Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Front Immunol ; 12: 632154, 2021.
Article in English | MEDLINE | ID: mdl-34093525

ABSTRACT

The human gastric pathogen Helicobacter pylori activates human epithelial cells by a particular combination of mechanisms, including NOD1 and ALPK1-TIFA activation. These mechanisms are characterized by a strong participation of the bacterial cag pathogenicity island, which forms a type IV secretion system (CagT4SS) that enables the bacteria to transport proteins and diverse bacterial metabolites, including DNA, glycans, and cell wall components, into human host cells. Building on previous findings, we sought to determine the contribution of lipopolysaccharide inner core heptose metabolites (ADP-heptose) in the activation of human phagocytic cells by H. pylori. Using human monocyte/macrophage-like Thp-1 cells and human primary monocytes and macrophages, we were able to determine that a substantial part of early phagocytic cell activation, including NF-κB activation and IL-8 production, by live H. pylori is triggered by bacterial heptose metabolites. This effect was very pronounced in Thp-1 cells exposed to bacterial purified lysates or pure ADP-heptose, in the absence of other bacterial MAMPs, and was significantly reduced upon TIFA knock-down. Pure ADP-heptose on its own was able to strongly activate Thp-1 cells and human primary monocytes/macrophages. Comprehensive transcriptome analysis of Thp-1 cells co-incubated with live H. pylori or pure ADP-heptose confirmed a signature of ADP-heptose-dependent transcript activation in monocyte/macrophages. Bacterial enzyme-treated lysates (ETL) and pure ADP-heptose-dependent activation differentiated monocytes into macrophages of predominantly M1 type. In Thp-1 cells, the active CagT4SS was less required for the heptose-induced proinflammatory response than in epithelial cells, while active heptose biosynthesis or pure ADP-heptose was required and sufficient for their early innate response and NF-κB activation. The present data suggest that early activation and maturation of incoming and resident phagocytic cells (monocytes, macrophages) in the H. pylori-colonized stomach strongly depend on bacterial LPS inner core heptose metabolites, also with a significant contribution of an active CagT4SS.


Subject(s)
Genomic Islands/physiology , Helicobacter pylori/metabolism , Heptoses/metabolism , Macrophages/immunology , Monocytes/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Biosynthetic Pathways , Helicobacter pylori/pathogenicity , Humans , Immunity, Innate , Lipopolysaccharides/metabolism , Macrophage Activation , Macrophages/metabolism , Monocytes/metabolism , Signal Transduction , Transcriptome , Type IV Secretion Systems/genetics , Type IV Secretion Systems/metabolism
2.
J Bacteriol ; 202(17)2020 08 10.
Article in English | MEDLINE | ID: mdl-32571967

ABSTRACT

Salmonella enterica serovar Typhimurium uses a type three secretion system (T3SS) encoded on the Salmonella pathogenicity island 1 (SPI1) to invade intestinal epithelial cells and induce inflammatory diarrhea. The SPI1 T3SS is regulated by numerous environmental and physiological signals, integrated to either activate or repress invasion. Transcription of hilA, encoding the transcriptional activator of the SPI1 structural genes, is activated by three AraC-like regulators, HilD, HilC, and RtsA, that act in a complex feed-forward loop. Deletion of bamB, encoding a component of the ß-barrel assembly machinery, causes a dramatic repression of SPI1, but the mechanism was unknown. Here, we show that partially defective ß-barrel assembly activates the RcsCDB regulon, leading to decreased hilA transcription. This regulation is independent of RpoE activation. Though Rcs has been previously shown to repress SPI1 when disulfide bond formation is impaired, we show that activation of Rcs in a bamB background is dependent on the sensor protein RcsF, whereas disulfide bond status is sensed independently. Rcs decreases transcription of the flagellar regulon, including fliZ, the product of which indirectly activates HilD protein activity. Rcs also represses hilD, hilC, and rtsA promoters by an unknown mechanism. Both dsbA and bamB mutants have motility defects, though this is simply regulatory in a bamB background; motility is restored in the absence of Rcs. Effector secretion assays show that repression of SPI1 in a bamB background is also regulatory; if expressed, the SPI1 T3SS is functional in a bamB background. This emphasizes the sensitivity of SPI1 regulation to overall envelope homeostasis.IMPORTANCESalmonella causes worldwide foodborne illness, leading to massive disease burden and an estimated 600,000 deaths per year. Salmonella infects orally and invades intestinal epithelial cells using a type 3 secretion system that directly injects effector proteins into host cells. This first step in invasion is tightly regulated by a variety of inputs. In this work, we demonstrate that Salmonella senses the functionality of outer membrane assembly in determining regulation of invasion machinery, and we show that Salmonella uses distinct mechanisms to detect specific perturbations in envelope assembly.


Subject(s)
Genomic Islands/physiology , Salmonella typhimurium/physiology , Stress, Physiological , Type III Secretion Systems/physiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , RNA, Bacterial/genetics , RNA, Bacterial/metabolism
3.
Ann N Y Acad Sci ; 1447(1): 21-52, 2019 07.
Article in English | MEDLINE | ID: mdl-30900279

ABSTRACT

Conventional 20th century evolution thinking was based on the idea of isolated genomes for each species. Any possibility of life-history inputs to the germ line was strictly excluded by Weismann's doctrine, and genome change was attributed to random copying errors. Today, we know that many life-history events lead to rapid and nonrandom evolutionary change mediated by specific cellular functions. There are many ways that genomes, viruses, cells, and organisms interact to generate evolutionary variation. These include cell mergers and activation of natural genetic engineering by stress, infection, and interspecific hybridization. In addition, we know molecular mechanisms for transmitting life-history information across generations through gametes. These discoveries require a new agenda for evolutionary theory and novel experimental designs to investigate the genomic impacts of stresses, biotic interactions, and sensory inputs coming from the environment. The review will offer some generic recommendations for enriching evolution experiments to incorporate new knowledge and find answers to previously excluded questions.


Subject(s)
Evolution, Molecular , Genomic Islands/physiology , Animals , Genome/physiology , Humans , Microbiota/physiology
4.
Int J Med Microbiol ; 308(8): 1043-1052, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30466554

ABSTRACT

Salmonella pathogenicity island 13 (SPI-13) contributes to the virulence of Salmonella. The majority of the SPI-13 genes encode proteins putatively involved in bacterial metabolism, however, their functions largely remain uncharacterized. It is currently unknown if SPI-13 contributes to metabolic fitness of Salmonella and, if so, what are the metabolic substrates for the protein encoded by genes within SPI-13. We employed Phenotype Microarray (Biolog, USA) to compare the metabolic properties of SPI-13 deficient mutant (ΔSPI-13) and the WT parent strain of non-typhoidal Salmonella enterica sub sp. enterica serovar Enteritidis (S. Enteritidis). The results of Phenotype Microarray revealed that SPI-13 is required for efficient utilization of two micronutrients, namely, d-glucuronic acid (DGA) and tyramine (TYR), as sole sources of carbon and/or nitrogen. By systematic deletion of the individual gene(s), we identified specific genes within SPI-13 that are required for efficient utilization of DGA (SEN2977-80) and TYR (SEN2967 and SEN2971-72) as sole nutrient sources. The results show that SPI-13 mediated DGA and TYR metabolic pathways afford nutritional fitness to S. Enteritidis. Comparative genomics analysis of the SPI-13 locus from 247 Salmonella strains belonging to 57 different serovars revealed that SPI-13 genes specifically involved in the metabolism of DGA and TYR are highly conserved in Salmonella enterica. Because DGA and TYR are naturally present as metabolic byproducts in the gastrointestinal tract and other host tissues, we propose a metabolic model that shows that the role of SPI-13 mediated DGA and TYR metabolism in the nutritional fitness of Salmonella is likely linked to nutritional virulence of this pathogen.


Subject(s)
Genome, Bacterial/genetics , Genomic Islands/physiology , Salmonella enteritidis/genetics , Salmonella enteritidis/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Deletion , Genes, Bacterial/genetics , Genomic Islands/genetics , Glucuronic Acid/metabolism , Models, Biological , Models, Chemical , Salmonella enteritidis/growth & development , Salmonella enteritidis/pathogenicity , Serogroup , Tyramine/metabolism , Virulence
5.
Res Vet Sci ; 120: 41-46, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30199780

ABSTRACT

To investigate the effects of pathogenic Escherichia coli high pathogenicity island (HPI) on the expression of inflammatory factors via ubiquitin proteasome pathway. Firstly, the UBC-sus-263 shRNA plasmid was successfully established and transfected into porcine small intestine epithelial cells (IPEC-J2) by liposome to silence the ubiquitinntion gene. Then the IPEC-J2 was infected with E. coli HPI+ and HPI- strains, respectively. Finally, the mRNA of intracellular NF-κB and IκB-α,and the protein levels of NF-κB, IκB-α, TNF-α and IL-1 in IPEC-J2 cell line transfected with UBC-sus-263 shRNA (Ub-shRNA) were detected. The results showed that the Ub-shRNA was effectively inhibited ubiquitination pathway in the IPEC-J2 cell. After infected with HPI+, the mRNA and protein levels of NF-κB and IκB-α were dramatically decreased in Ub-hsRNA transfected IPEC-J2 cells compared to the control and HPI--infected groups. Consistently, the production of downstream cytokines such as TNF-α and IL-1 were highly expressed after HPI+-infection than that of HPI--infected groups. However, whether the HPI+ or HPI-, both could induce increasingly expression of NF-κB and IκB-α and its downstream cytokines in normal IPEC-J2 cells. Thus, the E. coli HPI can upregulate the expression of IκB-α to promote the releasing of TNF-α and IL-1 via the ubiquitination pathway.


Subject(s)
Epithelial Cells/metabolism , Escherichia coli/physiology , Genomic Islands/physiology , Intestine, Small/cytology , Proteasome Endopeptidase Complex/metabolism , Swine , Animals , Cell Line , Cytokines/genetics , Cytokines/metabolism , Epithelial Cells/microbiology , Gene Expression Regulation , Inflammation/metabolism , NF-kappa B/metabolism , Ubiquitins
6.
Cell Microbiol ; 20(2)2018 02.
Article in English | MEDLINE | ID: mdl-29156489

ABSTRACT

The human gastrointestinal tract is a complex ecosystem in which epithelial cells and microorganisms of the intestinal microbiota live in symbiosis. Certain members of the microbiota, in particular Escherichia coli strains of the B2 phylotype, carry the polyketide synthase-island encoding the genotoxin colibactin. Colibactin is a nonribosomal peptide or polyketide-nonribosomal peptide hybrid of still unsolved structure, which induces DNA double strand breaks (DSBs) in eukaryotic cells. However, direct contact between live bacteria and host cell is required in order to elicit these genotoxic effects. In this study, we used a variety of cell culture models, among them, a 3D cell culture approach based on decellularised small intestinal submucosa, to investigate whether the intestinal mucus layer has the potential to interfere with colibactin activity. We demonstrate that the expression of mucins and the formation of an adherent mucus layer significantly increased with increasing complexity of cell culture. Moreover, we show that the presence of an adherent mucus layer on epithelial cells attenuates the genotoxic activity of colibactin, by preventing the induction of DNA-DSBs. Removal of the adherent mucus layer restored the occurrence of DNA-DSBs.


Subject(s)
Gastrointestinal Tract/microbiology , Mucus/microbiology , Mutagens/metabolism , Peptides/metabolism , Polyketides/metabolism , Cell Line, Tumor , DNA Damage/physiology , Escherichia coli/pathogenicity , Gastrointestinal Microbiome/physiology , Genomic Islands/physiology , HT29 Cells , Humans , Symbiosis/physiology , Virulence/physiology
7.
Appl Environ Microbiol ; 84(2)2018 01 15.
Article in English | MEDLINE | ID: mdl-29079617

ABSTRACT

Two Gram-stain-positive, coagulase-negative staphylococcal strains were isolated from abiotic sources comprising stone fragments and sandy soil in James Ross Island, Antarctica. Here, we describe properties of a novel species of the genus Staphylococcus that has a 16S rRNA gene sequence nearly identical to that of Staphylococcus saprophyticus However, compared to S. saprophyticus and the next closest relatives, the new species demonstrates considerable phylogenetic distance at the whole-genome level, with an average nucleotide identity of <85% and inferred DNA-DNA hybridization of <30%. It forms a separate branch in the S. saprophyticus phylogenetic clade as confirmed by multilocus sequence analysis of six housekeeping genes, rpoB, hsp60, tuf, dnaJ, gap, and sod Matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS) and key biochemical characteristics allowed these bacteria to be distinguished from their nearest phylogenetic neighbors. In contrast to S. saprophyticus subsp. saprophyticus, the novel strains are pyrrolidonyl arylamidase and ß-glucuronidase positive and ß-galactosidase negative, nitrate is reduced, and acid produced aerobically from d-mannose. Whole-genome sequencing of the 2.69-Mb large chromosome revealed the presence of a number of mobile genetic elements, including the 27-kb pseudo-staphylococcus cassette chromosome mec of strain P5085T (ψSCCmecP5085), harboring the mecC gene, two composite phage-inducible chromosomal islands probably essential to adaptation to extreme environments, and one complete and one defective prophage. Both strains are resistant to penicillin G, ampicillin, ceftazidime, methicillin, cefoxitin, and fosfomycin. We hypothesize that antibiotic resistance might represent an evolutionary advantage against beta-lactam producers, which are common in a polar environment. Based on these results, a novel species of the genus Staphylococcus is described and named Staphylococcus edaphicus sp. nov. The type strain is P5085T (= CCM 8730T = DSM 104441T).IMPORTANCE The description of Staphylococcus edaphicus sp. nov. enables the comparison of multidrug-resistant staphylococci from human and veterinary sources evolved in the globalized world to their geographically distant relative from the extreme Antarctic environment. Although this new species was not exposed to the pressure of antibiotic treatment in human or veterinary practice, mobile genetic elements carrying antimicrobial resistance genes were found in the genome. The genomic characteristics presented here elucidate the evolutionary relationships in the Staphylococcus genus with a special focus on antimicrobial resistance, pathogenicity, and survival traits. Genes encoded on mobile genetic elements were arranged in unique combinations but retained conserved locations for the integration of mobile genetic elements. These findings point to enormous plasticity of the staphylococcal pangenome, shaped by horizontal gene transfer. Thus, S. edaphicus can act not only as a reservoir of antibiotic resistance in a natural environment but also as a mediator for the spread and evolution of resistance genes.


Subject(s)
Adaptation, Biological/genetics , Extreme Cold , Extreme Environments , Genes, Bacterial/physiology , Genomic Islands/physiology , Staphylococcus/classification , Antarctic Regions , Staphylococcus/genetics , Staphylococcus/physiology
8.
J Biol Chem ; 292(52): 21366-21380, 2017 12 29.
Article in English | MEDLINE | ID: mdl-29123034

ABSTRACT

Studies of virulence determinants in the bacterial phytopathogen Erwinia amylovora, the cause of devastating fire blight disease in apple and pear, have shown that HsvA, a putative amidinotransferase enzyme located in the Hrp pathogenicity island, is required for systemic infection in apple. However, the mechanism by which HsvA contributes to virulence is unclear. To investigate the role of HsvA in virulence, we carried out a series of biochemical and structural studies to characterize the amidinotransferase activity of HsvA. We found that HsvA displays a preference for linear aliphatic polyamines as the amidino acceptor substrate, especially for spermidine and putrescine (Km values of 33 µm and 3.9 mm, respectively). The three-dimensional structure, determined at 2.30 Å resolution using X-ray crystallography, revealed that the overall architecture of HsvA is similar to that of the human arginine-glycine amidinotransferase in the creatine biosynthesis pathway. The active site is located in the core of the protein at the base of a long, narrow substrate access channel. Specific amino acids near the entrance of the channel may serve as major determinants of the substrate specificity, including a glutamate residue at the rim of the channel entrance that appears to be positioned to interact with the distal primary amine in the putrescine substrate as well as the internal and distal amines in the spermidine substrate. These results suggest potential in vivo functions for HsvA as a virulence factor in fire blight and may also provide a basis for strategies to control fire blight by inhibiting HsvA activity.


Subject(s)
Amidinotransferases/metabolism , Erwinia amylovora/metabolism , Amidinotransferases/physiology , Crystallography, X-Ray/methods , Erwinia amylovora/pathogenicity , Genomic Islands/genetics , Genomic Islands/physiology , Malus/microbiology , Plant Diseases/microbiology , Polyamines/metabolism , Pyrus/microbiology , Virulence , Virulence Factors/metabolism
9.
Microbiology (Reading) ; 163(11): 1664-1679, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29034854

ABSTRACT

During conditions of nutrient limitation bacteria undergo a series of global gene expression changes to survive conditions of amino acid and fatty acid starvation. Rapid reallocation of cellular resources is brought about by gene expression changes coordinated by the signalling nucleotides' guanosine tetraphosphate or pentaphosphate, collectively termed (p)ppGpp and is known as the stringent response. The stringent response has been implicated in bacterial virulence, with elevated (p)ppGpp levels being associated with increased virulence gene expression. This has been observed in the highly pathogenic Francisella tularensis sub spp. tularensis SCHU S4, the causative agent of tularaemia. Here, we aimed to artificially induce the stringent response by culturing F. tularensis in the presence of the amino acid analogue l-serine hydroxamate. Serine hydroxamate competitively inhibits tRNAser aminoacylation, causing an accumulation of uncharged tRNA. The uncharged tRNA enters the A site on the translating bacterial ribosome and causes ribosome stalling, in turn stimulating the production of (p)ppGpp and activation of the stringent response. Using the essential virulence gene iglC, which is encoded on the Francisella pathogenicity island (FPI) as a marker of active stringent response, we optimized the culture conditions required for the investigation of virulence gene expression under conditions of nutrient limitation. We subsequently used whole genome RNA-seq to show how F. tularensis alters gene expression on a global scale during active stringent response. Key findings included up-regulation of genes involved in virulence, stress responses and metabolism, and down-regulation of genes involved in metabolite transport and cell division. F. tularensis is a highly virulent intracellular pathogen capable of causing debilitating or fatal disease at extremely low infectious doses. However, virulence mechanisms are still poorly understood. The stringent response is widely recognized as a diverse and complex bacterial stress response implicated in virulence. This work describes the global gene expression profile of F. tularensis SCHU S4 under active stringent response for the first time. Herein we provide evidence for an association of active stringent response with FPI virulence gene expression. Our results further the understanding of the molecular basis of virulence and regulation thereof in F. tularensis. These results also support research into genes involved in (p)ppGpp production and polyphosphate biosynthesis and their applicability as targets for novel antimicrobials.


Subject(s)
Adaptation, Biological/physiology , Francisella tularensis/physiology , Gene Expression Regulation, Bacterial/physiology , Genomic Islands/genetics , Transcriptome/physiology , Virulence/physiology , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Gene Expression Regulation, Bacterial/genetics , Genes, Bacterial/genetics , Genes, Regulator/genetics , Genes, Regulator/physiology , Genomic Islands/physiology , High-Throughput Nucleotide Sequencing , Metabolic Networks and Pathways/genetics , Metabolic Networks and Pathways/physiology , Oxidative Stress/genetics , Oxidative Stress/physiology , Proteome/physiology , Sequence Analysis, RNA , Serine/analogs & derivatives , Serine/toxicity , Stress, Physiological , Transcriptional Activation/genetics , Transcriptional Activation/physiology , Transcriptome/genetics , Virulence/genetics
10.
Biol Res ; 50(1): 5, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-28202086

ABSTRACT

BACKGROUND: Salmonella pathogenicity island (SPI)-13 is conserved in many serovars of S. enterica, including S. Enteritidis, S. Typhimurium and S. Gallinarum. However, it is absent in typhoid serovars such as S. Typhi and Paratyphi A, which carry SPI-8 at the same genomic location. Because the interaction with macrophages is a critical step in Salmonella pathogenicity, in this study we investigated the role played by SPI-13 and SPI-8 in the interaction of S. Enteritidis and S. Typhi with cultured murine (RAW264.7) and human (THP-1) macrophages. RESULTS: Our results showed that SPI-13 was required for internalization of S. Enteritidis in murine but not human macrophages. On the other hand, SPI-8 was not required for the interaction of S. Typhi with human or murine macrophages. Of note, the presence of an intact copy of SPI-13 in a S. Typhi mutant carrying a deletion of SPI-8 did not improve its ability to be internalized by, or survive in human or murine macrophages. CONCLUSIONS: Altogether, our results point out to different roles for SPI-13 and SPI-8 during Salmonella infection. While SPI-13 contributes to the interaction of S. Enteritidis with murine macrophages, SPI-8 is not required in the interaction of S. Typhi with murine or human macrophages. We hypothesized that typhoid serovars have lost SPI-13 and maintained SPI-8 to improve their fitness during another phase of human infection.


Subject(s)
Genomic Islands/physiology , Macrophages/microbiology , Salmonella Infections/microbiology , Salmonella enteritidis/genetics , Salmonella typhi/genetics , Analysis of Variance , Animals , Bacterial Physiological Phenomena , Cell Survival , Cells, Cultured , Genome, Bacterial , Genomic Islands/genetics , Humans , Mice , Microbial Interactions/genetics , Muridae , Polymerase Chain Reaction , RAW 264.7 Cells , Serogroup , Species Specificity
11.
Biol. Res ; 50: 5, 2017. tab, graf
Article in English | LILACS | ID: biblio-838972

ABSTRACT

BACKGROUND: Salmonella pathogenicity island (SPI)-13 is conserved in many serovars of S. enterica, including S. Enteritidis, S. Typhimurium and S. Gallinarum. However, it is absent in typhoid serovars such as S. Typhi and Paratyphi A, which carry SPI-8 at the same genomic location. Because the interaction with macrophages is a critical step in Salmonella pathogenicity, in this study we investigated the role played by SPI-13 and SPI-8 in the interaction of S. Enteritidis and S. Typhi with cultured murine (RAW264.7) and human (THP-1) macrophages. RESULTS: Our results showed that SPI-13 was required for internalization of S. Enteritidis in murine but not human macrophages. On the other hand, SPI-8 was not required for the interaction of S. Typhi with human or murine macrophages. Of note, the presence of an intact copy of SPI-13 in a S. Typhi mutant carrying a deletion of SPI-8 did not improve its ability to be internalized by, or survive in human or murine macrophages. CONCLUSIONS: Altogether, our results point out to different roles for SPI-13 and SPI-8 during Salmonella infection. While SPI-13 contributes to the interaction of S. Enteritidis with murine macrophages, SPI-8 is not required in the interaction of S. Typhi with murine or human macrophages. We hypothesized that typhoid serovars have lost SPI-13 and maintained SPI-8 to improve their fitness during another phase of human infection.


Subject(s)
Humans , Animals , Mice , Salmonella enteritidis/genetics , Salmonella Infections/microbiology , Salmonella typhi/genetics , Genomic Islands/physiology , Macrophages/microbiology , Species Specificity , Cell Survival , Cells, Cultured , Polymerase Chain Reaction , Analysis of Variance , Genome, Bacterial , Bacterial Physiological Phenomena , Genomic Islands/genetics , Microbial Interactions/genetics , Serogroup , RAW 264.7 Cells , Muridae
12.
J Bacteriol ; 198(5): 766-76, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26668266

ABSTRACT

UNLABELLED: Pathogenicity islands (PAIs) are mobile integrated genetic elements (MIGEs) that contain a diverse range of virulence factors and are essential in the evolution of pathogenic bacteria. PAIs are widespread among bacteria and integrate into the host genome, commonly at a tRNA locus, via integrase-mediated site-specific recombination. The excision of PAIs is the first step in the horizontal transfer of these elements and is not well understood. In this study, we examined the role of recombination directionality factors (RDFs) and their relationship with integrases in the excision of two PAIs essential for Vibrio cholerae host colonization: Vibrio pathogenicity island 1 (VPI-1) and VPI-2. VPI-1 does not contain an RDF, which allowed us to answer the question of whether RDFs are an absolute requirement for excision. We found that an RDF was required for efficient excision of VPI-2 but not VPI-1 and that RDFs can induce excision of both islands. Expression data revealed that the RDFs act as transcriptional repressors to both VPI-1- and VPI-2-encoded integrases. We demonstrated that the RDFs Vibrio excision factor A (VefA) and VefB bind at the attachment sites (overlapping the int promoter region) of VPI-1 and VPI-2, thus supporting this mode of integrase repression. In addition, V. cholerae RDFs are promiscuous due to their dual functions of promoting excision of both VPI-1 and VPI-2 and acting as negative transcriptional regulators of the integrases. This is the first demonstration of cross talk between PAIs mediated via RDFs which reveals the complex interactions that occur between separately acquired MIGEs. IMPORTANCE: Deciphering the mechanisms of pathogenicity island excision is necessary for understanding the evolution and spread of these elements to their nonpathogenic counterparts. Such mechanistic insight would assist in predicting the mobility of uncharacterized genetic elements. This study identified extensive RDF-mediated cross talk between two nonhomologous VPIs and demonstrated the dual functionality of RDF proteins: (i) inducing PAI excision and (ii) acting as transcriptional regulators. Findings from this study may be implicated in determining the mobilome contribution of other bacteria with multiple MIGEs.


Subject(s)
Chromosomes, Bacterial/genetics , Genomic Islands/physiology , Recombination, Genetic/physiology , Vibrio cholerae/genetics , Vibrio cholerae/physiology , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Escherichia coli , Gene Expression Regulation, Bacterial/physiology , Genomic Islands/genetics , Mutation , Plasmids , Vibrio cholerae/classification
13.
Nihon Saikingaku Zasshi ; 69(4): 577-88, 2014.
Article in Japanese | MEDLINE | ID: mdl-25447983

ABSTRACT

Chromobacterium violaceum is a Gram-negative bacterium that infects humans and animals with fatal sepsis. The infection with C. violaceum is rare in case of those who are healthy, but once established, C. violaceum causes sever disease accompanied by abscess formation in the lungs, liver and spleen. Furthermore, C. violaceum is resistant to a broad range of antibiotics, which in some cases renders the antimicrobial therapy for this infection difficult. Thus, the infection with C. violaceum displays high mortality rates unless initial proper antimicrobial therapy. In contrast, the infection mechanism had completely remained unknown. To this end, we have tried to identify virulence factors-associated with C. violaceum infection. Two distinct type III secretion systems (TTSSs) were thought to be one of the most important virulence factors, which are encoded by Chromobacterium pathogenicity island 1/1a and 2 (Cpi-1/-1a and -2) respectively. Our results have shown that Cpi-1/-1a-encoded TTSS, but not Cpi-2, is indispensable for the virulence in a mouse infection model. C. violaceum caused fulminant hepatitis in a Cpi-1/-1a-encoded TTSS-dependent manner. We next have identified 16 novel effectors secreted from Cpi-1/-1a-encoded TTS machinery. From these effectors, we found that CopE (Chromobacterium outer protein E) has similarities to a guanine nucleotide exchange factor (GEF) for Rho GTPases. CopE acts as GEF for Rac1 and Cdc42, leading to induction of actin cytoskeletal rearrangement. Interestingly, C. violaceum invades cultured human epithelial cells in a CopE-dependent manner. Finally, an inactivation of CopE by disruption of copE gene or amino acid point mutation leading to loss of GEF activity attenuates significantly the mouse virulence of C. violaceum. These results suggest that Cpi-1/-1a-encoded TTSS is a major virulence determinant for C. violaceum infection, and that CopE contributes to the virulence in part of this pathogen.


Subject(s)
Chromobacterium/genetics , Chromobacterium/pathogenicity , Genomic Islands/genetics , Genomic Islands/physiology , Gram-Negative Bacterial Infections/microbiology , Animals , Disease Models, Animal , Humans , Mice , Multigene Family/genetics , Virulence , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism
14.
PLoS Pathog ; 10(11): e1004500, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25375226

ABSTRACT

The bacterial H-NS protein silences expression from sequences with higher AT-content than the host genome and is believed to buffer the fitness consequences associated with foreign gene acquisition. Loss of H-NS results in severe growth defects in Salmonella, but the underlying reasons were unclear. An experimental evolution approach was employed to determine which secondary mutations could compensate for the loss of H-NS in Salmonella. Six independently derived S. Typhimurium hns mutant strains were serially passaged for 300 generations prior to whole genome sequencing. Growth rates of all lineages dramatically improved during the course of the experiment. Each of the hns mutant lineages acquired missense mutations in the gene encoding the H-NS paralog StpA encoding a poorly understood H-NS paralog, while 5 of the mutant lineages acquired deletions in the genes encoding the Salmonella Pathogenicity Island-1 (SPI-1) Type 3 secretion system critical to invoke inflammation. We further demonstrate that SPI-1 misregulation is a primary contributor to the decreased fitness in Salmonella hns mutants. Three of the lineages acquired additional loss of function mutations in the PhoPQ virulence regulatory system. Similarly passaged wild type Salmonella lineages did not acquire these mutations. The stpA missense mutations arose in the oligomerization domain and generated proteins that could compensate for the loss of H-NS to varying degrees. StpA variants most able to functionally substitute for H-NS displayed altered DNA binding and oligomerization properties that resembled those of H-NS. These findings indicate that H-NS was central to the evolution of the Salmonellae by buffering the negative fitness consequences caused by the secretion system that is the defining characteristic of the species.


Subject(s)
Bacterial Proteins , DNA-Binding Proteins , Evolution, Molecular , Gene Expression Regulation, Bacterial/physiology , Gene Silencing/physiology , Genomic Islands/physiology , Salmonella , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Mutation , Salmonella/genetics , Salmonella/metabolism
15.
J Theor Biol ; 363: 188-97, 2014 Dec 21.
Article in English | MEDLINE | ID: mdl-25128737

ABSTRACT

BACKGROUND: Serovars of Salmonella enterica, namely Typhi and Typhimurium, reportedly, are the bacterial pathogens causing systemic infections like gastroenteritis and typhoid fever. To elucidate the role and importance in such infection, the proteins of the Type III secretion system of Salmonella pathogenicity islands and two component signal transduction systems, have been mainly focused. However, the most indispensable of these virulent ones and their hierarchical role has not yet been studied extensively. RESULTS: We have adopted a theoretical approach to build an interactome comprising the proteins from the Salmonella pathogeneicity islands (SPI) and two component signal transduction systems. This interactome was then analyzed by using network parameters like centrality and k-core measures. An initial step to capture the fingerprint of the core network resulted in a set of proteins which are involved in the process of invasion and colonization, thereby becoming more important in the process of infection. These proteins pertained to the Inv, Org, Prg, Sip, Spa, Ssa and Sse operons along with chaperone protein SicA. Amongst them, SicA was figured out to be the most indispensable protein from different network parametric analyses. Subsequently, the gene expression levels of all these theoretically identified important proteins were confirmed by microarray data analysis. Finally, we have proposed a hierarchy of the proteins involved in the total infection process. This theoretical approach is the first of its kind to figure out potential virulence determinants encoded by SPI for therapeutic targets for enteric infection. CONCLUSIONS: A set of responsible virulent proteins was identified and the expression level of their genes was validated by using independent, published microarray data. The result was a targeted set of proteins that could serve as sensitive predictors and form the foundation for a series of trials in the wet-lab setting. Understanding these regulatory and virulent proteins would provide insight into conditions which are encountered by this intracellular enteric pathogen during the course of infection. This would further contribute in identifying novel targets for antimicrobial agents.


Subject(s)
Bacterial Secretion Systems/genetics , Genomic Islands/physiology , Protein Interaction Mapping/methods , Salmonella/metabolism , Salmonella/pathogenicity , Signal Transduction/physiology , Bacterial Proteins/metabolism , Gene Regulatory Networks/genetics , Microarray Analysis , Molecular Chaperones/metabolism , Salmonella/genetics
16.
Infect Immun ; 82(6): 2203-10, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24643535

ABSTRACT

In order to survive inside macrophages, Salmonella produces a series of proteins encoded by genes within Salmonella pathogenicity island 2 (SPI-2). In the present study, we report that Fur, a central regulator of iron utilization, negatively controls the expression of SPI-2 genes. Time course analysis of SPI-2 expression after the entry of Salmonella into macrophages revealed that SPI-2 genes are induced earlier and at higher levels in the absence of the Fur regulator. It was hypothesized that Fur repressed the SPI-2 expression that was activated during acidification of the phagosome. Indeed, as pH was lowered from pH 7.0 to pH 5.5, the lack of Fur enabled SPI-2 gene expression to be induced at higher pH and to be expressed at higher levels. Fur controlled SPI-2 genes via repression of the SsrB response regulator, a primary activator of SPI-2 expression. Fur repressed ssrB expression both inside macrophages and under acidic conditions, which we ascribe to the direct binding of Fur to the ssrB promoter. Our study suggests that Salmonella could employ iron inside the phagosome to precisely control the timing and levels of SPI-2 expression inside macrophages.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Proteins/physiology , Gene Expression Regulation, Bacterial , Genomic Islands/physiology , Membrane Proteins/metabolism , Repressor Proteins/physiology , Salmonella typhimurium/pathogenicity , Gene Expression Regulation, Bacterial/genetics , Genomic Islands/genetics , Hydrogen-Ion Concentration , Iron/metabolism , Macrophages/microbiology , Salmonella typhimurium/genetics , Transcription Factors/metabolism
17.
PLoS Pathog ; 10(1): e1003874, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24415940

ABSTRACT

Expression of genes of the locus of enterocyte effacement (LEE) is essential for adherence of enterohemorrhagic Escherichia coli (EHEC) to intestinal epithelial cells. Gut factors that may modulate LEE gene expression may therefore influence the outcome of the infection. Because nitric oxide (NO) is a critical effector of the intestinal immune response that may induce transcriptional regulation in enterobacteria, we investigated its influence on LEE expression in EHEC O157:H7. We demonstrate that NO inhibits the expression of genes belonging to LEE1, LEE4, and LEE5 operons, and that the NO sensor nitrite-sensitive repressor (NsrR) is a positive regulator of these operons by interacting directly with the RNA polymerase complex. In the presence of NO, NsrR detaches from the LEE1/4/5 promoter regions and does not activate transcription. In parallel, two regulators of the acid resistance pathway, GadE and GadX, are induced by NO through an indirect NsrR-dependent mechanism. In this context, we show that the NO-dependent LEE1 down-regulation is due to absence of NsrR-mediated activation and to the repressor effect of GadX. Moreover, the inhibition of expression of LEE4 and LEE5 by NO is due to loss of NsrR-mediated activation, to LEE1 down-regulation and to GadE up-regulation. Lastly, we establish that chemical or cellular sources of NO inhibit the adherence of EHEC to human intestinal epithelial cells. These results highlight the critical effect of NsrR in the regulation of the LEE pathogenicity island and the potential role of NO in the limitation of colonization by EHEC.


Subject(s)
AraC Transcription Factor/biosynthesis , DNA-Binding Proteins/biosynthesis , Escherichia coli O157/metabolism , Escherichia coli Proteins/biosynthesis , Genomic Islands/physiology , Nitric Oxide/metabolism , Transcription Factors/biosynthesis , AraC Transcription Factor/genetics , Bacterial Adhesion/physiology , DNA-Binding Proteins/genetics , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Escherichia coli O157/genetics , Escherichia coli Proteins/genetics , Gene Expression Regulation, Bacterial/physiology , HeLa Cells , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Transcription Factors/genetics
18.
Mol Genet Genomics ; 288(11): 549-57, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23933962

ABSTRACT

DT104 emerged as a new branch of Salmonella typhimurium with resistance to multiple antimicrobials. To reveal some general genomic features of DT104 for clues of evolutionary events possibly associated with the emergence of this relatively new type of this pathogen, we mapped 11 independent DT104 strains and compared them with non-DT104 S. typhimurium strains. We found that all 11 DT104 strains contained three insertions absent in non-DT104 strains, i.e., the previously reported ST104, ST104B and ST64B. However, SGI-1, a genomic island known to be responsible for DT104 multidrug resistance, was not present in all DT104 strains examined in this study: one DT104 strain did not contain SGI-1 but carried a 144 kb plasmid, suggesting possible evolutionary relationships between the two DNA elements in the development of antimicrobial resistance.


Subject(s)
Genome, Bacterial/genetics , Genomics , Salmonella Infections/microbiology , Salmonella typhimurium/genetics , Biological Evolution , Chromosome Mapping , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Deoxyribonucleases, Type II Site-Specific , Drug Resistance, Multiple, Bacterial/genetics , Endodeoxyribonucleases , Gene Rearrangement , Genomic Islands/physiology , Plasmids/genetics , Species Specificity
19.
Infect Immun ; 81(6): 2076-84, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23529623

ABSTRACT

Modulation of host cell death pathways appears to be a prerequisite for the successful lifestyles of many intracellular pathogens. The facultative intracellular bacterium Francisella tularensis is highly pathogenic, and effective proliferation in the macrophage cytosol leading to host cell death is a requirement for its virulence. To better understand the prerequisites of this cell death, macrophages were infected with the F. tularensis live vaccine strain (LVS), and the effects were compared to those resulting from infections with deletion mutants lacking expression of either of the pdpC, iglC, iglG, or iglI genes, which encode components of the Francisella pathogenicity island (FPI), a type VI secretion system. Within 12 h, a majority of the J774 cells infected with the LVS strain showed production of mitochondrial superoxide and, after 24 h, marked signs of mitochondrial damage, caspase-9 and caspase-3 activation, phosphatidylserine expression, nucleosome formation, and membrane leakage. In contrast, neither of these events occurred after infection with the ΔiglI or ΔiglC mutants, although the former strain replicated. The ΔiglG mutant replicated effectively but induced only marginal cytopathogenic effects after 24 h and intermediate effects after 48 h. In contrast, the ΔpdpC mutant showed no replication but induced marked mitochondrial superoxide production and mitochondrial damage, caspase-3 activation, nucleosome formation, and phosphatidylserine expression, although the effects were delayed compared to those obtained with LVS. The unique phenotypes of the mutants provide insights regarding the roles of individual FPI components for the modulation of the cytopathogenic effects resulting from the F. tularensis infection.


Subject(s)
Bacterial Proteins/metabolism , Francisella tularensis/physiology , Genomic Islands/physiology , Macrophages/microbiology , Animals , Annexin A5/metabolism , Bacterial Proteins/genetics , Caspase 9/metabolism , Cell Death/physiology , Cell Line , Cytokines/genetics , Cytokines/metabolism , Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Gene Expression Regulation, Bacterial/physiology , Genomic Islands/genetics , Macrophages/metabolism , Macrophages/pathology , Mice , Mutation , Virulence
20.
Infect Immun ; 80(6): 1996-2007, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22493086

ABSTRACT

The enteropathogen Salmonella enterica serovar Typhimurium employs a suite of tightly regulated virulence factors within the intracellular compartment of phagocytic host cells resulting in systemic dissemination in mice. A type VI secretion system (T6SS) within Salmonella pathogenicity island 6 (SPI-6) has been implicated in this process; however, the regulatory inputs and the roles of noncore genes in this system are not well understood. Here we describe four clusters of noncore T6SS genes in SPI-6 based on a comparative relationship with the T6SS-3 of Burkholderia mallei and report that the disruption of these genes results in defects in intracellular replication and systemic dissemination in mice. In addition, we show that the expression of the SPI-6-encoded Hcp and VgrG orthologs is enhanced during late stages of macrophage infection. We identify six regions that are transcriptionally active during cell infections and that have regulatory contributions from the regulators of virulence SsrB, PhoP, and SlyA. We show that levels of protein expression are very weak under in vitro conditions and that expression is not enhanced upon the deletion of ssrB, phoP, slyA, qseC, ompR, or hfq, suggesting an unknown activating factor. These data suggest that the SPI-6 T6SS has been integrated into the Salmonella Typhimurium virulence network and customized for host-pathogen interactions through the action of noncore genes.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial/physiology , Genomic Islands/physiology , Multigene Family , Salmonella typhimurium/pathogenicity , Animals , Bacterial Proteins/genetics , Cell Line , Female , Genomic Islands/genetics , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...