Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 719
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 22(2): 216-228, 2021 02.
Article in English | MEDLINE | ID: mdl-33462454

ABSTRACT

CD4+ effector lymphocytes (Teff) are traditionally classified by the cytokines they produce. To determine the states that Teff cells actually adopt in frontline tissues in vivo, we applied single-cell transcriptome and chromatin analyses to colonic Teff cells in germ-free or conventional mice or in mice after challenge with a range of phenotypically biasing microbes. Unexpected subsets were marked by the expression of the interferon (IFN) signature or myeloid-specific transcripts, but transcriptome or chromatin structure could not resolve discrete clusters fitting classic helper T cell (TH) subsets. At baseline or at different times of infection, transcripts encoding cytokines or proteins commonly used as TH markers were distributed in a polarized continuum, which was functionally validated. Clones derived from single progenitors gave rise to both IFN-γ- and interleukin (IL)-17-producing cells. Most of the transcriptional variance was tied to the infecting agent, independent of the cytokines produced, and chromatin variance primarily reflected activities of activator protein (AP)-1 and IFN-regulatory factor (IRF) transcription factor (TF) families, not the canonical subset master regulators T-bet, GATA3 or RORγ.


Subject(s)
Bacteria/pathogenicity , Bacterial Infections/microbiology , CD4-Positive T-Lymphocytes/microbiology , CD4-Positive T-Lymphocytes/parasitology , Colon/microbiology , Colon/parasitology , Gastrointestinal Microbiome , Heligmosomatoidea/pathogenicity , Intestinal Diseases, Parasitic/parasitology , Animals , Bacteria/immunology , Bacterial Infections/genetics , Bacterial Infections/immunology , Bacterial Infections/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Chromatin/genetics , Chromatin/metabolism , Citrobacter rodentium/immunology , Citrobacter rodentium/pathogenicity , Colon/immunology , Colon/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression Profiling , Heligmosomatoidea/immunology , Host-Pathogen Interactions , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Intestinal Diseases, Parasitic/genetics , Intestinal Diseases, Parasitic/immunology , Intestinal Diseases, Parasitic/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Nematospiroides dubius/immunology , Nematospiroides dubius/pathogenicity , Nippostrongylus/immunology , Nippostrongylus/pathogenicity , Phenotype , Salmonella enterica/immunology , Salmonella enterica/pathogenicity , Single-Cell Analysis , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcriptome
2.
Cell ; 175(5): 1307-1320.e22, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30392957

ABSTRACT

In the small intestine, a niche of accessory cell types supports the generation of mature epithelial cell types from intestinal stem cells (ISCs). It is unclear, however, if and how immune cells in the niche affect ISC fate or the balance between self-renewal and differentiation. Here, we use single-cell RNA sequencing (scRNA-seq) to identify MHC class II (MHCII) machinery enrichment in two subsets of Lgr5+ ISCs. We show that MHCII+ Lgr5+ ISCs are non-conventional antigen-presenting cells in co-cultures with CD4+ T helper (Th) cells. Stimulation of intestinal organoids with key Th cytokines affects Lgr5+ ISC renewal and differentiation in opposing ways: pro-inflammatory signals promote differentiation, while regulatory cells and cytokines reduce it. In vivo genetic perturbation of Th cells or MHCII expression on Lgr5+ ISCs impacts epithelial cell differentiation and IEC fate during infection. These interactions between Th cells and Lgr5+ ISCs, thus, orchestrate tissue-wide responses to external signals.


Subject(s)
Cell Differentiation , Cell Self Renewal , Interleukin-10/metabolism , Stem Cells/cytology , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Cell Differentiation/drug effects , Cell Self Renewal/drug effects , Cytokines/pharmacology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Histocompatibility Antigens Class II/metabolism , Immune System/metabolism , Intestines/cytology , Intestines/microbiology , Male , Mice , Mice, Inbred C57BL , Organoids/cytology , Organoids/drug effects , Organoids/metabolism , Receptors, G-Protein-Coupled/metabolism , Salmonella enterica/pathogenicity , Stem Cells/metabolism , T-Lymphocytes, Helper-Inducer/cytology
3.
Mol Cell ; 82(3): 629-644.e4, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35063132

ABSTRACT

The envelope of Gram-negative bacteria is a vital barrier that must balance protection and nutrient uptake. Small RNAs are crucial regulators of the envelope composition and function. Here, using RIL-seq to capture the Hfq-mediated RNA-RNA interactome in Salmonella enterica, we discover envelope-related riboregulators, including OppX. We show that OppX acts as an RNA sponge of MicF sRNA, a prototypical porin repressor. OppX originates from the 5' UTR of oppABCDF, encoding the major inner-membrane oligopeptide transporter, and sequesters MicF's seed region to derepress the synthesis of the porin OmpF. Intriguingly, OppX operates as a true sponge, storing MicF in an inactive complex without affecting its levels or stability. Conservation of the opp-OppX-MicF-ompF axis in related bacteria suggests that it serves an important mechanism, adjusting envelope porosity to specific transport capacity. These data also highlight the resource value of this Salmonella RNA interactome, which will aid in unraveling RNA-centric regulation in enteric pathogens.


Subject(s)
5' Untranslated Regions , Cell Membrane/genetics , Escherichia coli Proteins/genetics , Host Factor 1 Protein/genetics , RNA, Bacterial/genetics , Salmonella enterica/genetics , Biological Transport , Cell Membrane/metabolism , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial , Host Factor 1 Protein/metabolism , Host-Pathogen Interactions , Permeability , Porins/genetics , Porins/metabolism , RNA, Bacterial/metabolism , RNA-Seq , Salmonella enterica/metabolism , Salmonella enterica/pathogenicity
4.
Int J Mol Sci ; 25(8)2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38673776

ABSTRACT

Salmonella enterica is a leading cause of bacterial food-borne illness in humans and is responsible for millions of cases annually. A critical strategy for the survival of this pathogen is the translocation of bacterial virulence factors termed effectors into host cells, which primarily function via protein-protein interactions with host proteins. The Salmonella genome encodes several paralogous effectors believed to have arisen from duplication events throughout the course of evolution. These paralogs can share structural similarities and enzymatic activities but have also demonstrated divergence in host cell targets or interaction partners and contributions to the intracellular lifecycle of Salmonella. The paralog effectors SopD and SopD2 share 63% amino acid sequence similarity and extensive structural homology yet have demonstrated divergence in secretion kinetics, intracellular localization, host targets, and roles in infection. SopD and SopD2 target host Rab GTPases, which represent critical regulators of intracellular trafficking that mediate diverse cellular functions. While SopD and SopD2 both manipulate Rab function, these paralogs display differences in Rab specificity, and the effectors have also evolved multiple mechanisms of action for GTPase manipulation. Here, we highlight this intriguing pair of paralog effectors in the context of host-pathogen interactions and discuss how this research has presented valuable insights into effector evolution.


Subject(s)
Bacterial Proteins , Host-Pathogen Interactions , Salmonella Infections , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Humans , Host-Pathogen Interactions/genetics , Salmonella Infections/microbiology , Salmonella Infections/metabolism , Salmonella enterica/metabolism , Salmonella enterica/genetics , Salmonella enterica/pathogenicity , Virulence Factors/metabolism , Virulence Factors/genetics , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Animals , Evolution, Molecular
5.
Int J Mol Sci ; 25(11)2024 May 23.
Article in English | MEDLINE | ID: mdl-38891852

ABSTRACT

Salmonella diarizonae (IIIb) is frequently isolated from reptiles and less frequently from birds and mammals. However, its isolation from invasive human infections has not been widely reported. Migratory mallard ducks are excellent bioindicators of pathogen presence and pathogen antibiotic resistance (AMR). We present the first isolation from a mallard duck in central Europe of the antibiotic-resistant Salmonella enterica subsp. diarizonae with the unique antigenic pattern 58:r:z53 and report its whole-genome sequencing, serosequencing, and genotyping, which enabled the prediction of its pathogenicity and comparison with phenotypic AMR. The isolated strain was highly similar to S. diarizonae isolated from humans and food. Twenty-four AMR genes were detected, including those encoding aminoglycoside, fluoroquinolone, macrolide, carbapenem, tetracycline, cephalosporin, nitroimidazole, peptide antibiotic, and disinfecting agent/antiseptic resistance. Six Salmonella pathogenicity islands were found (SPI-1, SPI-2, SPI-3, SPI-5, SPI-9, and SPI-13). An iron transport system was detected in SPI-1 centisome C63PI. Plasmid profile analyses showed three to be present. Sequence mutations in the invA and invF genes were noted, which truncated and elongated the proteins, respectively. The strain also harbored genes encoding type-III secretion-system effector proteins and many virulence factors found in S. diarizonae associated with human infections. This study aims to elucidate the AMR and virulence genes in S. enterica subsp. diarizonae that may most seriously threaten human health.


Subject(s)
Ducks , Animals , Ducks/microbiology , Humans , Salmonella/genetics , Salmonella/pathogenicity , Salmonella/isolation & purification , Salmonella/drug effects , Whole Genome Sequencing , Genomic Islands/genetics , Salmonella Infections, Animal/microbiology , Anti-Bacterial Agents/pharmacology , Salmonella enterica/genetics , Salmonella enterica/pathogenicity , Salmonella enterica/isolation & purification , Salmonella enterica/drug effects , Drug Resistance, Multiple, Bacterial/genetics , Genome, Bacterial , Phylogeny , Drug Resistance, Bacterial/genetics , Plasmids/genetics
6.
Cell Immunol ; 383: 104651, 2023 01.
Article in English | MEDLINE | ID: mdl-36493524

ABSTRACT

Lipopolysaccharides (LPS) is one of the most potent pathogen-associated signals for the immune system of vertebrates. In addition to the canonical pathway of LPS detection mediated by toll-like receptor 4 (TLR4) signaling pathway, TRP channel-mediated pathways endow sensory neurons and epithelial cells with the ability to detect and react to bacterial endotoxins. Previous work revealed that LPS triggers TRPV4-dependent calcium influx in urothelial cells (UCs) and mouse tracheobronchial epithelial cells (mTEC). In marked contrast, here we show that most subtypes of LPS could not directly activate TRPV4 channel. Although LPS from Salmonella enterica serotype Minnesota evoked a [Ca2+]i response in freshly isolated human bronchial epithelial cells (ECs), freshly isolated mouse ear skin single-cell suspensions, or HEK293T cells transiently transfected with mTRPV4, this activation occurred in a TRPV4-independent manner. Additionally, LPS from either E. coli strains or Salmonella enterica serotype Minnesota did not evoke significant difference in inflammation and pain hyperalgesia between wild type and TRPV4 deficient mice. In summary, our results demonstrate that in vitro and in vivo effects induced by LPS are independent of TRPV4, thus providing a clarity to the questioned role of LPS in TRPV4 activation.


Subject(s)
Calcium Signaling , Lipopolysaccharides , TRPV Cation Channels , Animals , Humans , Mice , Calcium/metabolism , Calcium Signaling/physiology , Escherichia coli/pathogenicity , HEK293 Cells , Lipopolysaccharides/pharmacology , Lipopolysaccharides/metabolism , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , TRPV Cation Channels/pharmacology , Salmonella enterica/pathogenicity
7.
Nucleic Acids Res ; 49(9): 5319-5335, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33939833

ABSTRACT

FinO-domain proteins represent an emerging family of RNA-binding proteins (RBPs) with diverse roles in bacterial post-transcriptional control and physiology. They exhibit an intriguing targeting spectrum, ranging from an assumed single RNA pair (FinP/traJ) for the plasmid-encoded FinO protein, to transcriptome-wide activity as documented for chromosomally encoded ProQ proteins. Thus, the shared FinO domain might bear an unusual plasticity enabling it to act either selectively or promiscuously on the same cellular RNA pool. One caveat to this model is that the full suite of in vivo targets of the assumedly highly selective FinO protein is unknown. Here, we have extensively profiled cellular transcripts associated with the virulence plasmid-encoded FinO in Salmonella enterica. While our analysis confirms the FinP sRNA of plasmid pSLT as the primary FinO target, we identify a second major ligand: the RepX sRNA of the unrelated antibiotic resistance plasmid pRSF1010. FinP and RepX are strikingly similar in length and structure, but not in primary sequence, and so may provide clues to understanding the high selectivity of FinO-RNA interactions. Moreover, we observe that the FinO RBP encoded on the Salmonella virulence plasmid controls the replication of a cohabitating antibiotic resistance plasmid, suggesting cross-regulation of plasmids on the RNA level.


Subject(s)
Bacterial Proteins/metabolism , Plasmids/genetics , RNA, Small Untranslated/metabolism , RNA-Binding Proteins/metabolism , Salmonella enterica/genetics , Ligands , RNA, Antisense/metabolism , RNA, Small Untranslated/chemistry , Salmonella enterica/metabolism , Salmonella enterica/pathogenicity , Virulence
8.
Annu Rev Genet ; 48: 361-82, 2014.
Article in English | MEDLINE | ID: mdl-25251855

ABSTRACT

The gastrointestinal (GI) microbiota is a complex community of microorganisms residing within the mammalian gastrointestinal tract. The GI microbiota is vital to the development of the host immune system and plays a crucial role in human health and disease. The composition of the GI microbiota differs immensely among individuals yet specific shifts in composition and diversity have been linked to inflammatory bowel disease, obesity, atopy, and susceptibility to infection. In this review, we describe the GI microbiota and its role in enteric diseases caused by pathogenic Escherichia coli, Salmonella enterica, and Clostridium difficile. We discuss the central role of the GI microbiota in protective immunity, resistance to enteric pathogens, and resolution of enteric colitis.


Subject(s)
Colitis/genetics , Gastrointestinal Tract/microbiology , Microbiota/genetics , Animals , Clostridioides difficile/immunology , Clostridioides difficile/pathogenicity , Colitis/immunology , Colitis/microbiology , Escherichia coli/immunology , Escherichia coli/pathogenicity , Gastrointestinal Tract/immunology , Humans , Microbiota/immunology , Salmonella enterica/immunology , Salmonella enterica/pathogenicity
9.
PLoS Pathog ; 16(2): e1008336, 2020 02.
Article in English | MEDLINE | ID: mdl-32084237

ABSTRACT

Typhoid toxin is an A2B5 toxin secreted from Salmonella Typhi-infected cells during human infection and is suggested to contribute to typhoid disease progression and the establishment of chronic infection. To deliver the enzymatic 'A' subunits of the toxin to the site of action in host cells, the receptor-binding 'B' subunit PltB binds to the trisaccharide glycan receptor moieties terminated in N-acetylneuraminic acid (Neu5Ac) that is α2-3 or α2-6 linked to the underlying disaccharide, galactose (Gal) and N-acetylglucosamine (GlcNAc). Neu5Ac is present in both unmodified and modified forms, with 9-O-acetylated Neu5Ac being the most common modification in humans. Here we show that host cells associated with typhoid toxin-mediated clinical signs express both unmodified and 9-O-acetylated glycan receptor moieties. We found that PltB binds to 9-O-acetylated α2-3 glycan receptor moieties with a markedly increased affinity, while the binding affinity to 9-O-acetylated α2-6 glycans is only slightly higher, as compared to the affinities of PltB to the unmodified counterparts, respectively. We also present X-ray co-crystal structures of PltB bound to related glycan moieties, which supports the different effects of 9-O-acetylated α2-3 and α2-6 glycan receptor moieties on the toxin binding. Lastly, we demonstrate that the cells exclusively expressing unmodified glycan receptor moieties are less susceptible to typhoid toxin than the cells expressing 9-O-acetylated counterparts, although typhoid toxin intoxicates both cells. These results reveal a fine-tuning mechanism of a bacterial toxin that exploits specific chemical modifications of its glycan receptor moieties for virulence and provide useful insights into the development of therapeutics against typhoid fever.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Proteins/ultrastructure , Bacterial Toxins/metabolism , Salmonella typhi/metabolism , Acetylation , Animals , Cell Line , Humans , Mice , Mice, Knockout , N-Acetylneuraminic Acid/metabolism , Polysaccharides/metabolism , Protein Binding , Salmonella enterica/metabolism , Salmonella enterica/pathogenicity , Salmonella typhi/pathogenicity , Trisaccharides/metabolism , Typhoid Fever/microbiology , Virulence
10.
Cell Microbiol ; 23(4): e13315, 2021 04.
Article in English | MEDLINE | ID: mdl-33534187

ABSTRACT

Salmonella enterica serovars infect a broad range of mammalian hosts including humans, causing both gastrointestinal and systemic diseases. Following uptake into host cells, bacteria replicate within vacuoles (Salmonella-containing vacuoles; SCVs). Clusters of SCVs are frequently associated with a meshwork of F-actin. This meshwork is dependent on the Salmonella pathogenicity island 2 encoded type III secretion system and its effector SteC. SteC contains a region with weak similarity to conserved subdomains of eukaryotic kinases and has kinase activity that is required for the formation of the F-actin meshwork. Several substrates of SteC have been identified. In this mini-review, we attempt to integrate these findings and propose a more unified model to explain SCV-associated F-actin: SteC (i) phosphorylates the actin sequestering protein Hsp27, which increases the local G-actin concentration (ii) binds to and phosphorylates formin family FMNL proteins, which enables actin polymerisation and (iii) phosphorylates MEK, resulting in activation of the MEK/ERK/MLCK/Myosin II pathway, leading to F-actin bundling. We also consider the possible physiological functions of SCV-associated F-actin and similar structures produced by other intracellular bacterial pathogens.


Subject(s)
Actins/metabolism , Host-Pathogen Interactions , Salmonella enterica/pathogenicity , Shiga-Toxigenic Escherichia coli/metabolism , Actin Cytoskeleton , Actins/genetics , Animals , Epithelial Cells/microbiology , Genomic Islands , Humans , Mice , Phosphorylation , Vacuoles
11.
Nature ; 540(7632): 280-283, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27798599

ABSTRACT

The Enterobacteriaceae are a family of Gram-negative bacteria that include commensal organisms as well as primary and opportunistic pathogens that are among the leading causes of morbidity and mortality worldwide. Although Enterobacteriaceae often comprise less than 1% of a healthy intestine's microbiota, some of these organisms can bloom in the inflamed gut; expansion of enterobacteria is a hallmark of microbial imbalance known as dysbiosis. Microcins are small secreted proteins that possess antimicrobial activity in vitro, but whose role in vivo has been unclear. Here we demonstrate that microcins enable the probiotic bacterium Escherichia coli Nissle 1917 (EcN) to limit the expansion of competing Enterobacteriaceae (including pathogens and pathobionts) during intestinal inflammation. Microcin-producing EcN limits the growth of competitors in the inflamed intestine, including commensal E. coli, adherent-invasive E. coli and the related pathogen Salmonella enterica. Moreover, only therapeutic administration of the wild-type, microcin-producing EcN to mice previously infected with S. enterica substantially reduced intestinal colonization by the pathogen. Our work provides the first evidence that microcins mediate inter- and intraspecies competition among the Enterobacteriaceae in the inflamed gut. Moreover, we show that microcins can act as narrow-spectrum therapeutics to inhibit enteric pathogens and reduce enterobacterial blooms.


Subject(s)
Bacteriocins/metabolism , Enterobacteriaceae/growth & development , Escherichia coli/metabolism , Inflammation/microbiology , Inflammation/pathology , Intestines/microbiology , Intestines/pathology , Animals , Bacteriocins/genetics , Bacteriocins/therapeutic use , Dysbiosis/microbiology , Enterobacteriaceae/pathogenicity , Escherichia coli/classification , Escherichia coli/growth & development , Female , Inflammation/metabolism , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Probiotics/metabolism , Salmonella enterica/growth & development , Salmonella enterica/pathogenicity , Symbiosis
12.
PLoS Pathog ; 15(2): e1007566, 2019 02.
Article in English | MEDLINE | ID: mdl-30716138

ABSTRACT

The cellular invasion machinery of the enteric pathogen Salmonella consists of a type III secretion system (T3SS) with injectable virulence factors that induce uptake by macropinocytosis. Salmonella invasion at the apical surface of intestinal epithelial cells is inefficient, presumably because of a glycosylated barrier formed by transmembrane mucins that prevents T3SS contact with host cells. We observed that Salmonella is capable of apical invasion of intestinal epithelial cells that express the transmembrane mucin MUC1. Knockout of MUC1 in HT29-MTX cells or removal of MUC1 sialic acids by neuraminidase treatment reduced Salmonella apical invasion but did not affect lateral invasion that is not hampered by a defensive barrier. A Salmonella deletion strain lacking the SiiE giant adhesin was unable to invade intestinal epithelial cells through MUC1. SiiE-positive Salmonella closely associated with the MUC1 layer at the apical surface, but invaded Salmonella were negative for the adhesin. Our findings uncover that the transmembrane mucin MUC1 is required for Salmonella SiiE-mediated entry of enterocytes via the apical route.


Subject(s)
Adhesins, Bacterial/metabolism , Mucin-1/physiology , Salmonella Infections/metabolism , Bacterial Proteins , Cell Line , Elongin/metabolism , Enterocytes , Epithelial Cells , Humans , Mucin-1/genetics , Mucin-1/metabolism , Salmonella enterica/pathogenicity , Salmonella typhimurium/pathogenicity , Virulence Factors
13.
Appl Environ Microbiol ; 87(24): e0115121, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34613760

ABSTRACT

Mitigation strategies to prevent microbial contamination of crops are lacking. We tested the hypothesis that induction of plant systemic resistance by biological (induced systemic resistance [ISR]) and chemical (systemic acquired resistance [SAR]) elicitors reduces endophytic colonization of leaves by Salmonella enterica serovars Senftenberg and Typhimurium. S. Senftenberg had greater endophytic fitness than S. Typhimurium in basil and lettuce. The apoplastic population sizes of serovars Senftenberg and Typhimurium in basil and lettuce, respectively, were significantly reduced approximately 10- to 100-fold by root treatment with microbial inducers of systemic resistance compared to H2O treatment. Rhodotorula glutinis effected the lowest population increases of S. Typhimurium in lettuce and S. Senftenberg in basil leaves, respectively 120- and 60-fold lower than those seen with the H2O treatment over 10 days postinoculation. Trichoderma harzianum and Pichia guilliermondii did not have any significant effect on S. Senftenberg in the basil apoplast. The chemical elicitors acidobenzolar-S-methyl and dl-ß-amino-butyric acid inhibited S. Typhimurium multiplication in the lettuce apoplast 10- and 2-fold, respectively, compared to H2O-treated plants. All ISR and SAR inducers applied to lettuce roots in this study increased leaf expression of the defense gene PR1, as did Salmonella apoplastic colonization in H2O-treated lettuce plants. Remarkably, both acidobenzolar-S-methyl upregulation and R. glutinis upregulation of PR1 were repressed by the presence of Salmonella in the leaves. However, enhanced PR1 expression was sustained longer and at greater levels upon elicitor treatment than by Salmonella induction alone. These results serve as a proof of concept that priming of plant immunity may provide an intrinsic hurdle against the endophytic establishment of enteric pathogens in leafy vegetables. IMPORTANCE Fruit and vegetables consumed raw have become an important vehicle of foodborne illness despite a continuous effort to improve their microbial safety. Salmonella enterica has caused numerous recalls and outbreaks of infection associated with contaminated leafy vegetables. Evidence is increasing that enteric pathogens can reach the leaf apoplast, where they confront plant innate immunity. Plants may be triggered for induction of their defense signaling pathways by exposure to chemical or microbial elicitors. This priming for recognition of microbes by plant defense pathways has been used to inhibit plant pathogens and limit disease. Given that current mitigation strategies are insufficient in preventing microbial contamination of produce and associated outbreaks, we investigated the effect of plant-induced resistance on S. enterica colonization of the lettuce and basil leaf apoplast in order to gain a proof of concept for the use of such an intrinsic approach to inhibit human pathogens in leafy vegetables.


Subject(s)
Disease Resistance , Lactuca , Ocimum basilicum , Salmonella enterica , Lactuca/immunology , Lactuca/microbiology , Ocimum basilicum/immunology , Ocimum basilicum/microbiology , Plant Immunity , Plant Leaves , Plant Proteins , Salmonella enterica/pathogenicity , Salmonella typhimurium
14.
Crit Rev Microbiol ; 47(4): 397-434, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33751923

ABSTRACT

Adaptation to environments with constant fluctuations imposes challenges that are only overcome with sophisticated strategies that allow bacteria to perceive environmental conditions and develop an appropriate response. The gastrointestinal environment is a complex ecosystem that is home to trillions of microorganisms. Termed microbiota, this microbial ensemble plays important roles in host health and provides colonization resistance against pathogens, although pathogens have evolved strategies to circumvent this barrier. Among the strategies used by bacteria to monitor their environment, one of the most important are the sensing and signalling machineries of two-component systems (TCSs), which play relevant roles in the behaviour of all bacteria. Salmonella enterica is no exception, and here we present our current understanding of how this important human pathogen uses TCSs as an integral part of its lifestyle. We describe important aspects of these systems, such as the stimuli and responses involved, the processes regulated, and their roles in virulence. We also dissect the genomic organization of histidine kinases and response regulators, as well as the input and output domains for each TCS. Lastly, we explore how these systems may be promising targets for the development of antivirulence therapeutics to combat antibiotic-resistant infections.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Salmonella Infections/microbiology , Salmonella enterica/metabolism , Salmonella enterica/pathogenicity , Animals , Bacterial Proteins/genetics , Ecosystem , Humans , Salmonella enterica/genetics , Signal Transduction , Virulence
15.
Cell Microbiol ; 22(5): e13155, 2020 05.
Article in English | MEDLINE | ID: mdl-31872936

ABSTRACT

Salmonella enterica serovar Typhimurium (STM) is an invasive, facultative intracellular pathogen that has evolved sophisticated molecular mechanisms to establish an intracellular niche within a specialised vesicular compartment, the Salmonella-containing vacuole (SCV). The loss of the SCV and release of STM into the cytosol of infected host cells was observed, and a bimodal intracellular lifestyle of STM in the SCV versus life in the cytosol is currently discussed. We set out to investigate the parameters affecting SCV integrity and cytosolic release. A fluorescent protein-based cytosolic reporter approach was established to quantify, time-resolved, and on a single cell level, the release of STM into the cytosol of host cells. We observed that the extent of SCV damage and cytosolic release is highly dependent on experimental conditions such as multiplicity of infection, type of host cell line, and STM strain background. Trigger invasion mediated by the Salmonella Pathogenicity Island 1-encoded type III secretion system (SPI1-T3SS) and its effector proteins promoted cytosolic release, whereas cytosolic bacteria were rarely observed if entry was mediated by zipper invasion. Presence of SPI1-T3SS effector SopE was identified as major factor for damage of the SCV in the early phase after STM invasion and sopE-expressing strains showed higher levels of cytosolic release.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Salmonella enterica/genetics , Salmonella enterica/metabolism , Vacuoles/microbiology , Animals , Cytosol/metabolism , Genomic Islands/genetics , HeLa Cells , Host-Pathogen Interactions/physiology , Humans , Kinetics , Mice , RAW 264.7 Cells , Salmonella enterica/pathogenicity , Salmonella typhimurium , Type III Secretion Systems
16.
J Appl Microbiol ; 130(6): 2123-2131, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33150646

ABSTRACT

AIMS: This study compared the capacity of strains of Salmonella enterica serovars Enteritidis and Dublin isolated in Brazil to invade epithelial cells, to be internalized by and survive within macrophages, and to stimulate cytokine release in vitro. METHODS AND RESULTS: Both serovars infected 75 and 73% Caco-2 (human) and MDBK (bovine) epithelial cells respectively. Salmonella Dublin and S. Enteritidis (i) were internalized at the respective rates of 79·6 and 65·0% (P ≤ 0·05) by U937 (human) macrophages, and 70·4 and 66·9% by HD11 (chicken) macrophages; and (ii) multiplied at the respective rates of 3·2- and 2·7-fold within U937 cells, and 1·9- and 1·1-fold (P ≤ 0·05) within HD11 cells respectively. Seventy per cent of 10 S. Dublin strains stimulated IL-8 production, while 70% of S. Enteritidis strains enhanced production of IL-1ß, IL-6, IL-8, IL-10, IL-12p70 and TNF in Caco-2 cells. CONCLUSIONS: Compared with S. Enteritidis, S. Dublin had stronger ability to survive within macrophages and induced weak cytokine production, which may explain the higher incidence of invasive diseases caused by S. Dublin in humans. SIGNIFICANCE AND IMPACT OF THE STUDY: This study compared S. enterica serovars Enteritidis and Dublin to provide comparative data about the profile of the two serovars in cells from humans, the common host and their respective natural animal hosts and vice versa in order to check the differences between these two phylogenetically closely related serovars that share antigenic properties but present different phenotypic behaviours.


Subject(s)
Cytokines/metabolism , Epithelial Cells/microbiology , Macrophages/microbiology , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella enterica/immunology , Salmonella enterica/pathogenicity , Animals , Brazil , Caco-2 Cells , Cattle , Chickens , Epithelial Cells/immunology , Humans , Macrophages/immunology , Microbial Viability , Serogroup , U937 Cells
17.
Lett Appl Microbiol ; 73(1): 54-63, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33765334

ABSTRACT

Lactic Acid Bacteria (LAB) regulate and maintain the stability of healthy microbial flora, inhibit the adhesion of pathogenic bacteria and promote the colonization of beneficial micro-organisms. The drug resistance and pathogenicity of Salmonella enteritis SE47 isolated from retail eggs were investigated. Meanwhile, Enterococcus faecalis L76 and Lactobacillus salivarius LAB35 were isolated from intestine of chicken. With SE47 as indicator bacteria, the diameters of L76 and LAB35 inhibition zones were 12 mm and 8·5 mm, respectively, by agar inhibition circle method, which indicated that both of them had inhibitory effect on Salmonella, and L76 had better antibacterial effect; two chicken-derived lactic acid bacteria isolates and Salmonella SE47 were incubated with Caco-2. The adhesion index of L76 was 17·5%, which was much higher than that of LAB35 (10·21%) and SE47 (4·89%), this experiment shows that the higher the bacteriostatic effect of potential probiotics, the stronger the adhesion ability; then Caco-2 cells were incubated with different bacteria, and the survival of Caco-2 cells was observed by flow cytometry. Compared with Salmonella SE47, the results showed that lactic acid bacteria isolates could effectively protect Caco-2 cells; finally, after different bacteria incubated Caco-2 cells, according to the cytokine detection kit, the RNA of Caco-2 cells was extracted and transcribed into cDNA, then detected by fluorescence quantitative PCR, the results showed that L76 could protect Caco-2 cells from the invasion of Salmonella SE47, with less cell membrane rupture and lower expression of MIF and TNF genes. Therefore, the lactic acid bacteria isolates can effectively inhibit the adhesion of Salmonella and protect the integrity of intestinal barrier.


Subject(s)
Antibiosis/physiology , Eggs/microbiology , Lactobacillales/physiology , Salmonella Infections/microbiology , Salmonella enterica/physiology , Animals , Caco-2 Cells , Chickens/microbiology , Drug Resistance, Bacterial/physiology , Enterococcus faecalis/isolation & purification , Enterococcus faecalis/physiology , Humans , Ligilactobacillus salivarius/isolation & purification , Ligilactobacillus salivarius/physiology , Probiotics/isolation & purification , Probiotics/pharmacology , Salmonella enterica/pathogenicity
18.
PLoS Genet ; 14(5): e1007333, 2018 05.
Article in English | MEDLINE | ID: mdl-29738521

ABSTRACT

Emerging pathogens are a major threat to public health, however understanding how pathogens adapt to new niches remains a challenge. New methods are urgently required to provide functional insights into pathogens from the massive genomic data sets now being generated from routine pathogen surveillance for epidemiological purposes. Here, we measure the burden of atypical mutations in protein coding genes across independently evolved Salmonella enterica lineages, and use these as input to train a random forest classifier to identify strains associated with extraintestinal disease. Members of the species fall along a continuum, from pathovars which cause gastrointestinal infection and low mortality, associated with a broad host-range, to those that cause invasive infection and high mortality, associated with a narrowed host range. Our random forest classifier learned to perfectly discriminate long-established gastrointestinal and invasive serovars of Salmonella. Additionally, it was able to discriminate recently emerged Salmonella Enteritidis and Typhimurium lineages associated with invasive disease in immunocompromised populations in sub-Saharan Africa, and within-host adaptation to invasive infection. We dissect the architecture of the model to identify the genes that were most informative of phenotype, revealing a common theme of degradation of metabolic pathways in extraintestinal lineages. This approach accurately identifies patterns of gene degradation and diversifying selection specific to invasive serovars that have been captured by more labour-intensive investigations, but can be readily scaled to larger analyses.


Subject(s)
Adaptation, Physiological/genetics , Bacterial Proteins/genetics , Machine Learning , Salmonella enterica/genetics , Animals , Host Specificity , Humans , Mutation , Phylogeny , Salmonella Infections/microbiology , Salmonella Infections, Animal/microbiology , Salmonella enterica/classification , Salmonella enterica/pathogenicity , Virulence/genetics
19.
Genomics ; 112(6): 4863-4874, 2020 11.
Article in English | MEDLINE | ID: mdl-32898642

ABSTRACT

The G-quadruplex structure is a highly conserved drug target for preventing infection of several human pathogens. We tried to explore G-quadruplex forming motifs as promising drug targets in the genome of Salmonella enterica that causes enteric fever in humans. Herein, we report three highly conserved G-quadruplex motifs (SE-PGQ-1, 2, and 3) in the genome of Salmonella enterica. Bioinformatics analysis inferred the presence of SE-PGQ-1 in the regulatory region of mgtA, SE-PGQ-2 in ORF of entA, and SE-PGQ-3 in the promoter region of malE and malK genes. The G-quadruplex forming sequences were confirmed by biophysical and biomolecular techniques. Cellular studies affirm the inhibitory effect of G-quadruplex specific ligands on Salmonella enterica growth. Further, PCR inhibition, reporter based assay, and RT-qPCR assays emphasize the biological relevance of G-quadruplexes in these genes. Thus, this study confirmed the presence of G-quadruplex motifs in Salmonella enterica and characterized them as a promising drug target.


Subject(s)
G-Quadruplexes , Maltose-Binding Proteins/genetics , Salmonella enterica/genetics , Virulence/genetics , ATP-Binding Cassette Transporters/genetics , Bacterial Proteins/genetics , Ion Transport , Promoter Regions, Genetic , Salmonella enterica/pathogenicity
20.
Clin Microbiol Rev ; 32(1)2019 01.
Article in English | MEDLINE | ID: mdl-30487167

ABSTRACT

The ability of pathogenic bacteria to affect higher organisms and cause disease is one of the most dramatic properties of microorganisms. Some pathogens can establish transient colonization only, but others are capable of infecting their host for many years or even for a lifetime. Long-term infection is called persistence, and this phenotype is fundamental for the biology of important human pathogens, including Helicobacter pylori, Mycobacterium tuberculosis, and Salmonella enterica Both typhoidal and nontyphoidal serovars of the species Salmonella enterica can cause persistent infection in humans; however, as these two Salmonella groups cause clinically distinct diseases, the characteristics of their persistent infections in humans differ significantly. Here, following a general summary of Salmonella pathogenicity, host specificity, epidemiology, and laboratory diagnosis, I review the current knowledge about Salmonella persistence and discuss the relevant epidemiology of persistence (including carrier rate, duration of shedding, and host and pathogen risk factors), the host response to Salmonella persistence, Salmonella genes involved in this lifestyle, as well as genetic and phenotypic changes acquired during prolonged infection within the host. Additionally, I highlight differences between the persistence of typhoidal and nontyphoidal Salmonella strains in humans and summarize the current gaps and limitations in our understanding, diagnosis, and curing of persistent Salmonella infections.


Subject(s)
Salmonella Infections/microbiology , Salmonella enterica/pathogenicity , Carrier State , Humans , Risk Factors , Salmonella Infections/diagnosis , Salmonella Infections/drug therapy , Salmonella Infections/pathology , Salmonella enterica/classification , Serogroup
SELECTION OF CITATIONS
SEARCH DETAIL