Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Nat Commun ; 15(1): 2778, 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38555361

ABSTRACT

Bacterial genotoxins damage host cells by targeting their chromosomal DNA. In the present study, we demonstrate that a genotoxin of Salmonella Typhi, typhoid toxin, triggers the senescence-associated secretory phenotype (SASP) by damaging mitochondrial DNA. The actions of typhoid toxin disrupt mitochondrial DNA integrity, leading to mitochondrial dysfunction and disturbance of redox homeostasis. Consequently, it facilitates the release of damaged mitochondrial DNA into the cytosol, activating type I interferon via the cGAS-STING pathway. We also reveal that the GCN2-mediated integrated stress response plays a role in the upregulation of inflammatory components depending on the STING signaling axis. These SASP factors can propagate the senescence effect on T cells, leading to senescence in these cells. These findings provide insights into how a bacterial genotoxin targets mitochondria to trigger a proinflammatory SASP, highlighting a potential therapeutic target for an anti-toxin intervention.


Subject(s)
Senescence-Associated Secretory Phenotype , Typhoid Fever , Humans , Typhoid Fever/metabolism , Mutagens/metabolism , Cellular Senescence/physiology , Mitochondria/metabolism , DNA, Mitochondrial/metabolism , Salmonella , Phenotype
2.
Front Immunol ; 12: 728685, 2021.
Article in English | MEDLINE | ID: mdl-34659215

ABSTRACT

Mucosal-associated invariant T (MAIT) cells are an innate-like population of T cells that display a TCR Vα7.2+ CD161+ phenotype and are restricted by the nonclassical MHC-related molecule 1 (MR1). Although B cells control MAIT cell development and function, little is known about the mechanisms underlying their interaction(s). Here, we report, for the first time, that during Salmonella enterica serovar Typhi (S. Typhi) infection, HLA-G expression on B cells downregulates IFN-γ production by MAIT cells. In contrast, blocking HLA-G expression on S. Typhi-infected B cells increases IFN-γ production by MAIT cells. After interacting with MAIT cells, kinetic studies show that B cells upregulate HLA-G expression and downregulate the inhibitory HLA-G receptor CD85j on MAIT cells resulting in their loss. These results provide a new role for HLA-G as a negative feedback loop by which B cells control MAIT cell responses to antigens.


Subject(s)
Antigens, CD/metabolism , B-Lymphocytes/metabolism , HLA-G Antigens/metabolism , Leukocyte Immunoglobulin-like Receptor B1/metabolism , Mucosal-Associated Invariant T Cells/metabolism , Salmonella typhi/pathogenicity , Typhoid Fever/metabolism , Adult , Antigens, CD/genetics , B-Lymphocytes/immunology , B-Lymphocytes/microbiology , Cells, Cultured , Coculture Techniques , Female , Host-Pathogen Interactions , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , Kinetics , Leukocyte Immunoglobulin-like Receptor B1/genetics , Male , Middle Aged , Mucosal-Associated Invariant T Cells/immunology , Mucosal-Associated Invariant T Cells/microbiology , Phenotype , Salmonella typhi/immunology , Signal Transduction , Typhoid Fever/genetics , Typhoid Fever/immunology , Typhoid Fever/microbiology , Young Adult
3.
Int J Mol Sci ; 22(18)2021 Sep 16.
Article in English | MEDLINE | ID: mdl-34576166

ABSTRACT

Salmonella enterica serovar Typhi (S. Typhi) is a human-limited intracellular pathogen and the cause of typhoid fever, a severe systemic disease. Pathogen-host interaction at the metabolic level affects the pathogenicity of intracellular pathogens, but it remains unclear how S. Typhi infection influences host metabolism for its own benefit. Herein, using metabolomics and transcriptomics analyses, combined with in vitro and in vivo infection assays, we investigated metabolic responses in human macrophages during S. Typhi infection, and the impact of these responses on S. Typhi intracellular replication and systemic pathogenicity. We observed increased glucose content, higher rates of glucose uptake and glycolysis, and decreased oxidative phosphorylation in S. Typhi-infected human primary macrophages. Replication in human macrophages and the bacterial burden in systemic organs of humanized mice were reduced by either the inhibition of host glucose uptake or a mutation of the bacterial glucose uptake system, indicating that S. Typhi utilizes host-derived glucose to enhance intracellular replication and virulence. Thus, S. Typhi promotes its pathogenicity by inducing metabolic changes in host macrophages and utilizing the glucose that subsequently accumulates as a nutrient for intracellular replication. Our findings provide the first metabolic signature of S. Typhi-infected host cells and identifies a new strategy utilized by S. Typhi for intracellular replication.


Subject(s)
Glucose/metabolism , Salmonella typhi/pathogenicity , Typhoid Fever/metabolism , Typhoid Fever/microbiology , Host-Pathogen Interactions , Humans , Macrophages/metabolism , Macrophages/microbiology , Virulence
4.
PLoS Pathog ; 16(7): e1008591, 2020 07.
Article in English | MEDLINE | ID: mdl-32645118

ABSTRACT

Reactive arthritis, an autoimmune disorder, occurs following gastrointestinal infection with invasive enteric pathogens, such as Salmonella enterica. Curli, an extracellular, bacterial amyloid with cross beta-sheet structure can trigger inflammatory responses by stimulating pattern recognition receptors. Here we show that S. Typhimurium produces curli amyloids in the cecum and colon of mice after natural oral infection, in both acute and chronic infection models. Production of curli was associated with an increase in anti-dsDNA autoantibodies and joint inflammation in infected mice. The negative impacts on the host appeared to be dependent on invasive systemic exposure of curli to immune cells. We hypothesize that in vivo synthesis of curli contributes to known complications of enteric infections and suggest that cross-seeding interactions can occur between pathogen-produced amyloids and amyloidogenic proteins of the host.


Subject(s)
Arthritis, Infectious/immunology , Bacterial Proteins/immunology , Typhoid Fever/immunology , Animals , Antibodies, Antinuclear/immunology , Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Arthritis, Infectious/metabolism , Bacterial Proteins/biosynthesis , Intestine, Large/immunology , Intestine, Large/microbiology , Mice , Typhoid Fever/metabolism
5.
Cell Rep ; 31(11): 107772, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32553163

ABSTRACT

ISG15 is a ubiquitin-like modifier that also functions extracellularly, signaling through the LFA-1 integrin to promote interferon (IFN)-γ release from natural killer (NK) and T cells. The signals that lead to the production of extracellular ISG15 and the relationship between its two core functions remain unclear. We show that both epithelial cells and lymphocytes can secrete ISG15, which then signals in either an autocrine or paracrine manner to LFA-1-expressing cells. Microbial pathogens and Toll-like receptor (TLR) agonists result in both IFN-ß-dependent and -independent secretion of ISG15, and residues required for ISG15 secretion are mapped. Intracellular ISGylation inhibits secretion, and viral effector proteins, influenza B NS1, and viral de-ISGylases, including SARS-CoV-2 PLpro, have opposing effects on secretion of ISG15. These results establish extracellular ISG15 as a cytokine-like protein that bridges early innate and IFN-γ-dependent immune responses, and indicate that pathogens have evolved to differentially inhibit the intracellular and extracellular functions of ISG15.


Subject(s)
Cytokines/metabolism , Signal Transduction , Ubiquitins/metabolism , Animals , HEK293 Cells , Humans , Influenza, Human/immunology , Influenza, Human/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Jurkat Cells , Mice , Mice, Inbred C57BL , Mycobacterium Infections/immunology , Mycobacterium Infections/metabolism , Pathogen-Associated Molecular Pattern Molecules , Typhoid Fever/immunology , Typhoid Fever/metabolism , Viral Nonstructural Proteins/metabolism
6.
FASEB J ; 33(12): 13386-13397, 2019 12.
Article in English | MEDLINE | ID: mdl-31550172

ABSTRACT

Gut-homing γδ T cells are induced by chemokines and cell adhesion molecules and play a critical role in homeostasis and mucosal immunity; however, little is known regarding their upstream regulators. We investigated the role of Axl as a specific regulator of chemokines and cell adhesion molecule in the distribution of intestinal γδ T cells. The population of γδ T-cell receptor-positive cells including Vγ1 and Vγ7 subsets was remarkably increased in the intraepithelial lymphocytes of Axl-/- mice compared with those of wild-type (WT) mice. An increased number of migrated γδ T cells were observed in the coculture with intraepithelial cells from Axl-/- mice. The mRNA expression level of chemokine (C-C motif) ligand (CCL) 25 was specifically higher in the small intestine of Axl-/- mice than in WT mice. In adoptive transfer, the migration of both thymic and extrathymic γδ T cells was increased in Axl-/- mice. The activation of Axl signaling down-regulated CCL25 expression via ERK signaling pathway and reduced the population of γδ T cells. Systemic dissemination was suppressed in Axl-/- mice infected with Salmonella typhimurium. Thus, our findings suggest that Axl plays a critical role in regulating the migration of γδ T cells for the maintenance of homeostasis and bacterial resistance.-Kim, S.-M., Park, M., Yee, S.-M., Ji, K.-Y., Lee, E.-H., Nguyen, T.-V., Nguyen, T. H.-L., Jang, J., Kim, E.-M., Choi, H.-R., Yun, C.-H., Kang, H.-S. Axl is a key regulator of intestinal γδ T-cell homeostasis.


Subject(s)
Epithelial Cells/immunology , Homeostasis , Intestine, Small/immunology , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , Typhoid Fever/immunology , Animals , Cell Movement , Cells, Cultured , Chemokines, CC/metabolism , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Immunity, Mucosal , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestine, Small/metabolism , Intestine, Small/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Salmonella typhimurium/physiology , Typhoid Fever/metabolism , Typhoid Fever/microbiology , Axl Receptor Tyrosine Kinase
7.
Proc Natl Acad Sci U S A ; 116(38): 19077-19082, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31484771

ABSTRACT

Serum amyloid A (SAA) proteins are strongly induced in the liver by systemic infection and in the intestine by bacterial colonization. In infected mice, SAA proteins circulate in association with the vitamin A derivative retinol, suggesting that SAAs transport retinol during infection. Here we illuminate a structural basis for the retinol-SAA interaction. In the bloodstream of infected mice, most SAA is complexed with high-density lipoprotein (HDL). However, we found that the majority of the circulating retinol was associated with the small fraction of SAA proteins that circulate without binding to HDL, thus identifying free SAA as the predominant retinol-binding form in vivo. We then determined the crystal structure of retinol-bound mouse SAA3 at a resolution of 2.2 Å. Retinol-bound SAA3 formed a novel asymmetric trimeric assembly that was generated by the hydrophobic packing of the conserved amphipathic helices α1 and α3. This hydrophobic packing created a retinol-binding pocket in the center of the trimer, which was confirmed by mutagenesis studies. Together, these findings illuminate the molecular basis for retinol transport by SAA proteins during infection.


Subject(s)
Salmonella typhimurium/metabolism , Serum Amyloid A Protein/chemistry , Serum Amyloid A Protein/metabolism , Typhoid Fever/metabolism , Vitamin A/metabolism , Vitamins/metabolism , Animals , Crystallography, X-Ray , Mice , Mice, Knockout , Models, Molecular , Mutation , Protein Conformation , Serum Amyloid A Protein/genetics , Typhoid Fever/virology
8.
PLoS Pathog ; 15(4): e1007704, 2019 04.
Article in English | MEDLINE | ID: mdl-30951565

ABSTRACT

Typhoid toxin is a virulence factor for Salmonella Typhi and Paratyphi, the cause of typhoid fever in humans. This toxin has a unique architecture in that its pentameric B subunit, made of PltB, is linked to two enzymatic A subunits, the ADP ribosyl transferase PltA and the deoxyribonuclease CdtB. Typhoid toxin is uniquely adapted to humans, recognizing surface glycoprotein sialoglycans terminated in acetyl neuraminic acid, which are preferentially expressed by human cells. The transport pathway to its cellular targets followed by typhoid toxin after receptor binding is currently unknown. Through a genome-wide CRISPR/Cas9-mediated screen we have characterized the mechanisms by which typhoid toxin is transported within human cells. We found that typhoid toxin hijacks specific elements of the retrograde transport and endoplasmic reticulum-associated degradation machineries to reach its subcellular destination within target cells. Our study reveals unique and common features in the transport mechanisms of bacterial toxins that could serve as the bases for the development of novel anti-toxin therapeutic strategies.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Endoplasmic Reticulum-Associated Degradation , Salmonella typhi/pathogenicity , Typhoid Fever/microbiology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Biological Transport , CRISPR-Cas Systems , HeLa Cells , Humans , Intracellular Space/metabolism , Protein Binding , Salmonella typhi/genetics , Typhoid Fever/genetics , Typhoid Fever/metabolism
9.
Cell Microbiol ; 21(8): e13034, 2019 08.
Article in English | MEDLINE | ID: mdl-31013389

ABSTRACT

How Salmonella enterica serovar Typhi (S. Typhi), an important human pathogen, survives the stressful microenvironments inside the gastrointestinal tract and within macrophages remains poorly understood. We report here that S. Typhi has a bonafide stringent response (SR) system, which is mediated by (p)ppGpp and regulates multiple virulence-associated traits and the pathogenicity of the S. Typhi Ty2 strain. In an iron overload mouse model of S. Typhi infection, the (p)ppGpp0 (Ty2ΔRelAΔSpoT) strain showed minimal systemic spread and no mortality, as opposed to 100% death of the mice challenged with the isogenic wild-type strain. Ty2ΔRelAΔSpoT had markedly elongated morphology with incomplete septa formation and demonstrated severely attenuated motility and chemotaxis due to the loss of flagella. Absence of the Vi-polysaccharide capsule rendered the mutant strain highly susceptible to complement-mediated lysis. The phenotypes of Ty2ΔRelAΔSpoT was contributed by transcriptional repression of several genes, including fliC, tviA, and ftsZ, as found by reverse transcriptase quantitative polymerase chain reaction and gene complementation studies. Finally, Ty2ΔRelAΔSpoT had markedly reduced invasion into intestinal epithelial cells and significantly attenuated survival within macrophages. To the best of our knowledge, this was the first study that addressed SR in S. Typhi and showed that (p)ppGpp was essential for optimal pathogenic fitness of the organism.


Subject(s)
Bacterial Proteins/genetics , Guanosine Pentaphosphate/metabolism , Host-Pathogen Interactions/genetics , Salmonella typhi/genetics , Salmonella typhi/pathogenicity , Typhoid Fever/microbiology , Animals , Bacterial Proteins/metabolism , Caco-2 Cells , Disease Models, Animal , GTP Pyrophosphokinase/deficiency , GTP Pyrophosphokinase/genetics , Gene Expression Regulation, Bacterial , HT29 Cells , Humans , Iron Overload/metabolism , Iron Overload/microbiology , Iron Overload/mortality , Iron Overload/pathology , Liver/metabolism , Liver/microbiology , Liver/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Polysaccharides, Bacterial/deficiency , Pyrophosphatases/deficiency , Pyrophosphatases/genetics , RAW 264.7 Cells , Salmonella typhi/growth & development , Salmonella typhi/metabolism , Signal Transduction , Spleen/metabolism , Spleen/microbiology , Spleen/pathology , Survival Analysis , THP-1 Cells , Typhoid Fever/metabolism , Typhoid Fever/mortality , Typhoid Fever/pathology , Virulence
10.
PLoS Pathog ; 15(4): e1007741, 2019 04.
Article in English | MEDLINE | ID: mdl-31009521

ABSTRACT

Intracellular Salmonella enterica serovar Typhimurium (STM) deploy the Salmonella Pathogenicity Island 2-encoded type III secretion system (SPI2-T3SS) for the massive remodeling of the endosomal system for host cells. This activity results in formation of an extensive interconnected tubular network of Salmonella-induced filaments (SIFs) connected to the Salmonella-containing vacuole (SCV). Such network is absent in cells infected with SPI2-T3SS-deficient mutant strains such as ΔssaV. A tubular network with reduced dimensions is formed if SPI2-T3SS effector protein SseF is absent. Previous single cell live microscopy-based analyses revealed that intracellular proliferation of STM is directly correlated to the ability to transform the host cell endosomal system into a complex tubular network. This network may also abrogate host defense mechanisms such as delivery of antimicrobial effectors to the SCV. To test the role of SIFs in STM patho-metabolism, we performed quantitative comparative proteomics of STM recovered from infected murine macrophages. We infected RAW264.7 cells with STM wild type (WT), ΔsseF or ΔssaV strains, recovered bacteria 12 h after infection and determined proteome compositions. Increased numbers of proteins characteristic for nutritional starvation were detected in STM ΔsseF and ΔssaV compared to WT. In addition, STM ΔssaV, but not ΔsseF showed signatures of increased exposure to stress by antimicrobial defenses, in particular reactive oxygen species, of the host cells. The proteomics analyses presented here support and extend the role of SIFs for the intracellular lifestyle of STM. We conclude that efficient manipulation of the host cell endosomal system by effector proteins of the SPI2-T3SS contributes to nutrition, as well as to resistance against antimicrobial host defense mechanisms.


Subject(s)
Antioxidants/metabolism , Bacterial Proteins/metabolism , Macrophages/metabolism , Proteome/analysis , Proteomics/methods , Salmonella typhi/metabolism , Typhoid Fever/metabolism , Animals , Cells, Cultured , Computational Biology , Genomic Islands , Macrophages/microbiology , Mice , Protein Interaction Maps , Typhoid Fever/microbiology
11.
Sci Adv ; 5(2): eaau6849, 2019 02.
Article in English | MEDLINE | ID: mdl-30820452

ABSTRACT

Epidemiological and immunological evidence suggests that some vaccines can reduce all-cause mortality through nonspecific changes made to innate immune cells. Here, we present the first data to describe the nonspecific immunological impact of oral vaccination with live-attenuated Salmonella Typhi strain Ty21a. We vaccinated healthy adults with Ty21a and assessed aspects of innate and adaptive immunity over the course of 6 months. Changes to monocyte phenotype/function were observed for at least 3 months. Changes to innate and adaptive immune cell cytokine production in response to stimulation with vaccine and unrelated nonvaccine antigens were observed over the 6-month study period. The changes that we have observed could influence susceptibility to infection through altered immune responses mounted to subsequently encountered pathogens. These changes could influence all-cause mortality.


Subject(s)
Polysaccharides, Bacterial/immunology , Salmonella typhi/immunology , Typhoid Fever/prevention & control , Typhoid-Paratyphoid Vaccines/immunology , Vaccination , Vaccines, Attenuated/immunology , Administration, Oral , Adult , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Female , Healthy Volunteers , Humans , Male , Middle Aged , Monocytes/immunology , Monocytes/metabolism , Polysaccharides, Bacterial/administration & dosage , Typhoid Fever/immunology , Typhoid Fever/metabolism , Typhoid-Paratyphoid Vaccines/administration & dosage , Vaccines, Attenuated/administration & dosage , Young Adult
12.
J Exp Med ; 216(4): 757-771, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30804000

ABSTRACT

Salmonella species are among the world's most prevalent pathogens. Because the cell wall interfaces with the host, we designed a lipidomics approach to reveal pathogen-specific cell wall compounds. Among the molecules differentially expressed between Salmonella Paratyphi and S. Typhi, we focused on lipids that are enriched in S. Typhi, because it causes typhoid fever. We discovered a previously unknown family of trehalose phospholipids, 6,6'-diphosphatidyltrehalose (diPT) and 6-phosphatidyltrehalose (PT). Cardiolipin synthase B (ClsB) is essential for PT and diPT but not for cardiolipin biosynthesis. Chemotyping outperformed clsB homology analysis in evaluating synthesis of diPT. DiPT is restricted to a subset of Gram-negative bacteria: large amounts are produced by S. Typhi, lower amounts by other pathogens, and variable amounts by Escherichia coli strains. DiPT activates Mincle, a macrophage activating receptor that also recognizes mycobacterial cord factor (6,6'-trehalose dimycolate). Thus, Gram-negative bacteria show convergent function with mycobacteria. Overall, we discovered a previously unknown immunostimulant that is selectively expressed among medically important bacterial species.


Subject(s)
Mycobacterium/metabolism , Phospholipids/metabolism , Salmonella typhi/metabolism , Trehalose/metabolism , Animals , Cell Membrane , Escherichia coli/metabolism , Feces/microbiology , Humans , Lectins, C-Type/metabolism , Membrane Proteins/metabolism , Mice , Phylogeny , Receptors, Immunologic/metabolism , Transferases (Other Substituted Phosphate Groups)/metabolism , Typhoid Fever/metabolism , Typhoid Fever/microbiology
13.
Nat Commun ; 9(1): 5094, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30504848

ABSTRACT

There is paucity of data regarding the geographical distribution, incidence, and phylogenetics of multi-drug resistant (MDR) Salmonella Typhi in sub-Saharan Africa. Here we present a phylogenetic reconstruction of whole genome sequenced 249 contemporaneous S. Typhi isolated between 2008-2015 in 11 sub-Saharan African countries, in context of the 2,057 global S. Typhi genomic framework. Despite the broad genetic diversity, the majority of organisms (225/249; 90%) belong to only three genotypes, 4.3.1 (H58) (99/249; 40%), 3.1.1 (97/249; 39%), and 2.3.2 (29/249; 12%). Genotypes 4.3.1 and 3.1.1 are confined within East and West Africa, respectively. MDR phenotype is found in over 50% of organisms restricted within these dominant genotypes. High incidences of MDR S. Typhi are calculated in locations with a high burden of typhoid, specifically in children aged <15 years. Antimicrobial stewardship, MDR surveillance, and the introduction of typhoid conjugate vaccines will be critical for the control of MDR typhoid in Africa.


Subject(s)
Salmonella Infections/drug therapy , Africa South of the Sahara , Anti-Bacterial Agents/therapeutic use , Drug Resistance, Multiple, Bacterial/genetics , Genetic Variation/genetics , Genotype , Humans , Incidence , Phylogeny , Phylogeography , Salmonella Infections/genetics , Salmonella Infections/metabolism , Salmonella typhi/classification , Salmonella typhi/pathogenicity , Typhoid Fever/drug therapy , Typhoid Fever/genetics , Typhoid Fever/metabolism
14.
Nat Microbiol ; 3(11): 1243-1254, 2018 11.
Article in English | MEDLINE | ID: mdl-30250245

ABSTRACT

Protein secretion mechanisms are essential for the virulence of most bacterial pathogens. Typhoid toxin is an essential virulence factor for Salmonella Typhi, the cause of typhoid fever in humans. This toxin is unique in that it is only produced within mammalian cells, and it must be trafficked to the extracellular space before intoxicating target cells. An essential and poorly understood aspect of this transport pathway is the secretion of typhoid toxin from the bacterium into the S. Typhi-containing vacuole. We show here that typhoid toxin secretion requires its translocation to the trans side of the peptidoglycan layer at the bacterial poles for subsequent release through the outer membrane. This translocation process depends on a specialized muramidase, the activity of which requires the localized editing of peptidoglycan by a specific ld-transpeptidase. These studies describe a protein export mechanism that is probably conserved in other bacterial species.


Subject(s)
Bacterial Toxins/metabolism , Muramidase/metabolism , Peptidoglycan/metabolism , Peptidyl Transferases/metabolism , Salmonella typhi/pathogenicity , Typhoid Fever/metabolism , Bacterial Proteins/metabolism , Cell Line , Cell Wall/chemistry , Cell Wall/enzymology , Cell Wall/metabolism , Humans , Muramidase/genetics , Peptidyl Transferases/genetics , Protein Transport , Virulence/genetics
15.
Article in English | MEDLINE | ID: mdl-29473020

ABSTRACT

The human-specific pathogen Salmonella enterica serovar Typhi causes typhoid, a major public health issue in developing countries. Several aspects of its pathogenesis are still poorly understood. S. Typhi possesses 14 fimbrial gene clusters including 12 chaperone-usher fimbriae (stg, sth, bcf, fim, saf, sef, sta, stb, stc, std, ste, and tcf). These fimbriae are weakly expressed in laboratory conditions and only a few are actually characterized. In this study, expression of all S. Typhi chaperone-usher fimbriae and their potential roles in pathogenesis such as interaction with host cells, motility, or biofilm formation were assessed. All S. Typhi fimbriae were better expressed in minimal broth. Each system was overexpressed and only the fimbrial gene clusters without pseudogenes demonstrated a putative major subunits of about 17 kDa on SDS-PAGE. Six of these (Fim, Saf, Sta, Stb, Std, and Tcf) also show extracellular structure by electron microscopy. The impact of fimbrial deletion in a wild-type strain or addition of each individual fimbrial system to an S. Typhi afimbrial strain were tested for interactions with host cells, biofilm formation and motility. Several fimbriae modified bacterial interactions with human cells (THP-1 and INT-407) and biofilm formation. However, only Fim fimbriae had a deleterious effect on motility when overexpressed. Overall, chaperone-usher fimbriae seem to be an important part of the balance between the different steps (motility, adhesion, host invasion and persistence) of S. Typhi pathogenesis.


Subject(s)
Fimbriae Proteins/genetics , Fimbriae, Bacterial/genetics , Gene Expression Regulation, Bacterial , Multigene Family , Salmonella typhi/physiology , Typhoid Fever/microbiology , Bacterial Adhesion , Biofilms , Fimbriae Proteins/metabolism , Fimbriae, Bacterial/metabolism , Humans , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Mutation , Operon , Typhoid Fever/immunology , Typhoid Fever/metabolism
16.
Infect Immun ; 86(3)2018 03.
Article in English | MEDLINE | ID: mdl-29229736

ABSTRACT

Salmonella enterica serovars Typhi and Typhimurium cause typhoid fever and gastroenteritis, respectively. A unique feature of typhoid infection is asymptomatic carriage within the gallbladder, which is linked with S Typhi transmission. Despite this, S Typhi responses to bile have been poorly studied. Transcriptome sequencing (RNA-Seq) of S Typhi Ty2 and a clinical S Typhi isolate belonging to the globally dominant H58 lineage (strain 129-0238), as well as S Typhimurium 14028, revealed that 249, 389, and 453 genes, respectively, were differentially expressed in the presence of 3% bile compared to control cultures lacking bile. fad genes, the actP-acs operon, and putative sialic acid uptake and metabolism genes (t1787 to t1790) were upregulated in all strains following bile exposure, which may represent adaptation to the small intestine environment. Genes within the Salmonella pathogenicity island 1 (SPI-1), those encoding a type IIII secretion system (T3SS), and motility genes were significantly upregulated in both S Typhi strains in bile but downregulated in S Typhimurium. Western blots of the SPI-1 proteins SipC, SipD, SopB, and SopE validated the gene expression data. Consistent with this, bile significantly increased S Typhi HeLa cell invasion, while S Typhimurium invasion was significantly repressed. Protein stability assays demonstrated that in S Typhi the half-life of HilD, the dominant regulator of SPI-1, is three times longer in the presence of bile; this increase in stability was independent of the acetyltransferase Pat. Overall, we found that S Typhi exhibits a specific response to bile, especially with regard to virulence gene expression, which could impact pathogenesis and transmission.


Subject(s)
Bile/microbiology , Salmonella Infections/metabolism , Salmonella typhi/metabolism , Salmonella typhimurium/metabolism , Typhoid Fever/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bile/metabolism , Genomic Islands , HeLa Cells , Humans , Salmonella Infections/microbiology , Salmonella typhi/genetics , Salmonella typhimurium/genetics , Serogroup , Species Specificity , Typhoid Fever/microbiology
17.
Int J Mol Sci ; 18(9)2017 Aug 31.
Article in English | MEDLINE | ID: mdl-28858232

ABSTRACT

Salmonella enterica subspecies enterica serovar Typhi is the aetiological agent of typhoid or enteric fever. In a subset of individuals, S. Typhi colonizes the gallbladder causing an asymptomatic chronic infection. Nonetheless, these asymptomatic carriers provide a reservoir for further spreading of the disease. Epidemiological studies performed in regions where S. Typhi is endemic, revealed that the majority of chronically infected carriers also harbour gallstones, which in turn, have been indicated as a primary predisposing factor for the onset of gallbladder cancer (GC). It is now well recognised, that S. Typhi produces a typhoid toxin with a carcinogenic potential, that induces DNA damage and cell cycle alterations in intoxicated cells. In addition, biofilm production by S. Typhi may represent a key factor for the promotion of a persistent infection in the gallbladder, thus sustaining a chronic local inflammatory response and exposing the epithelium to repeated damage caused by carcinogenic toxins. This review aims to highlight the putative connection between the chronic colonization by highly pathogenic strains of S. Typhi capable of combining biofilm and toxin production and the onset of GC. Considering the high risk of GC associated with the asymptomatic carrier status, the rapid identification and profiling of biofilm production by S. Typhi strains would be key for effective therapeutic management and cancer prevention.


Subject(s)
Biofilms/growth & development , Gallbladder Neoplasms , Salmonella typhi/physiology , Typhoid Fever , Animals , Gallbladder Neoplasms/metabolism , Gallbladder Neoplasms/microbiology , Gallbladder Neoplasms/pathology , Gallbladder Neoplasms/prevention & control , Humans , Typhoid Fever/metabolism , Typhoid Fever/pathology , Typhoid Fever/therapy
18.
Proc Natl Acad Sci U S A ; 114(37): E7746-E7755, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28827342

ABSTRACT

Risk, severity, and outcome of infection depend on the interplay of pathogen virulence and host susceptibility. Systematic identification of genetic susceptibility to infection is being undertaken through genome-wide association studies, but how to expeditiously move from genetic differences to functional mechanisms is unclear. Here, we use genetic association of molecular, cellular, and human disease traits and experimental validation to demonstrate that genetic variation affects expression of VAC14, a phosphoinositide-regulating protein, to influence susceptibility to Salmonella enterica serovar Typhi (S Typhi) infection. Decreased VAC14 expression increased plasma membrane cholesterol, facilitating Salmonella docking and invasion. This increased susceptibility at the cellular level manifests as increased susceptibility to typhoid fever in a Vietnamese population. Furthermore, treating zebrafish with a cholesterol-lowering agent, ezetimibe, reduced susceptibility to S Typhi. Thus, coupling multiple genetic association studies with mechanistic dissection revealed how VAC14 regulates Salmonella invasion and typhoid fever susceptibility and may open doors to new prophylactic/therapeutic approaches.


Subject(s)
Membrane Proteins/genetics , Membrane Proteins/metabolism , Salmonella typhi/genetics , Cell Line, Tumor , Cholesterol/genetics , Cholesterol/metabolism , Ezetimibe , Genetic Variation/genetics , Genome-Wide Association Study , Humans , Intracellular Signaling Peptides and Proteins , Polymorphism, Single Nucleotide , Salmonella/genetics , Salmonella/pathogenicity , Salmonella typhi/metabolism , Salmonella typhi/pathogenicity , Typhoid Fever/metabolism , Typhoid Fever/physiopathology , Virulence/genetics
19.
Am J Trop Med Hyg ; 95(4): 938-944, 2016 Oct 05.
Article in English | MEDLINE | ID: mdl-27527624

ABSTRACT

The present controlled cross-sectional study aimed to assess elevated values of C-reactive protein (CRP), a positive acute-phase protein, induced by imported infectious diseases (IDs) seen in patients consulting the University of Munich (1999-2015) after being in the tropics/subtropics. The analysis investigated data sets from 11,079 diseased German travelers (cases) returning from Latin America (1,986), Africa (3,387), and Asia (5,706), and from 714 healthy Germans who had not recently traveled (controls). The proportions of elevated values of CRP (> 0.5 mg/dL) were significantly larger among cases (44.3%) than among controls (20.7%). Among cases, this proportion was largest among males (49.2%) in comparison to females (39.9%), among travelers with short travel duration of 1-14 days (49.6%) in comparison to travelers with a travel duration of > 180 days (30.8%), and with travel destination in Africa (47.0%) in comparison to Asia (44.2%) and Latin America (39.9%), among all-inclusive travelers (47.4%) in comparison to business travelers (46.7%) and backpackers (44.1%), and among patients presenting with fever (70.9%) and arthralgia (54.3%). The study identified various imported IDs with significantly larger proportions of elevated values of CRP including viral (cytomegalovirus infection [94.7%], influenza [88.9%], infectious mononucleosis [71.8%]), bacterial (typhoid fever [100%], paratyphoid fever [92.9%], shigellosis [76.8%], rickettsiosis [74.2%], Salmonella enteritis [71.3%], Campylobacter infection [68.7%]), and protozoan (vivax malaria [100%], ovale malaria [100%], falciparum malaria [95.4%], noninvasive Entamoeba infection [65.9%]) IDs. This study demonstrates that elevated values of CRP can be a useful laboratory finding for travelers returning from the tropics/subtropics, as these findings are typically caused mainly by certain imported bacterial IDs, but also by viral and protozoan IDs.


Subject(s)
Bacterial Infections/metabolism , C-Reactive Protein/metabolism , Parasitic Diseases/metabolism , Travel , Virus Diseases/metabolism , Adolescent , Adult , Africa , Aged , Aged, 80 and over , Asia , Campylobacter Infections/metabolism , Case-Control Studies , Child , Child, Preschool , Cross-Sectional Studies , Cytomegalovirus Infections/metabolism , Dysentery, Bacillary/metabolism , Entamoebiasis/metabolism , Enteritis/metabolism , Epstein-Barr Virus Infections/metabolism , Female , Germany , Humans , Infant , Influenza, Human/metabolism , Latin America , Malaria/metabolism , Male , Middle Aged , Paratyphoid Fever/metabolism , Rickettsia Infections/metabolism , Salmonella Infections/metabolism , Sex Factors , Typhoid Fever/metabolism , Young Adult
20.
Microb Pathog ; 85: 40-3, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26026834

ABSTRACT

The sufC gene of Escherichia coli (E. coli) is required for the biogenesis of iron-sulfur (Fe-S) cluster under oxidative stress conditions. In order to investigate the roles of sufC in Salmonella enterica serovar Typhi (S. Typhi), isogenic S. Typhi strain GIFU10007 harboring a non-polar mutation of sufC (ΔsufC) was constructed and the results showed that the sufC deleted mutant grew more slowly than the wild type strain when encounter oxidative stresses. Moreover, the deletion of sufC gene decreased S. Typhi survival within macrophages. After macrophages infected by sufC deleted mutant and wild type strain, we detected IL-6 and TNF-α released into the supernatant, and found the expression of IL-6 and TNF-α decreased in the supernatant of sufC deleted mutant infected groups than the wild type strain infected ones. In summary, our results showed that SufC may promote S. Typhi coping oxidative stress and help S. Typhi survival in macrophages.


Subject(s)
Adenosine Triphosphatases/metabolism , Bacterial Proteins/metabolism , Macrophages/microbiology , Salmonella typhi/enzymology , Salmonella typhi/growth & development , Typhoid Fever/microbiology , Adenosine Triphosphatases/genetics , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Macrophages/metabolism , Microbial Viability , Oxidative Stress , Salmonella typhi/genetics , Salmonella typhi/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Typhoid Fever/genetics , Typhoid Fever/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...