Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 390
Filter
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.
Immunity ; 49(6): 1090-1102.e7, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30552021

ABSTRACT

Salmonella enterica (Se) bacteria cause persistent intracellular infections while stimulating a robust interferon-γ-producing CD4+ T (Th1) cell response. We addressed this paradox of concomitant infection and immunity by tracking fluorescent Se organisms in mice. Se bacteria persisted in nitric oxide synthase (iNOS)-producing resident and recruited macrophages while inducing genes related to protection from nitric oxide. Se-infected cells occupied iNOS+ splenic granulomas that excluded T cells but were surrounded by mononuclear phagocytes producing the chemokines CXCL9 and CXCL10, and Se epitope-specific Th1 cells expressing CXCR3, the receptor for these chemokines. Blockade of CXCR3 inhibited Th1 occupancy of CXCL9/10-dense regions, reduced activation of the Th1 cells, and led to increased Se growth. Thus, intracellular Se bacteria survive in their hosts by counteracting toxic products of the innate immune response and by residing in T cell-sparse granulomas, away from abundant Th1 cells positioned via CXCR3 in a bordering region that act to limit infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Granuloma/immunology , Receptors, CXCR3/immunology , Salmonella Infections/immunology , Salmonella enterica/immunology , Th1 Cells/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/microbiology , Chemokine CXCL10/immunology , Chemokine CXCL10/metabolism , Chemokine CXCL9/immunology , Chemokine CXCL9/metabolism , Granuloma/metabolism , Granuloma/microbiology , Host-Pathogen Interactions/immunology , Ligands , Macrophage Activation/immunology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Receptors, CXCR3/metabolism , Salmonella Infections/metabolism , Salmonella Infections/microbiology , Salmonella enterica/physiology , Th1 Cells/metabolism , Th1 Cells/microbiology
3.
Immunity ; 44(6): 1392-405, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27287411

ABSTRACT

Although numerous polymorphisms have been associated with inflammatory bowel disease (IBD), identifying the function of these genetic factors has proved challenging. Here we identified a role for nine genes in IBD susceptibility loci in antibacterial autophagy and characterized a role for one of these genes, GPR65, in maintaining lysosome function. Mice lacking Gpr65, a proton-sensing G protein-coupled receptor, showed increased susceptibly to bacteria-induced colitis. Epithelial cells and macrophages lacking GPR65 exhibited impaired clearance of intracellular bacteria and accumulation of aberrant lysosomes. Similarly, IBD patient cells and epithelial cells expressing an IBD-associated missense variant, GPR65 I231L, displayed aberrant lysosomal pH resulting in lysosomal dysfunction, impaired bacterial restriction, and altered lipid droplet formation. The GPR65 I231L polymorphism was sufficient to confer decreased GPR65 signaling. Collectively, these data establish a role for GPR65 in IBD susceptibility and identify lysosomal dysfunction as a potentially causative element in IBD pathogenesis with effects on cellular homeostasis and defense.


Subject(s)
Colitis/immunology , Epithelial Cells/immunology , Inflammatory Bowel Diseases/genetics , Lysosomes/physiology , Receptors, G-Protein-Coupled/metabolism , Salmonella Infections/immunology , Salmonella enterica/immunology , Salmonella typhimurium/immunology , Animals , Genetic Predisposition to Disease , HeLa Cells , Humans , Inflammatory Bowel Diseases/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagosomes/physiology , Polymorphism, Genetic , Receptors, G-Protein-Coupled/genetics , Risk
4.
PLoS Pathog ; 18(1): e1010241, 2022 01.
Article in English | MEDLINE | ID: mdl-35077524

ABSTRACT

Salmonella enterica causes intracellular infections that can be limited to the intestine or spread to deeper tissues. In most cases, intracellular bacteria show moderate growth. How these bacteria face host defenses that recognize peptidoglycan, is poorly understood. Here, we report a high-resolution structural analysis of the minute amounts of peptidoglycan purified from S. enterica serovar Typhimurium (S. Typhimurium) infecting fibroblasts, a cell type in which this pathogen undergoes moderate growth and persists for days intracellularly. The peptidoglycan of these non-proliferating bacteria contains atypical crosslinked muropeptides with stem peptides trimmed at the L-alanine-D-glutamic acid-(γ) or D-glutamic acid-(γ)-meso-diaminopimelic acid motifs, both sensed by intracellular immune receptors. This peptidoglycan has a reduced glycan chain average length and ~30% increase in the L,D-crosslink, a type of bridge shared by all the atypical crosslinked muropeptides identified. The L,D-transpeptidases LdtD (YcbB) and LdtE (YnhG) are responsible for the formation of these L,D-bridges in the peptidoglycan of intracellular bacteria. We also identified in a fraction of muropeptides an unprecedented modification in the peptidoglycan of intracellular S. Typhimurium consisting of the amino alcohol alaninol replacing the terminal (fourth) D-alanine. Alaninol was still detectable in the peptidoglycan of a double mutant lacking LdtD and LdtE, thereby ruling out the contribution of these enzymes to this chemical modification. Remarkably, all multiple mutants tested lacking candidate enzymes that either trim stem peptides or form the L,D-bridges retain the capacity to modify the terminal D-alanine to alaninol and all attenuate NF-κB nuclear translocation. These data inferred a potential role of alaninol-containing muropeptides in attenuating pro-inflammatory signaling, which was confirmed with a synthetic tetrapeptide bearing such amino alcohol. We suggest that the modification of D-alanine to alaninol in the peptidoglycan of non-proliferating intracellular S. Typhimurium is an editing process exploited by this pathogen to evade immune recognition inside host cells.


Subject(s)
Peptidoglycan/chemistry , Peptidoglycan/immunology , Salmonella Infections/immunology , Salmonella enterica/immunology , Salmonella enterica/metabolism , Cell Line , Cell Wall/chemistry , Cell Wall/immunology , Cell Wall/metabolism , Humans , Immune Tolerance/immunology , Peptidoglycan/metabolism
5.
Am J Hum Biol ; 36(6): e24061, 2024 06.
Article in English | MEDLINE | ID: mdl-38429916

ABSTRACT

OBJECTIVES: The immune system of milk (ISOM) creates a mother-infant immune axis that plays an important role in protecting infants against infectious disease (ID). Tradeoffs in the immune system suggest the potential for both protection and harm, so we conceive of two dimensions via which the ISOM impacts infants: promotion of protective activity and control of activity directed at benign targets. High variability in ISOM activity across mother-infant dyads suggests investment the ISOM may have evolved to be sensitive to maternal and/or infant characteristics. We assessed predictors of appropriate and misdirected proinflammatory ISOM activity in an environment of high ID risk, testing predictions drawn from life history theory and other evolutionary perspectives. METHODS: We characterized milk in vitro interleukin-6 (IL-6) responses to Salmonella enterica (a target of protective immune activity; N = 96) and Escherichia coli (a benign target; N = 85) among mother-infant dyads in rural Kilimanjaro, Tanzania. We used ordered logistic regression and mixture models to evaluate maternal and infant characteristics as predictors of IL-6 responses. RESULTS: In all models, IL-6 responses to S. enterica increased with maternal age and decreased with gravidity. In mixture models, IL-6 responses to E. coli declined with maternal age and increased with gravidity. No other considered variables were consistently associated with IL-6 responses. CONCLUSIONS: The ISOM's capacities for appropriate proinflammatory activity and control of misdirected proinflammatory activity increases with maternal age and decreases with gravidity. These findings are consistent with the hypothesis that the mother-infant immune axis has evolved to respond to maternal life history characteristics.


Subject(s)
Interleukin-6 , Milk, Human , Salmonella enterica , Tanzania , Humans , Female , Salmonella enterica/immunology , Adult , Infant , Milk, Human/immunology , Milk, Human/chemistry , Escherichia coli/immunology , Young Adult , Infant, Newborn , Male
6.
Immunity ; 40(6): 896-909, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24882217

ABSTRACT

Animal host defense against infection requires the expression of defense genes at the right place and the right time. Understanding such tight control of host defense requires the elucidation of the transcription factors involved. By using an unbiased approach in the model Caenorhabditis elegans, we discovered that HLH-30 (known as TFEB in mammals) is a key transcription factor for host defense. HLH-30 was activated shortly after Staphylococcus aureus infection, and drove the expression of close to 80% of the host response, including antimicrobial and autophagy genes that were essential for host tolerance of infection. TFEB was also rapidly activated in murine macrophages upon S. aureus infection and was required for proper transcriptional induction of several proinflammatory cytokines and chemokines. Thus, our data suggest that TFEB is a previously unappreciated, evolutionarily ancient transcription factor in the host response to infection.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/immunology , Basic Helix-Loop-Helix Transcription Factors/immunology , Caenorhabditis elegans Proteins/immunology , Caenorhabditis elegans/immunology , Caenorhabditis elegans/microbiology , Staphylococcal Infections/immunology , Animals , Autophagy/genetics , Autophagy/immunology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Caenorhabditis elegans Proteins/genetics , Enterococcus faecalis/immunology , Immunity, Innate , Macrophages/immunology , Mice , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , RNA Interference , RNA, Small Interfering , Salmonella Infections/immunology , Salmonella enterica/immunology , Signal Transduction/immunology , Staphylococcus aureus/immunology , Transcriptional Activation/genetics , Transcriptional Activation/immunology
7.
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
8.
Vet Res ; 53(1): 46, 2022 Jun 22.
Article in English | MEDLINE | ID: mdl-35733156

ABSTRACT

A universal vaccine protecting against multiple serotypes of Streptococcus suis is urgently needed to improve animal welfare and reduce the consumption of antibiotics. In this study, a dual antigen expression cassette consisting of SS2-SaoA and SS9-Eno was delivered by a recombinant Salmonella Choleraesuis vector to form the vaccine candidate rSC0016(pS-SE). SaoA and Eno were simultaneously synthesized in rSC0016(pS-SE) without affecting the colonization of the recombinant vector in the lymphatic system. In addition, the antiserum of mice immunized with rSC0016(pS-SE) produced a broader and potent opsonophagocytic response against multiple serotypes of S. suis. Finally, rSC0016(pS-SE) provided mice with a 100% protection against a lethal dose of parent S. suis serotype 2 and serotype 9, and provided 90% and 80% protection against heterologous S. suis serotype 7 or 1/2. These values were significantly higher than those obtained with rSC0016(pS-SaoA) or rSC0016(pS-Eno). Together, this study serves as a foundation for developing a universal vaccine against multiple serotypes of S. suis.


Subject(s)
Bacterial Vaccines , Cross Protection , Salmonella enterica , Streptococcal Infections , Streptococcus suis , Animals , Bacterial Vaccines/immunology , Cross Protection/immunology , Disease Models, Animal , Mice , Salmonella enterica/genetics , Salmonella enterica/immunology , Serogroup , Streptococcal Infections/prevention & control , Streptococcal Infections/veterinary , Streptococcus suis/genetics , Streptococcus suis/immunology
9.
Nat Immunol ; 10(11): 1137-9, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19841643

ABSTRACT

Mammalian cells ubiquitinate bacteria that erroneously enter the cytosol and target these intruding microbes for destruction by autophagy. New work shows that the protein NDP52 directly binds to ubiquitinated bacteria and facilitates the assembly of an autophagic membrane that surrounds these invaders.


Subject(s)
Autophagy , Nuclear Proteins/immunology , Salmonella enterica/immunology , Ubiquitin/immunology , Humans , Nuclear Proteins/metabolism , Protein Binding , Salmonella Infections/immunology , Ubiquitin/metabolism , Ubiquitination
10.
Nat Immunol ; 10(11): 1215-21, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19820708

ABSTRACT

Cell-autonomous innate immune responses against bacteria attempting to colonize the cytosol of mammalian cells are incompletely understood. Polyubiquitylated proteins can accumulate on the surface of such bacteria, and bacterial growth is restricted by Tank-binding kinase (TBK1). Here we show that NDP52, not previously known to contribute to innate immunity, recognizes ubiquitin-coated Salmonella enterica in human cells and, by binding the adaptor proteins Nap1 and Sintbad, recruits TBK1. Knockdown of NDP52 and TBK1 facilitated bacterial proliferation and increased the number of cells containing ubiquitin-coated salmonella. NDP52 also recruited LC3, an autophagosomal marker, and knockdown of NDP52 impaired autophagy of salmonella. We conclude that human cells utilize the ubiquitin system and NDP52 to activate autophagy against bacteria attempting to colonize their cytosol.


Subject(s)
Autophagy , Nuclear Proteins/immunology , Salmonella enterica/immunology , Ubiquitin/immunology , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , HeLa Cells , Humans , Microtubule-Associated Proteins/immunology , Microtubule-Associated Proteins/metabolism , Molecular Sequence Data , Nuclear Proteins/metabolism , Protein Binding , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Proteins/immunology , Proteins/metabolism , RNA Interference , Salmonella Infections/immunology , Ubiquitin/metabolism , Ubiquitination , tRNA Methyltransferases
11.
J Immunol ; 202(3): 920-930, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30593537

ABSTRACT

Common IFN regulatory factor 5 (IRF5) variants associated with multiple immune-mediated diseases are a major determinant of interindividual variability in pattern recognition receptor (PRR)-induced cytokines in macrophages. PRR-initiated pathways also contribute to bacterial clearance, and dysregulation of bacterial clearance can contribute to immune-mediated diseases. However, the role of IRF5 in macrophage-mediated bacterial clearance is not well defined. Furthermore, it is unclear if macrophages from individuals who are carriers of low IRF5-expressing genetic variants associated with protection for immune-mediated diseases might be at a disadvantage in bacterial clearance. We found that IRF5 was required for optimal bacterial clearance in PRR-stimulated, M1-differentiated human macrophages. Mechanisms regulated by IRF5 included inducing reactive oxygen species through p40phox, p47phox and p67phox, NOS2, and autophagy through ATG5. Complementing these pathways in IRF5-deficient M1 macrophages restored bacterial clearance. Further, these antimicrobial pathways required the activation of IRF5-dependent MAPK, NF-κB, and Akt2 pathways. Importantly, relative to high IRF5-expressing rs2004640/rs2280714 TT/TT immune-mediated disease risk-carrier human macrophages, M1-differentiated GG/CC carrier macrophages demonstrated less reactive oxygen species, NOS2, and autophagy pathway induction and, consequently, reduced bacterial clearance. Increasing IRF5 expression to the rs2004640/rs2280714 TT/TT levels restored these antimicrobial pathways. We define mechanisms wherein common IRF5 genetic variants modulate bacterial clearance, thereby highlighting that immune-mediated disease risk IRF5 carriers might be relatively protected from microbial-associated diseases.


Subject(s)
Bacteria/immunology , Interferon Regulatory Factors/immunology , Macrophages/immunology , Macrophages/microbiology , Autophagy , Autophagy-Related Protein 5/genetics , Cell Differentiation , Cells, Cultured , Enterococcus faecalis/immunology , Genetic Predisposition to Disease , Genetic Variation , Humans , Interferon Regulatory Factors/genetics , Interferon-gamma/pharmacology , Nitric Oxide Synthase Type II/metabolism , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Receptors, Pattern Recognition/immunology , Salmonella enterica/immunology , Signal Transduction
12.
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
13.
J Bacteriol ; 202(5)2020 02 11.
Article in English | MEDLINE | ID: mdl-31792013

ABSTRACT

Bacterial lipopolysaccharides are major components and contributors to the integrity of Gram-negative outer membranes. The more conserved lipid A-core part of this complex glycolipid is synthesized separately from the hypervariable O-antigenic polysaccharide (OPS) part, and they are joined in the periplasm prior to translocation to the outer membrane. Three different biosynthesis strategies are recognized for OPS biosynthesis, and one, the synthase-dependent pathway, is currently confined to a single example: the O:54 antigen from Salmonella enterica serovar Borreze. Synthases are complex enzymes that have the capacity to both polymerize and export bacterial polysaccharides. Although synthases like cellulose synthase are widespread, they typically polymerize a glycan without employing a lipid-linked intermediate, unlike the O:54 synthase (WbbF), which produces an undecaprenol diphosphate-linked product. This raises questions about the overall similarity between WbbF and conventional synthases. In this study, we examine the topology of WbbF, revealing four membrane-spanning helices, compared to the eight in cellulose synthase. Molecular modeling of the glycosyltransferase domain of WbbF indicates a similar architecture, and site-directed mutagenesis confirmed that residues important for catalysis and processivity in cellulose synthase are conserved in WbbF and required for its activity. These findings indicate that the glycosyltransferase mechanism of WbbF and classic synthases are likely conserved despite the use of a lipid acceptor for chain extension by WbbF.IMPORTANCE Glycosyltransferases play a critical role in the synthesis of a wide variety of bacterial polysaccharides. These include O-antigenic polysaccharides, which form the distal component of lipopolysaccharides and provide a protective barrier important for survival and host-pathogen interactions. Synthases are a subset of glycosyltransferases capable of coupled synthesis and export of glycans. Currently, the O:54 antigen of Salmonella enterica serovar Borreze involves the only example of an O-polysaccharide synthase, and its generation of a lipid-linked product differentiates it from classical synthases. Here, we explore features conserved in the O:54 enzyme and classical synthases to shed light on the structure and function of the unusual O:54 enzyme.


Subject(s)
Catalytic Domain , Glycosyltransferases/chemistry , Models, Molecular , Salmonella enterica/enzymology , Amino Acid Sequence , Catalysis , O Antigens/biosynthesis , Recombinant Fusion Proteins , Salmonella enterica/immunology
14.
Infect Immun ; 88(4)2020 03 23.
Article in English | MEDLINE | ID: mdl-32014897

ABSTRACT

Nontyphoidal Salmonella species are globally disseminated pathogens and are the predominant cause of gastroenteritis. The pathogenesis of salmonellosis has been extensively studied using in vivo murine models and cell lines, typically challenged with Salmonella enterica serovar Typhimurium. Although S. enterica serovars Enteritidis and Typhimurium are responsible for most of the human infections reported to the Centers for Disease Control and Prevention (CDC), several other serovars also contribute to clinical cases of salmonellosis. Despite their epidemiological importance, little is known about their infection phenotypes. Here, we report the virulence characteristics and genomes of 10 atypical S. enterica serovars linked to multistate foodborne outbreaks in the United States. We show that the murine RAW 264.7 macrophage model of infection is unsuitable for inferring human-relevant differences in nontyphoidal Salmonella infections, whereas differentiated human THP-1 macrophages allowed these isolates to be further characterized in a more human-relevant context.


Subject(s)
Macrophages/immunology , Macrophages/microbiology , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella enterica/growth & development , Salmonella enterica/immunology , Animals , Humans , Mice , Models, Biological , RAW 264.7 Cells , THP-1 Cells , Virulence
15.
BMC Genomics ; 21(1): 202, 2020 Mar 04.
Article in English | MEDLINE | ID: mdl-32131727

ABSTRACT

BACKGROUND: Salmonella enterica subsp. enterica serovar Napoli (S. Napoli) is among the top serovars causing human infections in Italy, although it is relatively uncommon in other European countries; it is mainly isolated from humans and the environment, but neither the reservoir nor its route of infection are clearly defined. This serovar is characterized by high genomic diversity, and molecular evidences revealed important similarities with typhoidal serovars. RESULTS: 179 S. Napoli genomes as well as 239 genomes of typhoidal and non-typhoidal serovars were analyzed in a comparative genomic study. Phylogenetic analysis and draft genome characterization in terms of Multi Locus Sequence Typing (MLST), plasmid replicons, Salmonella Pathogenicity Islands (SPIs), antimicrobial resistance genes (ARGs), phages, biocide and metal-tolerance genes confirm the high genetic variability of S. Napoli, also revealing a within-serovar phylogenetic structure more complex than previously known. Our work also confirms genomic similarity of S. Napoli to typhoidal serovars (S. Typhi and S. Paratyphi A), with S. Napoli samples clustering primarily according to ST, each being characterized by specific genomic traits. Moreover, two major subclades of S. Napoli can be clearly identified, with ST-474 being biphyletic. All STs span among isolation sources and years of isolation, highlighting the challenge this serovar poses to define its epidemiology and evolution. Altogether, S. Napoli strains carry less SPIs and less ARGs than other non-typhoidal serovars and seldom acquire plasmids. However, we here report the second case of an extended-spectrum ß-lactamases (ESBLs) producing S. Napoli strain and the first cases of multidrug resistant (MDR) S. Napoli strains, all isolated from humans. CONCLUSIONS: Our results provide evidence of genomic plasticity of S. Napoli, highlighting genomic similarity with typhoidal serovars and genomic features typical of non-typhoidal serovars, supporting the possibility of survival in different niches, both enteric and non-enteric. Presence of horizontally acquired ARGs and MDR profiles rises concerns regarding possible selective pressure exerted by human environment on this pathogen.


Subject(s)
DNA, Bacterial/genetics , Salmonella Infections/microbiology , Salmonella enterica/classification , Whole Genome Sequencing/methods , Drug Resistance, Multiple, Bacterial , Genomic Islands , Genomics , High-Throughput Nucleotide Sequencing , Humans , Italy , Phylogeny , Plasmids/genetics , Salmonella enterica/genetics , Salmonella enterica/immunology , Salmonella enterica/isolation & purification , Serogroup , Typhoid Fever/microbiology , beta-Lactam Resistance
16.
J Immunol ; 200(10): 3626-3634, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29654208

ABSTRACT

The cytokine IFN-γ has well-established antibacterial properties against the bacterium Salmonella enterica in phagocytes, but less is known about the effects of IFN-γ on Salmonella-infected nonphagocytic cells, such as intestinal epithelial cells (IECs) and fibroblasts. In this article, we show that exposing human and murine IECs and fibroblasts to IFN-γ following infection with Salmonella triggers a novel form of cell death that is neither pyroptosis nor any of the major known forms of programmed cell death. Cell death required IFN-γ-signaling via STAT1-IRF1-mediated induction of guanylate binding proteins and the presence of live Salmonella in the cytosol. In vivo, ablating IFN-γ signaling selectively in murine IECs led to higher bacterial burden in colon contents and increased inflammation in the intestine of infected mice. Together, these results demonstrate that IFN-γ signaling triggers release of Salmonella from the Salmonella-containing vacuole into the cytosol of infected nonphagocytic cells, resulting in a form of nonpyroptotic cell death that prevents bacterial spread in the gut.


Subject(s)
Cell Death/immunology , Interferon-gamma/immunology , Phagocytes/immunology , Pyroptosis/immunology , Salmonella Infections/immunology , Salmonella enterica/immunology , 3T3 Cells , Animals , Cell Line , Cytosol/immunology , Cytosol/microbiology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Fibroblasts/immunology , Fibroblasts/microbiology , Humans , Inflammation/immunology , Inflammation/microbiology , Interferon Regulatory Factor-1/immunology , Intestines/immunology , Intestines/microbiology , Mice , Phagocytes/microbiology , STAT1 Transcription Factor/immunology , Salmonella Infections/microbiology
17.
Avian Pathol ; 49(6): 666-677, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32907345

ABSTRACT

A novel inactivated vaccine, comprising three serovars of Salmonella enterica (Enteritidis, serogroup O:9; Typhimurium, serogroup O:4; Infantis, serogroup O:7) grown under conditions of iron restriction and adjuvanted with aluminium hydroxide, was evaluated for efficacy following challenge by homologous and heterologous serovars. Chickens were vaccinated at 6 and 10 weeks of age by the intramuscular route and challenged 4 to 9 weeks after the second vaccination with serovars belonging to serogroup O:9 (Enteritidis), O:4 (Typhimurium and Heidelberg), O:7 (Infantis and Virchow), and O:8 (Hadar). All vaccinated birds produced a marked systemic antibody response against each of the component vaccine antigens by the time of challenge. Significant reductions in both colonization of the intestinal tract and invasion of internal organs were observed in vaccinated birds compared with non-vaccinated controls, irrespective of the challenge serovar. The findings suggest that broad serovar protection within the constitutive serogroups of an inactivated multi-valent vaccine is possible and could, therefore, play an important role in future Salmonella control programmes. RESEARCH HIGHLIGHTS Novel inactivated trivalent Salmonella chicken vaccine was developed and tested. Vaccine induced marked systemic antibody response against all vaccine antigens. Significant reductions in intestinal tract colonization and internal organ invasion. Vaccine efficacy demonstrated against homologous and heterologous serovars.


Subject(s)
Chickens/immunology , Poultry Diseases/prevention & control , Salmonella Infections, Animal/prevention & control , Salmonella Vaccines/immunology , Salmonella enterica/immunology , Vaccination/veterinary , Animals , Chickens/microbiology , Poultry Diseases/microbiology , Salmonella Infections, Animal/microbiology , Serogroup , Vaccines, Inactivated
18.
J Immunol ; 199(7): 2491-2502, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28827286

ABSTRACT

The anti-proliferative agent hexamethylene bisacetamide (HMBA) belongs to a class of hybrid bipolar compounds developed more than 30 y ago for their ability to induce terminal differentiation of transformed cells. Recently, HMBA has also been shown to trigger HIV transcription from latently infected cells, via a CDK9/HMBA inducible protein-1 dependent process. However, the effect of HMBA on the immune response has not been explored. We observed that pretreatment of human peripheral blood mononuclear cells with HMBA led to a markedly increased production of IL-12 and IFN-γ, but not of TNF-α, IL-6, and IL-8 upon subsequent infection with Burkholderia pseudomallei and Salmonella enterica HMBA treatment was also associated with better intracellular bacterial control. HMBA significantly improved IL-12p70 production from CD14+ monocytes during infection partly via the induction of type I IFN in these cells, which primed an increased transcription of the p35 subunit of IL-12p70 during infection. HMBA also increased early type I IFN transcription in human monocytic and epithelial cell lines, but this was surprisingly independent of its previously reported effects on positive transcription elongation factor b and HMBA inducible protein-1. Instead, the effect of HMBA was downstream of a calcium influx, and required the pattern recognition receptor and adaptor STING but not cGAS. Our work therefore links the STING-IRF3 axis to enhanced IL-12 production and intracellular bacterial control in primary monocytes. This raises the possibility that HMBA or related small molecules may be explored as therapeutic adjuvants to improve disease outcomes during intracellular bacterial infections.


Subject(s)
Acetamides/pharmacology , Adjuvants, Immunologic , Interferon Type I/biosynthesis , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/microbiology , Membrane Proteins/metabolism , Acetamides/therapeutic use , Burkholderia pseudomallei/drug effects , Burkholderia pseudomallei/immunology , Cell Line , Cells, Cultured , Cytoplasm/immunology , Cytoplasm/microbiology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/microbiology , Humans , Interferon Regulatory Factor-3/metabolism , Interferon Type I/genetics , Interferon Type I/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-12/biosynthesis , Interleukin-12/immunology , Interleukin-6/biosynthesis , Interleukin-6/immunology , Interleukin-8/biosynthesis , Interleukin-8/immunology , Leukocytes, Mononuclear/immunology , Membrane Proteins/immunology , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , Salmonella enterica/drug effects , Salmonella enterica/immunology , Signal Transduction , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
19.
Avian Pathol ; 48(5): 423-428, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31081347

ABSTRACT

Salmonella enterica serovar Gallinarum causes a disease in chickens known as fowl typhoid. Interferon-gamma (IFN-γ) has been shown to be crucial in eliminating salmonellosis infection because of its strong association with T-cell responses. This study was undertaken to compare the expression of IFN-γ in chickens generated by different vaccine formulations. Eighty one-day-old Lohmann layer chicks were divided into four groups of 20 birds each for the experiment. This comprised an unvaccinated negative control group (NEG), a group vaccinated with the live 9R vaccine by the injection route (SC), a group vaccinated with alginate-coated chitosan microparticles encapsulating live plasmid-cured S. Gallinarum strain 9 (PC) by the oral route, and a group vaccinated with a weak attenuated live S. Gallinarum strain 9 encapsulated in alginate-coated chitosan microparticles (VM) given orally. Vaccinations were done at 10 and 14 weeks of age followed by challenge at 16 weeks of age. IgG was measured using ELISA. qRT-PCR was used to compare the mRNA fold expression of IFN-γ in the PC, VM and SC groups using the unvaccinated/unchallenged group as the control. There were significant differences in the IgG levels between each vaccinated group and the unvaccinated group (P < 0.05) after booster vaccination and post-challenge. There was 100% protection of the birds in SC and VM groups, 80% protection in PC group and 0% protection in the NEG group. Using 2-ΔΔCT calculation, IFN-γ was more highly expressed in the PC group than in the SC group or VM group. In conclusion, the IFN-γ was more highly expressed in the PC group (though not significantly higher) compared to the SC and VM groups and this could be attributed to the alginate-coated chitosan microparticles which acted as an adjuvant.


Subject(s)
Chickens/immunology , Interferon-gamma/analysis , Poultry Diseases/prevention & control , Salmonella Infections, Animal/prevention & control , Salmonella Vaccines/administration & dosage , Salmonella enterica/immunology , Administration, Oral , Alginates/chemistry , Animals , Chitosan/chemistry , Female , Immunity, Cellular , Immunity, Humoral , Immunization, Secondary/veterinary , Plasmids/genetics , Poultry Diseases/microbiology , Salmonella Infections, Animal/microbiology , Salmonella Vaccines/immunology , Typhoid-Paratyphoid Vaccines/administration & dosage , Up-Regulation
20.
Adv Exp Med Biol ; 1111: 205-218, 2019.
Article in English | MEDLINE | ID: mdl-30411307

ABSTRACT

Bacteria deliver virulence proteins termed 'effectors' to counteract host innate immunity. Protein-protein interactions within the host cell ultimately subvert the generation of an inflammatory response to the infecting pathogen. Here we briefly describe a subset of T3SS effectors produced by enterohemorrhagic Escherichia coli (EHEC), enteropathogenic E. coli (EPEC), Citrobacter rodentium, and Salmonella enterica that inhibit innate immune pathways. These effectors are interesting for structural and mechanistic reasons, as well as for their potential utility in being engineered to treat human autoimmune disorders associated with perturbations in NF-κB signaling.


Subject(s)
Citrobacter rodentium/immunology , Enteropathogenic Escherichia coli/immunology , Escherichia coli Proteins/metabolism , Immunity, Innate/immunology , Salmonella enterica/immunology , Type III Secretion Systems/metabolism , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Citrobacter rodentium/metabolism , Enteropathogenic Escherichia coli/metabolism , Enteropathogenic Escherichia coli/pathogenicity , Humans , Salmonella enterica/metabolism , Virulence Factors/immunology , Virulence Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL