Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Elife ; 122023 10 11.
Article in English | MEDLINE | ID: mdl-37819270

ABSTRACT

Individual species of bacteria and yeast present in the food of wild fruit flies work together to provide the nutrients needed for larval growth.


Subject(s)
Drosophila melanogaster , Microbiota , Animals , Drosophila , Nutrients
2.
Front Cell Infect Microbiol ; 12: 898125, 2022.
Article in English | MEDLINE | ID: mdl-35909953

ABSTRACT

In Brazil, the production of KPC-type carbapenemases in Enterobacteriales is endemic, leading to widespread use of polymyxins. In the present study, 502 Klebsiella pneumoniae isolates were evaluated for resistance to polymyxins, their genetic determinants and clonality, in addition to the presence of carbapenem resistance genes and evaluation of antimicrobial resistance. Resistance to colistin (polymyxin E) was evaluated through initial selection on EMB agar containing 4% colistin sulfate, followed by Minimal Inhibitory Concentration (MIC) determination by broth microdilution. The susceptibility to 17 antimicrobials was assessed by disk diffusion. The presence of blaKPC, blaNDM and blaOXA-48-like carbapenemases was investigated by phenotypic methods and conventional PCR. Molecular typing was performed by PFGE and MLST. Allelic variants of the mcr gene were screened by PCR and chromosomal mutations in the pmrA, pmrB, phoP, phoQ and mgrB genes were investigated by sequencing. Our work showed a colistin resistance frequency of 29.5% (n = 148/502) in K. pneumoniae isolates. Colistin MICs from 4 to >128 µg/mL were identified (MIC50 = 64 µg/mL; MIC90 >128 µg/mL). All isolates were considered MDR, with the lowest resistance rates observed for amikacin (34.4%), and 19.6% of the isolates were resistant to all tested antimicrobials. The blaKPC gene was identified in 77% of the isolates, in consonance with the high rate of resistance to polymyxins related to its use as a therapeutic alternative. Through XbaI-PFGE, 51 pulsotypes were identified. MLST showed 21 STs, with ST437, ST258 and ST11 (CC11) being the most prevalent, and two new STs were determined: ST4868 and ST4869. The mcr-1 gene was identified in 3 K. pneumoniae isolates. Missense mutations in chromosomal genes were identified, as well as insertion sequences in mgrB. Furthermore, the identification of chromosomal mutations in K. pneumoniae isolates belonging from CC11 ensures its success as a high-risk epidemic clone in Brazil and worldwide.


Subject(s)
Anti-Bacterial Agents , Colistin , Drug Resistance, Bacterial , Klebsiella Infections , Klebsiella pneumoniae , beta-Lactamases , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Brazil , Colistin/pharmacology , Colistin/therapeutic use , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Humans , Klebsiella Infections/epidemiology , Klebsiella Infections/genetics , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/isolation & purification , Microbial Sensitivity Tests , Multilocus Sequence Typing , Polymyxins/adverse effects , Polymyxins/pharmacology , Polymyxins/therapeutic use , beta-Lactamases/genetics , beta-Lactamases/therapeutic use
3.
Crit Rev Microbiol ; 47(4): 397-434, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33751923

ABSTRACT

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


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Salmonella Infections/microbiology , Salmonella enterica/metabolism , Salmonella enterica/pathogenicity , Animals , Bacterial Proteins/genetics , Ecosystem , Humans , Salmonella enterica/genetics , Signal Transduction , Virulence
4.
Tuberculosis (Edinb) ; 126: 102043, 2021 01.
Article in English | MEDLINE | ID: mdl-33370646

ABSTRACT

Although treatable with antibiotics, tuberculosis is a leading cause of death. Mycobacterium tuberculosis antibiotic resistance is becoming increasingly common and disease control is challenging. Conventional drug susceptibility testing takes weeks to produce results, and treatment is often initiated empirically. Therefore, new methods to determine drug susceptibility profiles are urgent. Here, we used mass-spectrometry-based metabolomics to characterize the metabolic landscape of drug-susceptible (DS), multidrug-resistant (MDR) and extensively drug-resistant (XDR) M. tuberculosis. Direct infusion mass spectrometry data showed that DS, MDR, and XDR strains have distinct metabolic profiles, which can be used to predict drug susceptibility and resistance. This was later confirmed by Ultra-High-Performance Liquid Chromatography and High-Resolution Mass Spectrometry, where we found that levels of ions presumptively identified as isoleucine, proline, hercynine, betaine, and pantothenic acid varied significantly between strains with different drug susceptibility profiles. We then confirmed the identification of proline and isoleucine and determined their absolute concentrations in bacterial extracts, and found significantly higher levels of these amino acids in DS strains, as compared to drug-resistant strains (combined MDR and XDR strains). Our results advance the current understanding of the effect of drug resistance on bacterial metabolism and open avenues for the detection of drug resistance biomarkers.


Subject(s)
Antitubercular Agents/pharmacology , Extensively Drug-Resistant Tuberculosis/metabolism , Metabolome/physiology , Metabolomics/methods , Mycobacterium tuberculosis/metabolism , Extensively Drug-Resistant Tuberculosis/drug therapy , Extensively Drug-Resistant Tuberculosis/microbiology , Humans , Microbial Sensitivity Tests , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/isolation & purification
5.
Tuberculosis (Edinb) ; 113: 163-174, 2018 12.
Article in English | MEDLINE | ID: mdl-30514498

ABSTRACT

Tuberculosis patients taking second line drugs such as ethionamide (ETH) have often experienced previous treatment failure and usually have a complex history of disease and treatment that can span decades. Mutations in the ETH activating enzyme, EthA, confer resistance through undescribed mechanisms. To explore the impact of EthA mutations on ETH resistance, data from a total of 160 ETHR isolates was analysed. The most frequently mutated positions are within regions that display sequence conservation with the active site of OTEMO, another FAD-containing NADH-binding Baeyer-Villiger monooxygenase (BVMO), or with the sugar binding site of galectin-4N. Additionally, to look at a possible role of EthR on ETH resistance we purified an EthR mutant identified in a clinical isolate, F110L, and found it to bind the ethA-ethR intergenic region with higher affinity than the wild type regulator in gel shift assays. The ability of cyclic di-GMP to enhance DNA binding is maintained in the EthR mutant. To our knowledge, this is the first ETH resistance study that combines sequence and resistance data of clinical isolates with functional and structural information.


Subject(s)
Antitubercular Agents/therapeutic use , DNA, Bacterial/genetics , Drug Resistance, Bacterial/genetics , Ethionamide/therapeutic use , Genetic Loci , Mycobacterium tuberculosis/genetics , Tuberculosis/microbiology , Binding Sites , DNA, Bacterial/isolation & purification , Genotype , Humans , Mutation , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/isolation & purification , Oxidoreductases/genetics , Phenotype , Protein Binding , Protein Conformation , Repressor Proteins/genetics , Structure-Activity Relationship , Tuberculosis/diagnosis , Tuberculosis/drug therapy
6.
Bio Protoc ; 8(8): e2808, 2018 Apr 20.
Article in English | MEDLINE | ID: mdl-34286025

ABSTRACT

The human body is colonized by vast communities of microbes, collectively known as microbiota, or microbiome. Although microbes colonize every surface of our bodies that is exposed to the external environment, the biggest collection of microbes colonizing humans and other mammals can be found in the gastrointestinal tract. Given the fact that the human gut is colonized by several hundred microbial species, our group hypothesized that the chemical diversity of this environment should be significant, and that many of the molecules present in that environment would have important signaling roles. Therefore, we devised a protocol to extract these molecules from human feces and test their signaling properties. Potentially bioactive extracts can be tested through addition to culture medium and analyses of bacterial growth and gene expression, among other properties. The protocol described herein provides an easy and rapid method for the extraction and testing of metabolites from fecal samples using Salmonella enterica as a model organism. This protocol can also be adapted to the extraction of small molecules from other matrices, such as cultured mammalian cells, tissues, body fluids, and axenic microbial cultures, and the resulting extracts can be tested against various microbial species.

7.
Tuberculosis (Edinb) ; 107: 59-62, 2017 12.
Article in English | MEDLINE | ID: mdl-29050773

ABSTRACT

Tuberculosis is a major public health concern, and diagnostic strategies applied to animal populations are scarce. As part of ongoing efforts to control tuberculosis dissemination at our animal facility, two non-human primates (NHP, Saimiri sciureus) presenting cutaneous lesions were examined for mycobacterial infection. Both animals tested positive for acid-fast bacilli and Mycobacterium tuberculosis using a molecular assay (IS6110 PCR). Animals were euthanized and several samples were tested for M. tuberculosis using the Xpert MTB/RIF assay. Many samples were positive for M. tuberculosis and rifampicin resistance, and some produced mycobacterial growth. Oral swabs from cage mates were then tested with Xpert MTB/RIF, and the majority tested positive for M. tuberculosis and rifampicin resistance, and produced growth in culture. To our knowledge, this is the first report of multidrug-resistant mycobacterial infection in NHP. Additionally, our data shows that the Xpert MTB/RIF assay can be useful as a screening tool for tuberculosis infection in NHP.


Subject(s)
Bacteriological Techniques/veterinary , DNA, Bacterial/genetics , Monkey Diseases/diagnosis , Mycobacterium tuberculosis/genetics , Polymerase Chain Reaction/veterinary , Saimiri/microbiology , Tuberculosis, Cutaneous/veterinary , Tuberculosis, Multidrug-Resistant/veterinary , Animals , Antitubercular Agents/pharmacology , DNA, Bacterial/isolation & purification , Drug Resistance, Multiple, Bacterial/genetics , Genotype , Monkey Diseases/drug therapy , Monkey Diseases/microbiology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/isolation & purification , Predictive Value of Tests , Rifampin/pharmacology , Tuberculosis, Cutaneous/diagnosis , Tuberculosis, Cutaneous/drug therapy , Tuberculosis, Cutaneous/microbiology , Tuberculosis, Multidrug-Resistant/diagnosis , Tuberculosis, Multidrug-Resistant/drug therapy , Tuberculosis, Multidrug-Resistant/microbiology
8.
Appl Environ Microbiol ; 83(19)2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28754707

ABSTRACT

The human microbiome is a collection of microorganisms that inhabit every surface of the body that is exposed to the environment, generally coexisting peacefully with their host. These microbes have important functions, such as producing vitamins, aiding in maturation of the immune system, and protecting against pathogens. We have previously shown that a small-molecule extract from the human fecal microbiome has a strong repressive effect on Salmonella enterica serovar Typhimurium host cell invasion by modulating the expression of genes involved in this process. Here, we describe the characterization of this biological activity. Using a series of purification methods, we obtained fractions with biological activity and characterized them by mass spectrometry. These experiments revealed an abundance of aromatic compounds in the bioactive fraction. Selected compounds were obtained from commercial sources and tested with respect to their ability to repress the expression of hilA, the gene encoding the master regulator of invasion genes in Salmonella We found that the aromatic compound 3,4-dimethylbenzoic acid acts as a strong inhibitor of hilA expression and of invasion of cultured host cells by Salmonella Future studies should reveal the molecular details of this phenomenon, such as the signaling cascades involved in sensing this bioactive molecule.IMPORTANCE Microbes constantly sense and adapt to their environment. Often, this is achieved through the production and sensing of small extracellular molecules. The human body is colonized by complex communities of microbes, and, given their biological and chemical diversity, these ecosystems represent a platform where the production and sensing of molecules occur. In previous work, we showed that small molecules produced by microbes from the human gut can significantly impair the virulence of the enteric pathogen Salmonella enterica Here, we describe a specific compound from the human gut that produces this same effect. The results from this work not only shed light on an important biological phenomenon occurring in our bodies but also may represent an opportunity to develop drugs that can target these small-molecule interactions to protect us from enteric infections and other diseases.

9.
PLoS One ; 11(7): e0159676, 2016.
Article in English | MEDLINE | ID: mdl-27437699

ABSTRACT

Salmonella enterica serovar Typhimurium (S. Typhimurium) is a foodborne enteric pathogen and a major cause of gastroenteritis in humans. It is known that molecules derived from the human fecal microbiota downregulate S. Typhimurium virulence gene expression and induce a starvation-like response. In this study, S. Typhimurium was cultured in minimal media to mimic starvation conditions such as that experienced by S. Typhimurium in the human intestinal tract, and the pathogen's virulence in vitro and in vivo was measured. S. Typhimurium cultured in minimal media displayed a reduced ability to invade human epithelial cells in a manner that was at least partially independent of the Salmonella Pathogenicity Island 1 (SPI-1) type III secretion system. Nutrient deprivation did not, however, alter the ability of S. Typhimurium to replicate and survive inside epithelial cells. In a murine model of S. Typhimurium-induced gastroenteritis, prior cultivation in minimal media did not alter the pathogen's ability to colonize mice, nor did it affect levels of gastrointestinal inflammation. Upon examining the post-infection fecal gastrointestinal microbiota, we found that specifically in the 129Sv/ImJ murine strain S. Typhimurium cultured in minimal media induced differential microbiota compositional shifts compared to that of S. Typhimurium cultured in rich media. Together these findings demonstrate that S. Typhimurium remains a potent pathogen even in the face of nutritional deprivation, but nevertheless that nutrient deprivation encountered in this environment elicits significant changes in the bacterium genetic programme, as well as its capacity to alter host microbiota composition.


Subject(s)
Gastroenteritis/diet therapy , Gastrointestinal Microbiome/genetics , Genomic Islands/genetics , Salmonella Infections/diet therapy , Salmonella typhimurium/genetics , Animals , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Feces/microbiology , Gastroenteritis/genetics , Gastroenteritis/microbiology , Gene Expression Regulation, Bacterial , Humans , Intestines/microbiology , Intestines/pathology , Mice , Salmonella Infections/microbiology , Salmonella typhimurium/growth & development , Salmonella typhimurium/pathogenicity , Starvation/metabolism , Starvation/pathology
10.
mBio ; 6(5): e01421-15, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26396246

ABSTRACT

UNLABELLED: Nontyphoidal Salmonella enterica (NTS) infections are a major burden to global public health, as they lead to diseases ranging from gastroenteritis to systemic infections and there is currently no vaccine available. Here, we describe a highly effective component vaccine against S. enterica serovar Typhimurium in both gastroenteritis and systemic murine infection models. We devised an approach to generate supernatants of S. enterica serovar Typhimurium, an organism that is highly abundant in virulence factors. Immunization of mice with this supernatant resulted in dramatic protection against a challenge with serovar Typhimurium, showing increased survival in the systemic model and decreased intestinal pathology in the gastrointestinal model. Protection correlated with specific IgA and IgG levels in the serum and specific secretory IgA levels in the feces of immunized mice. Initial characterization of the protective antigens in the bacterial culture supernatants revealed a subset of antigens that exhibited remarkable stability, a highly desirable characteristic of an effective vaccine to be used under suboptimal environmental conditions in developing countries. We were able to purify a subset of the peptides present in the supernatants and show their potential for immunization of mice against serovar Typhimurium resulting in a decreased level of colonization. This component vaccine shows promise with regard to protecting against NTS, and further work should significantly help to establish vaccines against these prevalent infections. IMPORTANCE: Salmonella enterica infections other than typhoid and paratyphoid fever are a major global health burden, as they cause high morbidity and mortality worldwide. Strategies that prevent Salmonella-related diseases are greatly needed, and there is a significant push for the development of vaccines against nontyphoidal Salmonella enterica serovars. In this work, we describe an S. Typhimurium supernatant-derived vaccine that is effective in reducing bacterial colonization in mouse models of gastroenteritis as well as invasive disease. This is a component vaccine that shows high stability to heat, a feature that is important for use under suboptimal conditions, such as those found in sub-Saharan Africa.


Subject(s)
Culture Media/chemistry , Salmonella Infections/prevention & control , Salmonella Vaccines/administration & dosage , Salmonella Vaccines/immunology , Salmonella typhimurium/immunology , Animals , Antibodies, Bacterial/blood , Bacteremia/microbiology , Bacteremia/prevention & control , Disease Models, Animal , Feces/chemistry , Gastroenteritis/microbiology , Gastroenteritis/prevention & control , Immunoglobulin A/blood , Immunoglobulin A, Secretory/analysis , Immunoglobulin G/blood , Intestines/pathology , Mice , Salmonella Infections/microbiology , Salmonella Vaccines/isolation & purification , Salmonella typhimurium/growth & development , Survival Analysis , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Vaccines, Subunit/isolation & purification
11.
mBio ; 5(4): e01183-14, 2014 Jul 29.
Article in English | MEDLINE | ID: mdl-25073640

ABSTRACT

The mammalian gut contains a complex assembly of commensal microbes termed microbiota. Although much has been learned about the role of these microbes in health, the mechanisms underlying these functions are ill defined. We have recently shown that the mammalian gut contains thousands of small molecules, most of which are currently unidentified. Therefore, we hypothesized that these molecules function as chemical cues used by hosts and microbes during their interactions in health and disease. Thus, a search was initiated to identify molecules produced by the microbiota that are sensed by pathogens. We found that a secreted molecule produced by clostridia acts as a strong repressor of Salmonella virulence, obliterating expression of the Salmonella pathogenicity island 1 as well as host cell invasion. It has been known for decades that the microbiota protects its hosts from invading pathogens, and these data suggest that chemical sensing may be involved in this phenomenon. Further investigations should reveal the exact biological role of this molecule as well as its therapeutic potential. Importance: Microbes can communicate through the production and sensing of small molecules. Within the complex ecosystem formed by commensal microbes living in and on the human body, it is likely that these molecular messages are used extensively during the interactions between different microbial species as well as with host cells. Deciphering such a molecular dialect will be fundamental to our understanding of host-microbe interactions in health and disease and may prove useful for the design of new therapeutic strategies that target these mechanisms of communication.


Subject(s)
Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Metabolome/physiology , Animals , Feces/chemistry , Gastrointestinal Tract/metabolism , Host-Pathogen Interactions , Humans , Metabolome/genetics , Mice , Mice, Mutant Strains , Salmonella/pathogenicity
12.
BMC Microbiol ; 13: 238, 2013 Oct 29.
Article in English | MEDLINE | ID: mdl-24165751

ABSTRACT

BACKGROUND: Enterohepatic bacterial infections have the potential to affect multiple physiological processes of the body. Fibroblast growth factor 15/19 (FGF15 in mice, FGF19 in humans) is a hormone that functions as a central regulator of glucose, lipid and bile acid metabolism. FGF15/19 is produced in the intestine and exert its actions on the liver by signaling through the FGFR4-ßKlotho receptor complex. Here, we examined the in vivo effects of enterohepatic bacterial infection over the FGF15 endocrine axis. RESULTS: Infection triggered significant reductions in the intestinal expression of Fgf15 and its hepatic receptor components (Fgfr4 and Klb (ßKlotho)). Infection also resulted in alterations of the expression pattern of genes involved in hepatobiliary function, marked reduction in gallbladder bile volumes and accumulation of hepatic cholesterol and triglycerides. The decrease in ileal Fgf15 expression was associated with liver bacterial colonization and hepatobiliary pathophysiology rather than with direct intestinal bacterial pathogenesis. CONCLUSIONS: Bacterial pathogens of the enterohepatic system can disturb the homeostasis of the FGF15/19-FGFR4 endocrine axis. These results open up a possible link between FGF15/19-FGFR4 disruptions and the metabolic and nutritional disorders observed in infectious diseases.


Subject(s)
Fibroblast Growth Factors/metabolism , Gastrointestinal Tract/pathology , Listeriosis/pathology , Liver/pathology , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Salmonella Infections, Animal/pathology , Animals , Disease Models, Animal , Female , Gastrointestinal Tract/microbiology , Gene Expression Profiling , Liver/microbiology , Mice , Mice, Inbred C57BL
13.
PLoS One ; 8(7): e69759, 2013.
Article in English | MEDLINE | ID: mdl-23922794

ABSTRACT

15-deoxy-Δ(12,14)-prostaglandin J2 (15d-PGJ2) is an anti-inflammatory downstream product of the cyclooxygenase enzymes. It has been implicated to play a protective role in a variety of inflammatory mediated diseases, including rheumatoid arthritis, neural damage, and myocardial infarctions. Here we show that 15d-PGJ2 also plays a role in Salmonella infection. Salmonella enterica Typhimurium is a Gram-negative facultative intracellular pathogen that is able to survive and replicate inside phagocytic immune cells, allowing for bacterial dissemination to systemic sites. Salmonella species cause a wide range of morbidity and mortality due to gastroenteritis and typhoid fever. Previously we have shown that in mouse models of typhoid fever, Salmonella infection causes a major perturbation in the prostaglandin pathway. Specifically, we saw that 15d-PGJ2 production was significantly increased in both liver and feces. In this work we show that 15d-PGJ2 production is also significantly increased in macrophages infected with Salmonella. Furthermore, we show that the addition of 15d-PGJ2 to Salmonella infected RAW264.7, J774, and bone marrow derived macrophages is sufficient to significantly reduce bacterial colonization. We also show evidence that 15d-PGJ2 is reducing bacterial uptake by macrophages. 15d-PGJ2 reduces the inflammatory response of these infected macrophages, as evidenced by a reduction in the production of cytokines and reactive nitrogen species. The inflammatory response of the macrophage is important for full Salmonella virulence, as it can give the bacteria cues for virulence. The reduction in bacterial colonization is independent of the expression of Salmonella virulence genes SPI1 and SPI2, and is independent of the 15d-PGJ2 ligand PPAR-γ. 15d-PGJ2 also causes an increase in ERK1/2 phosphorylation in infected macrophages. In conclusion, we show here that 15d-PGJ2 mediates the outcome of bacterial infection, a previously unidentified role for this prostaglandin.


Subject(s)
Macrophages/drug effects , Macrophages/microbiology , Prostaglandin D2/analogs & derivatives , Salmonella typhimurium/growth & development , Animals , Arachidonic Acid/metabolism , Colony Count, Microbial , Cytokines/biosynthesis , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation, Bacterial/drug effects , Genes, Bacterial/genetics , HeLa Cells , Host-Pathogen Interactions/drug effects , Humans , Immunity/drug effects , Macrophages/enzymology , Macrophages/immunology , Mice , Mice, Inbred C57BL , PPAR gamma/metabolism , Phosphorylation/drug effects , Prostaglandin D2/pharmacology , Reactive Nitrogen Species/metabolism , Salmonella Infections/microbiology , Salmonella Infections/pathology , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Salmonella typhimurium/pathogenicity , Virulence/drug effects , Virulence/genetics
14.
PLoS One ; 7(11): e49646, 2012.
Article in English | MEDLINE | ID: mdl-23155475

ABSTRACT

The intestinal microbiota has been found to play a central role in the colonization of Salmonella enterica serovar Typhimurium in the gastrointestinal tract. In this study, we present a novel process through which Salmonella benefit from inflammatory induced changes in the microbiota in order to facilitate disease. We show that Salmonella infection in mice causes recruitment of neutrophils to the gut lumen, resulting in significant changes in the composition of the intestinal microbiota. This occurs through the production of the enzyme elastase by neutrophils. Administration of recombinant neutrophil elastase to infected animals under conditions that do not elicit neutrophil recruitment caused shifts in microbiota composition that favored Salmonella colonization, while inhibition of neutrophil elastase reduced colonization. This study reveals a new relationship between the microbiota and the host during infection.


Subject(s)
Intestines/microbiology , Leukocyte Elastase/pharmacology , Neutrophils/immunology , Salmonella Infections, Animal/immunology , Salmonella typhimurium/genetics , Animals , Intestines/drug effects , Intestines/immunology , Metagenome , Mice , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Salmonella Infections, Animal/genetics
15.
J Bacteriol ; 194(9): 2286-96, 2012 May.
Article in English | MEDLINE | ID: mdl-22366421

ABSTRACT

Infection with Salmonella enterica serovar Typhi in humans causes the life-threatening disease typhoid fever. In the laboratory, typhoid fever can be modeled through the inoculation of susceptible mice with Salmonella enterica serovar Typhimurium. Using this murine model, we previously characterized the interactions between Salmonella Typhimurium and host cells in the gallbladder and showed that this pathogen can successfully invade gallbladder epithelial cells and proliferate. Additionally, we showed that Salmonella Typhimurium can use bile phospholipids to grow at high rates. These abilities are likely important for quick colonization of the gallbladder during typhoid fever and further pathogen dissemination through fecal shedding. To further characterize the interactions between Salmonella and the gallbladder environment, we compared the transcriptomes of Salmonella cultures grown in LB broth or physiological murine bile. Our data showed that many genes involved in bacterial central metabolism are affected by bile, with the citric acid cycle being repressed and alternative respiratory systems being activated. Additionally, our study revealed a new aspect of Salmonella interactions with bile through the identification of the global regulator phoP as a bile-responsive gene. Repression of phoP expression could also be achieved using physiological, but not commercial, bovine bile. The biological activity does not involve PhoPQ sensing of a bile component and is not caused by bile acids, the most abundant organic components of bile. Bioactivity-guided purification allowed the identification of a subset of small molecules from bile that can elicit full activity; however, a single compound with phoP inhibitory activity could not be isolated, suggesting that multiple molecules may act in synergy to achieve this effect. Due to the critical role of phoP in Salmonella virulence, further studies in this area will likely reveal aspects of the interaction between Salmonella and bile that are relevant to disease.


Subject(s)
Bacterial Proteins/metabolism , Bile Acids and Salts/pharmacology , Bile , Gene Expression Regulation, Bacterial/drug effects , Salmonella enterica/drug effects , Salmonella enterica/metabolism , Animals , Bacterial Proteins/genetics , Bile/chemistry , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cattle , Gene Expression Profiling , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Array Analysis , Salmonella enterica/genetics
16.
Infect Immun ; 79(11): 4392-400, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21875964

ABSTRACT

To cause disease, Salmonella enterica serovar Typhimurium requires two type III secretion systems that are encoded by Salmonella pathogenicity islands 1 and 2 (SPI-1 and -2). These secretion systems serve to deliver specialized proteins (effectors) into the host cell cytosol. While the importance of these effectors to promote colonization and replication within the host has been established, the specific roles of individual secreted effectors in the disease process are not well understood. In this study, we used an in vivo gallbladder epithelial cell infection model to study the function of the SPI-2-encoded type III effector, SseL. The deletion of the sseL gene resulted in bacterial filamentation and elongation and the unusual localization of Salmonella within infected epithelial cells. Infection with the ΔsseL strain also caused dramatic changes in host cell lipid metabolism and led to the massive accumulation of lipid droplets in infected cells. This phenotype was directly attributable to the deubiquitinase activity of SseL, as a Salmonella strain carrying a single point mutation in the catalytic cysteine also resulted in extensive lipid droplet accumulation. The excessive buildup of lipids due to the absence of a functional sseL gene also was observed in murine livers during S. Typhimurium infection. These results suggest that SseL alters host lipid metabolism in infected epithelial cells by modifying the ubiquitination patterns of cellular targets.


Subject(s)
Bacterial Proteins/metabolism , Endopeptidases/metabolism , Genomic Islands/physiology , Lipid Metabolism/physiology , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/metabolism , Animals , Bacterial Proteins/genetics , Endopeptidases/genetics , Gallbladder/metabolism , Gallbladder/microbiology , Gene Expression Regulation, Bacterial , Genomic Islands/genetics , Liver/metabolism , Liver/microbiology , Mice , Salmonella typhimurium/enzymology , Salmonella typhimurium/genetics
17.
J Bacteriol ; 193(18): 4719-25, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21764918

ABSTRACT

During the colonization of hosts, bacterial pathogens are presented with many challenges that must be overcome for colonization to occur successfully. This requires the bacterial sensing of the surroundings and adaptation to the conditions encountered. One of the major impediments to the pathogen colonization of the mammalian gastrointestinal tract is the antibacterial action of bile. Salmonella enterica serovar Typhimurium has specific mechanisms involved in resistance to bile. Additionally, Salmonella can successfully multiply in bile, using it as a source of nutrients. This accomplishment is highly relevant to pathogenesis, as Salmonella colonizes the gallbladder of hosts, where it can be carried asymptomatically and promote further host spread and transmission. To gain insights into the mechanisms used by Salmonella to grow in bile, we studied the changes elicited by Salmonella in the chemical composition of bile during growth in vitro and in vivo through a metabolomics approach. Our data suggest that phospholipids are an important source of carbon and energy for Salmonella during growth in the laboratory as well as during gallbladder infections of mice. Further studies in this area will generate a better understanding of how Salmonella exploits this generally hostile environment for its own benefit.


Subject(s)
Bile/metabolism , Bile/microbiology , Metabolomics , Phospholipids/metabolism , Salmonella typhimurium/growth & development , Salmonella typhimurium/metabolism , Animals , Carbon/metabolism , Energy Metabolism , Mice , Mice, Inbred C57BL
18.
PLoS One ; 6(5): e20338, 2011.
Article in English | MEDLINE | ID: mdl-21633507

ABSTRACT

The intestinal microbiota is composed of hundreds of species of bacteria, fungi and protozoa and is critical for numerous biological processes, such as nutrient acquisition, vitamin production, and colonization resistance against bacterial pathogens. We studied the role of the intestinal microbiota on host resistance to Salmonella enterica serovar Typhimurium-induced colitis. Using multiple antibiotic treatments in 129S1/SvImJ mice, we showed that disruption of the intestinal microbiota alters host susceptibility to infection. Although all antibiotic treatments caused similar increases in pathogen colonization, the development of enterocolitis was seen only when streptomycin or vancomycin was used; no significant pathology was observed with the use of metronidazole. Interestingly, metronidazole-treated and infected C57BL/6 mice developed severe pathology. We hypothesized that the intestinal microbiota confers resistance to infectious colitis without affecting the ability of S. Typhimurium to colonize the intestine. Indeed, different antibiotic treatments caused distinct shifts in the intestinal microbiota prior to infection. Through fluorescence in situ hybridization, terminal restriction fragment length polymorphism, and real-time PCR, we showed that there is a strong correlation between the intestinal microbiota composition before infection and susceptibility to Salmonella-induced colitis. Members of the Bacteroidetes phylum were present at significantly higher levels in mice resistant to colitis. Further analysis revealed that Porphyromonadaceae levels were also increased in these mice. Conversely, there was a positive correlation between the abundance of Lactobacillus sp. and predisposition to colitis. Our data suggests that different members of the microbiota might be associated with S. Typhimurium colonization and colitis. Dissecting the mechanisms involved in resistance to infection and inflammation will be critical for the development of therapeutic and preventative measures against enteric pathogens.


Subject(s)
Enterocolitis/microbiology , Intestines/microbiology , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/physiology , Animals , Anti-Infective Agents/pharmacology , Bacteria/classification , Bacteria/drug effects , Bacteria/genetics , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Ecosystem , Feces/microbiology , Host-Pathogen Interactions , In Situ Hybridization, Fluorescence , Intestines/drug effects , Intestines/pathology , Metagenome/drug effects , Metagenome/genetics , Metronidazole/pharmacology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Salmonella typhimurium/drug effects , Salmonella typhimurium/genetics , Species Specificity , Streptomycin/pharmacology , Vancomycin/pharmacology
20.
F1000 Biol Rep ; 3: 4, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21399765

ABSTRACT

Chemical signaling via the production of small molecules such as hormones has been studied in detail in higher organisms. These molecules have important functions in maintaining physiological homeostasis as well as allowing organisms to respond to external insults. Virtually every living cell produces hormone-like diffusible small molecules that can be used to convey messages to neighboring cells-a vital step in adaptation, development, and survival within populations. Although most of our knowledge on cellular chemical communication comes from studies of multicellular eukaryotes, it is now understood that bacteria can also communicate using sophisticated signaling systems, in a way analogous to those used by higher organisms. Many of these microbes live in close association with higher eukaryotes, in mutualistic or commensal relationships. We suggest that there may be a wealth of unidentified bioactive small molecules in the human body, originating from both microbial and human cells and that have important biological functions. Because chemical signaling has important roles for the biology of both microbes and humans, detecting, identifying, and studying these chemical signals can further our understanding of the chemical interplay between microbiota and their hosts and provide us with an unexplored source of molecules that could be used for human benefit.

SELECTION OF CITATIONS
SEARCH DETAIL
...