as a replicative niche. Detection of flagella or T3SS proteins by IECs results in rapid host cell responses, i.e., the activation of inflammasomes. Here, we introduce a single-cell manipulation technology based on fluidic force microscopy (FluidFM) that enables direct bacteria delivery into the cytosol of single IECs within a murine enteroid monolayer. This approach allows to specifically study pathogen-host cell interactions in the cytosol uncoupled from preceding events such as docking, initiation of uptake, or vacuole escape. Consistent with current understanding, we show using a live-cell inflammasome reporter that exposure of the IEC cytosol to S. Tm induces NAIP/NLRC4 inflammasomes via its known ligands flagellin and T3SS rod and needle. Injected S. Tm mutants devoid of these invasion-relevant ligands were able to grow in the cytosol of IECs despite the absence of T3SS functions, suggesting that, in the absence of NAIP/NLRC4 inflammasome activation and the ensuing cell death, no effector-mediated host cell manipulation is required to render the epithelial cytosol growth-permissive for S. Tm. Overall, the experimental system to introduce S. Tm into single enteroid cells enables investigations into the molecular basis governing host-pathogen interactions in the cytosol with high spatiotemporal resolution.


Subject(s)
Calcium-Binding Proteins , Cytosol , Flagellin , Host-Pathogen Interactions , Inflammasomes , Salmonella typhimurium , Type III Secretion Systems , Cytosol/metabolism , Cytosol/microbiology , Animals , Salmonella typhimurium/pathogenicity , Salmonella typhimurium/metabolism , Type III Secretion Systems/metabolism , Inflammasomes/metabolism , Mice , Flagellin/metabolism , Neuronal Apoptosis-Inhibitory Protein/metabolism , Neuronal Apoptosis-Inhibitory Protein/genetics , Epithelial Cells/microbiology , Epithelial Cells/metabolism , Apoptosis Regulatory Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Mice, Inbred C57BL , CARD Signaling Adaptor Proteins/metabolism , CARD Signaling Adaptor Proteins/genetics , Single-Cell Analysis/methods , Salmonella Infections/microbiology , Salmonella Infections/metabolism , Salmonella Infections/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism
13.
FEBS J ; 291(14): 3104-3127, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38487972

ABSTRACT

Intestinal edema is a common manifestation of numerous gastrointestinal diseases and is characterized by the accumulation of fluid in the interstitial space of the intestinal wall. Technical advances in laser capture microdissection and low-biomass proteomics now allow us to specifically characterize the intestinal edema proteome. Using advanced proteomics, we identify peptides derived from antimicrobial factors with high signal intensity, but also highlight major contributions from the blood clotting system, extracellular matrix (ECM) and protease-protease inhibitor networks. The ECM is a complex fibrillar network of macromolecules that provides structural and mechanical support to the intestinal tissue. One abundant component of the ECM observed in Salmonella-driven intestinal edema is the glycoprotein fibronectin, recognized for its structure-function interplay regulated by mechanical forces. Using mechanosensitive staining of fibronectin fibers reveals that they are tensed in the edema, despite the high abundance of proteases able to cleave fibronectin. In contrast, fibronectin fibers increasingly relax in other cecal tissue areas as the infection progresses. Co-staining for fibrin(ogen) indicates the formation of a provisional matrix in the edema, similar to what is observed in response to skin injury, while collagen staining reveals a sparse and disrupted collagen fiber network. These observations plus the absence of low tensional fibronectin fibers and the additional finding of a high number of protease inhibitors in the edema proteome could indicate a critical role of stretched fibronectin fibers in maintaining tissue integrity in the severely inflamed cecum. Understanding these processes may also provide valuable functional diagnostic markers of intestinal disease progression in the future.


Subject(s)
Edema , Fibronectins , Animals , Fibronectins/metabolism , Mice , Edema/metabolism , Edema/pathology , Edema/microbiology , Extracellular Matrix/metabolism , Proteomics/methods , Mice, Inbred C57BL , Salmonella Infections/microbiology , Salmonella Infections/pathology , Salmonella Infections/metabolism , Intestines/microbiology , Intestines/pathology , Salmonella typhimurium/pathogenicity , Salmonella typhimurium/metabolism
14.
J Cell Physiol ; 239(1): 36-50, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37877586

ABSTRACT

Human enterocytes are primary targets of infection by invasive bacterium Salmonella Typhimurium, and studies using nonintestinal epithelial cells established that S. Typhimurium activates Rho family GTPases, primarily CDC42, to modulate the actin cytoskeletal network for invasion. The host intracellular protein network that engages CDC42 and influences the pathogen's invasive capacity are relatively unclear. Here, proteomic analyses of canonical and variant CDC42 interactomes identified a poorly characterized CDC42 interacting protein, CDC42EP1, whose intracellular localization is rapidly redistributed and aggregated around the invading bacteria. CDC42EP1 associates with SEPTIN-7 and Villin, and its relocalization and bacterial engagement depend on host CDC42 and S. Typhimurium's capability of activating CDC42. Unlike CDC42, CDC42EP1 is not required for S. Typhimurium's initial cellular entry but is found to associate with Salmonella-containing vacuoles after long-term infections, indicating a contribution to the pathogen's intracellular growth and replication. These results uncover a new host regulator of enteric Salmonella infections, which may be targeted to restrict bacterial load at the primary site of infection to prevent systemic spread.


Subject(s)
Cytoskeletal Proteins , Salmonella typhimurium , rho GTP-Binding Proteins , Humans , Actins/metabolism , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , Cytoskeleton/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , Salmonella typhimurium/pathogenicity , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism
15.
J Biol Chem ; 299(12): 105387, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37890783

ABSTRACT

The expression of virulence factors essential for the invasion of host cells by Salmonella enterica is tightly controlled by a network of transcription regulators. The AraC/XylS transcription factor HilD is the main integration point of environmental signals into this regulatory network, with many factors affecting HilD activity. Long-chain fatty acids, which are highly abundant throughout the host intestine, directly bind to and repress HilD, acting as environmental cues to coordinate virulence gene expression. The regulatory protein HilE also negatively regulates HilD activity, through a protein-protein interaction. Both of these regulators inhibit HilD dimerization, preventing HilD from binding to target DNA. We investigated the structural basis of these mechanisms of HilD repression. Long-chain fatty acids bind to a conserved pocket in HilD, in a comparable manner to that reported for other AraC/XylS regulators, whereas HilE forms a stable heterodimer with HilD by binding to the HilD dimerization interface. Our results highlight two distinct, mutually exclusive mechanisms by which HilD activity is repressed, which could be exploited for the development of new antivirulence leads.


Subject(s)
Bacterial Proteins , Intestines , Salmonella typhimurium , Bacterial Proteins/metabolism , Fatty Acids/metabolism , Gene Expression Regulation, Bacterial , Intestines/metabolism , Intestines/microbiology , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , Virulence , Animals , Salmonella Infections/metabolism , Salmonella Infections/microbiology
16.
Vet Microbiol ; 282: 109759, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37104940

ABSTRACT

This study presents the engineering of a less endotoxic Salmonella Typhimurium strain by manipulating the lipid-A structure of the lipopolysaccharide (LPS) component. Salmonella lipid A was dephosphorylated by using lpxE from Francisella tularensis. The 1-phosphate group from lipid-A was removed selectively, resulting in a close analog of monophosphoryl lipid A. We observed a significant impact of ∆pagL on major virulence factors such as biofilm formation, motility, persistency, and immune evasion. In correlation with biofilm and motility retardation, adhesion and invasion were elevated but with reduced intracellular survival, a favorable phenotype prospect of a vaccine strain. Western blotting and silver staining confirmed the absence of the O-antigen and truncated lipid-A core in the detoxified Salmonella mutant. In vitro and in vivo studies demonstrated that the dephosphorylated Salmonella mutant mediated lower pro-inflammatory cytokine secretion than the wild-type strain. The vaccine strains were present in the spleen and liver for five days and were cleared from the organs by day seven. However, the wild-type strain persisted in the spleen, liver, and brain, leading to sepsis-induced death. Histological evaluations of tissue samples further confirmed the reduced endotoxic activity of the detoxified Salmonella mutant. The detoxification strategy did not compromise the level of protective immunity, as the vaccine strain could enhance humoral and cellular immune responses and protect against the wild-type challenge in immunized mice.


Subject(s)
Salmonella Infections , Salmonella Vaccines , Salmonella typhimurium , Salmonella typhimurium/genetics , Salmonella typhimurium/pathogenicity , Salmonella typhimurium/physiology , Female , Animals , Mice , Mice, Inbred BALB C , Lipid A/metabolism , Salmonella Vaccines/adverse effects , Salmonella Vaccines/genetics , Salmonella Vaccines/immunology , Lipopolysaccharides/metabolism , Immunity, Humoral , Immunity, Cellular , Biofilms , Salmonella Infections/immunology , Salmonella Infections/microbiology , Carboxylic Ester Hydrolases/genetics
17.
Emerg Infect Dis ; 28(6): 1254-1256, 2022 06.
Article in English | MEDLINE | ID: mdl-35608817

ABSTRACT

Quantifying the effect of public health actions on population health is essential when justifying sustained public health investment. Using modeling, we conservatively estimated that rapid response to a multistate foodborne outbreak of Salmonella Typhimurium in the United States in 2018 potentially averted 94 reported cases and $633,181 in medical costs and productivity losses.


Subject(s)
Public Health , Salads , Salmonella Food Poisoning/epidemiology , Salmonella typhimurium , Animals , Chickens , Disease Outbreaks , Humans , Public Health/methods , Salads/adverse effects , Salads/microbiology , Salmonella Food Poisoning/economics , Salmonella Food Poisoning/etiology , Salmonella typhimurium/isolation & purification , Salmonella typhimurium/pathogenicity , United States/epidemiology
18.
Int J Mol Sci ; 23(3)2022 Jan 25.
Article in English | MEDLINE | ID: mdl-35163280

ABSTRACT

The rapid identification of bacterial antibiotic susceptibility is pivotal to the rational administration of antibacterial drugs. In this study, cefotaxime (CTX)-derived resistance in Salmonella typhimurium (abbr. CTXr-S. typhimurium) during 3 months of exposure was rapidly recorded using a portable Raman spectrometer. The molecular changes that occurred in the drug-resistant strains were sensitively monitored in whole cells by label-free surface-enhanced Raman scattering (SERS). Various degrees of resistant strains could be accurately discriminated by applying multivariate statistical analyses to bacterial SERS profiles. Minimum inhibitory concentration (MIC) values showed a positive linear correlation with the relative Raman intensities of I990/I1348, and the R2 reached 0.9962. The SERS results were consistent with the data obtained by MIC assays, mutant prevention concentration (MPC) determinations, and Kirby-Bauer antibiotic susceptibility tests (K-B tests). This preliminary proof-of-concept study indicates the high potential of the SERS method to supplement the time-consuming conventional method and help alleviate the challenges of antibiotic resistance in clinical therapy.


Subject(s)
Salmonella Infections/immunology , Salmonella typhimurium/immunology , Spectrum Analysis, Raman/methods , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Drug Resistance, Bacterial/genetics , Drug Resistance, Microbial/drug effects , Humans , Salmonella Infections/diagnosis , Salmonella typhimurium/drug effects , Salmonella typhimurium/pathogenicity
19.
PLoS Pathog ; 18(1): e1009718, 2022 01.
Article in English | MEDLINE | ID: mdl-35073381

ABSTRACT

Salmonella enterica serovar Typhimurium is a Gram-negative pathogen that uses two distinct type III secretion systems (T3SSs), termed Salmonella pathogenicity island (SPI)-1 and SPI-2, to deliver virulence factors into the host cell. The SPI-1 T3SS enables Salmonella to invade host cells, while the SPI-2 T3SS facilitates Salmonella's intracellular survival. In mice, a family of cytosolic immune sensors, including NAIP1, NAIP2, and NAIP5/6, recognizes the SPI-1 T3SS needle, inner rod, and flagellin proteins, respectively. Ligand recognition triggers assembly of the NAIP/NLRC4 inflammasome, which mediates caspase-1 activation, IL-1 family cytokine secretion, and pyroptosis of infected cells. In contrast to mice, humans encode a single NAIP that broadly recognizes all three ligands. The role of NAIP/NLRC4 or other inflammasomes during Salmonella infection of human macrophages is unclear. We find that although the NAIP/NLRC4 inflammasome is essential for detecting T3SS ligands in human macrophages, it is partially required for responses to infection, as Salmonella also activated the NLRP3 and CASP4/5 inflammasomes. Importantly, we demonstrate that combinatorial NAIP/NLRC4 and NLRP3 inflammasome activation restricts Salmonella replication in human macrophages. In contrast to SPI-1, the SPI-2 T3SS inner rod is not sensed by human or murine NAIPs, which is thought to allow Salmonella to evade host recognition and replicate intracellularly. Intriguingly, we find that human NAIP detects the SPI-2 T3SS needle protein. Critically, in the absence of both flagellin and the SPI-1 T3SS, the NAIP/NLRC4 inflammasome still controlled intracellular Salmonella burden. These findings reveal that recognition of Salmonella SPI-1 and SPI-2 T3SSs and engagement of both the NAIP/NLRC4 and NLRP3 inflammasomes control Salmonella infection in human macrophages.


Subject(s)
Inflammasomes/immunology , Macrophages/immunology , Macrophages/microbiology , Salmonella Infections/immunology , Type III Secretion Systems/immunology , CARD Signaling Adaptor Proteins/immunology , Calcium-Binding Proteins/immunology , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Neuronal Apoptosis-Inhibitory Protein/immunology , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Virulence
20.
Anal Bioanal Chem ; 414(2): 1073-1080, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34693471

ABSTRACT

In this study, Lba Cas12a (Cpf1) as one of the CRISPR systems from Lachnospiraceae bacterium was coupled with a hybridization chain reaction (HCR) to develop an electrochemical biosensor for detecting the pathogenic bacterium, Salmonella typhimurium. Autonomous cross-opening of functional DNA hairpin structures of HCR yielded polymer double-stranded DNA wires consisting of numerous single-stranded DNAs, which initiated the trans-cleavage activity of CRISPR-Cas12a to indiscriminately cleave random single-stranded DNA labeling electrochemical tags on the surface of the electrode. It led to a variation in the electron transfer of electrochemical tags. The polymer double-stranded DNA of HCR was immobilized on dynabeads (DBs) via the S. typhimurium aptamer and released from DBs. The established method could selectively and sensitively quantify S. typhimurium in samples with detection limits of 20 CFU/mL. Our study provides a novel insight for exploring universal analytical methods for pathogenic bacteria based on CRISPR-Cas12a coupled with HCR.


Subject(s)
Biosensing Techniques/methods , CRISPR-Cas Systems , Electrochemical Techniques/methods , Salmonella typhimurium/isolation & purification , Electrophoresis, Polyacrylamide Gel , Salmonella typhimurium/pathogenicity
SELECTION OF CITATIONS
SEARCH DETAIL