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










Publication year range
1.
Virulence ; 10(1): 643-656, 2019 12.
Article in English | MEDLINE | ID: mdl-31314675

ABSTRACT

Pathogens enhance their survival during infections by manipulating host defenses. Francisella tularensis evades innate immune responses, which we have found to be dependent on an understudied gene ybeX (FTL_0883/FTT_0615c). To understand the function of YbeX, we sought protein interactors in F. tularensis subsp. holarctica live vaccine strain (LVS). An unstudied Francisella protein co-immunoprecipitated with recombinant YbeX, which is a predicted glycosyltransferase with a DXD-motif. There are up to four genomic copies of this gene with identical sequence in strains of F. tularensis pathogenic to humans, despite ongoing genome decay. Disruption mutations were generated by intron insertion into all three copies of this glycosyltransferase domain containing gene in LVS, gdcA1-3. The resulting strains stimulated more cytokines from macrophages in vitro than wild-type LVS and were attenuated in two in vivo infection models. GdcA was released from LVS during culture and was sufficient to block NF-κB activation when expressed in eukaryotic cells. When co-expressed in zebrafish, GdcA and YbeX were synergistically lethal to embryo development. Glycosyltransferases with DXD-motifs are found in a variety of pathogens including NleB, an Escherichia coli type-III secretion system effector that inhibits NF-κB by antagonizing death receptor signaling. To our knowledge, GdcA is the first DXD-motif glycosyltransferase that inhibits NF-κB in immune cells. Together, these findings suggest DXD-motif glycosyltransferases may be a conserved virulence mechanism used by pathogenic bacteria to remodel host defenses.


Subject(s)
Bacterial Proteins/immunology , Francisella tularensis/enzymology , Glycosyltransferases/immunology , Host-Pathogen Interactions , Animals , Bacterial Proteins/genetics , Cytokines , Female , Francisella tularensis/genetics , Glycosyltransferases/genetics , Humans , Immunity, Innate , Jurkat Cells , Macrophages/microbiology , Mice, Inbred C57BL , Moths , Mutation , Tularemia/immunology , Tularemia/microbiology , Virulence , Zebrafish
2.
Nucleic Acids Res ; 43(3): e20, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25428368

ABSTRACT

Identifying conserved and divergent response patterns in gene networks is becoming increasingly important. A common approach is integrating expression information with gene association networks in order to find groups of connected genes that are activated or repressed. In many cases, researchers are also interested in comparisons across species (or conditions). Finding an active sub-network is a hard problem and applying it across species requires further considerations (e.g. orthology information, expression data and networks from different sources). To address these challenges we devised ModuleBlast, which uses both expression and network topology to search for highly relevant sub-networks. We have applied ModuleBlast to expression and interaction data from mouse, macaque and human to study immune response and aging. The immune response analysis identified several relevant modules, consistent with recent findings on apoptosis and NFκB activation following infection. Temporal analysis of these data revealed cascades of modules that are dynamically activated within and across species. We have experimentally validated some of the novel hypotheses resulting from the analysis of the ModuleBlast results leading to new insights into the mechanisms used by a key mammalian aging protein.


Subject(s)
Gene Regulatory Networks , Aging/genetics , Animals , Apoptosis , Humans , Macaca , Mice , Species Specificity
3.
Vaccine ; 32(52): 7098-107, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25454872

ABSTRACT

Here, we report on a first-in-man trial where the tuberculosis (TB) vaccine Ag85B-ESAT-6 (H1) was adjuvanted with escalating doses of a novel liposome adjuvant CAF01. On their own, protein antigens cannot sufficiently induce immune responses in humans, and require the addition of an adjuvant system to ensure appropriate delivery and concomitant immune activation. To date no approved adjuvants are available for induction of cellular immunity, which seems essential for a number of vaccines, including vaccines against TB. We vaccinated four groups of human volunteers: a non-adjuvanted H1 group, followed by three groups with escalating doses of CAF01-adjuvanted H1 vaccine. All subjects were vaccinated at 0 and 8 weeks and followed up for 150 weeks. Vaccination did not cause local or systemic adverse effects besides transient soreness at the injection site. Two vaccinations elicited strong antigen-specific T-cell responses which persisted after 150 weeks follow-up, indicating the induction of a long-lasting memory response in the vaccine recipients. These results show that CAF01 is a safe and tolerable, Th1-inducing adjuvant for human TB vaccination trials and for vaccination studies in general where cellular immunity is required.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Immunologic Memory , Liposomes/administration & dosage , Mycobacterium tuberculosis/immunology , T-Lymphocytes/immunology , Tuberculosis Vaccines/immunology , Adjuvants, Immunologic/adverse effects , Adolescent , Adult , Female , Healthy Volunteers , Humans , Male , Middle Aged , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/toxicity , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/adverse effects , Young Adult
4.
Microbes Infect ; 16(4): 300-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24374051

ABSTRACT

Biosynthesis and acquisition of nutrients during infection are integral to pathogenesis. Members of a metabolic pathway, the glycine cleavage system, have been identified in virulence screens of the intracellular bacterium Francisella tularensis but their role in pathogenesis remains unknown. This system generates 5,10-methylenetetrahydrofolate, a precursor of amino acid and DNA synthesis, from glycine degradation. To characterize this pathway, deletion of the gcvT homolog, an essential member of this system, was performed in attenuated and virulent F. tularensis strains. Deletion mutants were auxotrophic for serine but behaved similar to wild-type strains with respect to host cell invasion, intracellular replication, and stimulation of TNF-α. Unexpectedly, the glycine cleavage system was required for the pathogenesis of virulent F. tularensis in a murine model. Deletion of the gcvT homolog delayed mortality and lowered bacterial burden, particularly in the liver and bloodstream. To reconcile differences between the cell culture model and animal model, minimal tissue culture media was employed to mimic the nutritionally limiting environment of the host. This reevaluation demonstrated that the glycine cleavage system contributes to the intracellular replication of virulent F. tularensis in serine limiting environments. Thus, the glycine cleavage system is the serine biosynthetic pathway of F. tularensis and contributes to pathogenesis in vivo.


Subject(s)
Amino Acid Oxidoreductases , Carrier Proteins , Francisella tularensis/metabolism , Francisella tularensis/pathogenicity , Multienzyme Complexes , Transferases , Tularemia/microbiology , Tularemia/pathology , Animals , Bacterial Proteins/genetics , Disease Models, Animal , Francisella tularensis/genetics , Gene Deletion , Mice, Inbred C57BL , Tetrahydrofolates/metabolism , Virulence
5.
Article in English | MEDLINE | ID: mdl-24367766

ABSTRACT

The highly infectious and deadly pathogen, Francisella tularensis, is classified by the CDC as a Category A bioterrorism agent. Inhalation of a single bacterium results in an acute pneumonia with a 30-60% mortality rate without treatment. Due to the prevalence of antibiotic resistance, there is a strong need for new types of antibacterial drugs. Resazurin is commonly used to measure bacterial and eukaryotic cell viability through its reduction to the fluorescent product resorufin. When tested on various bacterial taxa at the recommended concentration of 44 µM, a potent bactericidal effect was observed against various Francisella and Neisseria species, including the human pathogens type A F. tularensis (Schu S4) and N. gonorrhoeae. As low as 4.4 µM resazurin was sufficient for a 10-fold reduction in F. tularensis growth. In broth culture, resazurin was reduced to resorufin by F. tularensis. Resorufin also suppressed the growth of F. tularensis suggesting that this compound is the biologically active form responsible for decreasing the viability of F. tularensis LVS bacteria. Replication of F. tularensis in primary human macrophages and non-phagocytic cells was abolished following treatment with 44 µM resazurin indicating this compound could be an effective therapy for tularemia in vivo.


Subject(s)
Anti-Infective Agents/pharmacology , Francisella tularensis/drug effects , Oxazines/pharmacology , Xanthenes/pharmacology , Anti-Infective Agents/metabolism , Biotransformation , Cell Line , Colony Count, Microbial , Epithelial Cells/microbiology , Humans , Macrophages/microbiology , Microbial Viability/drug effects , Oxazines/metabolism , Xanthenes/metabolism
6.
Microbes Infect ; 15(3): 201-11, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23211929

ABSTRACT

Pneumonic tularemia is a potentially fatal disease caused by the Category A bioterrorism agent Francisella tularensis. Understanding the pulmonary immune response to this bacterium is necessary for developing effective vaccines and therapeutics. In this study, characterization of immune cell populations in the lungs of mice infected with the type A strain Schu S4 revealed a significant loss in natural killer (NK) cells over time. Since this decline in NK cells correlated with morbidity and mortality, we hypothesized these cells contribute to host defense against Schu S4 infection. Depletion of NK cells prior to Schu S4 challenge significantly reduced IFN-γ and granzyme B in the lung but had no effect on bacterial burden or disease progression. Conversely, increasing NK cell numbers with the anti-apoptotic cytokine IL-15 and soluble receptor IL-15Rα had no significant impact on Schu S4 growth in vivo. A modest decrease in median time to death, however, was observed in live vaccine strain (LVS)-vaccinated mice depleted of NK1.1+ cells and challenged with Schu S4. Therefore, NK cells do not appear to contribute to host defense against acute respiratory infection with type A F. tularensis in vivo, but they play a minor role in protection elicited by LVS vaccination.


Subject(s)
Francisella tularensis/immunology , Killer Cells, Natural/immunology , Tularemia/immunology , Analysis of Variance , Animals , Cell Survival/immunology , Female , Granzymes/immunology , Granzymes/metabolism , Host-Pathogen Interactions , Interferon-gamma/metabolism , Interleukin-15/immunology , Interleukin-15/metabolism , Killer Cells, Natural/microbiology , Leukocytes/immunology , Lung/immunology , Lung/microbiology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Tularemia/microbiology
7.
PLoS One ; 7(2): e31172, 2012.
Article in English | MEDLINE | ID: mdl-22355343

ABSTRACT

Vaccination is a proven strategy to mitigate morbidity and mortality of infectious diseases. The methodology of identifying and testing new vaccine candidates could be improved with rational design and in vitro testing prior to animal experimentation. The tularemia vaccine, Francisella tularensis live vaccine strain (LVS), does not elicit complete protection against lethal challenge with a virulent type A Francisella strain. One factor that may contribute to this poor performance is limited stimulation of antigen-presenting cells. In this study, we examined whether the interaction of genetically modified LVS strains with human antigen-presenting cells correlated with effectiveness as tularemia vaccine candidates. Human dendritic cells infected with wild-type LVS secrete low levels of proinflammatory cytokines, fail to upregulate costimulatory molecules, and activate human T cells poorly in vitro. One LVS mutant, strain 13B47, stimulated higher levels of proinflammatory cytokines from dendritic cells and macrophages and increased costimulatory molecule expression on dendritic cells compared to wild type. Additionally, 13B47-infected dendritic cells activated T cells more efficiently than LVS-infected cells. A deletion allele of the same gene in LVS displayed similar in vitro characteristics, but vaccination with this strain did not improve survival after challenge with a virulent Francisella strain. In vivo, this mutant was attenuated for growth and did not stimulate T cell responses in the lung comparable to wild type. Therefore, stimulation of antigen-presenting cells in vitro was improved by genetic modification of LVS, but did not correlate with efficacy against challenge in vivo within this model system.


Subject(s)
Antigen-Presenting Cells/immunology , Bacterial Vaccines/administration & dosage , Francisella tularensis/immunology , Immunization , Tularemia/therapy , Vaccines, Attenuated/administration & dosage , Animals , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/virology , Female , Flow Cytometry , Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Humans , Lung/immunology , Lung/metabolism , Lung/virology , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Mice , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/metabolism , Monocytes/virology , Tularemia/immunology , Tularemia/mortality , Vaccination , Virulence
8.
PLoS One ; 6(7): e22401, 2011.
Article in English | MEDLINE | ID: mdl-21789257

ABSTRACT

Viral and bacterial infections of the lower respiratory tract are major causes of morbidity and mortality worldwide. Alveolar macrophages line the alveolar spaces and are the first cells of the immune system to respond to invading pathogens. To determine the similarities and differences between the responses of mice and macaques to invading pathogens we profiled alveolar macrophages from these species following infection with two viral (PR8 and Fuj/02 influenza A) and two bacterial (Mycobacterium tuberculosis and Francisella tularensis Schu S4) pathogens. Cells were collected at 6 time points following each infection and expression profiles were compared across and between species. Our analyses identified a core set of genes, activated in both species and across all pathogens that were predominantly part of the interferon response pathway. In addition, we identified similarities across species in the way innate immune cells respond to lethal versus non-lethal pathogens. On the other hand we also found several species and pathogen specific response patterns. These results provide new insights into mechanisms by which the innate immune system responds to, and interacts with, invading pathogens.


Subject(s)
Bacteria/immunology , Host-Pathogen Interactions/immunology , Immunity, Innate/genetics , Immunity, Innate/immunology , Macaca/microbiology , Macaca/virology , Viruses/immunology , Animals , Francisella tularensis/immunology , Gene Expression Profiling , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Influenza A virus/immunology , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/metabolism , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/virology , Mice , Mycobacterium tuberculosis/immunology , Oligonucleotide Array Sequence Analysis , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/virology , Signal Transduction/genetics , Species Specificity , Tuberculosis/genetics , Tuberculosis/microbiology , Tularemia/genetics , Tularemia/microbiology , Up-Regulation
9.
J Infect Dis ; 204(1): 51-9, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21628658

ABSTRACT

Francisella tularensis is the causative agent of tularemia and is classified as a category A biodefense agent by the Centers for Disease Control and Prevention because of its highly infectious nature. F. tularensis infects leukocytes and exhibits an extracellular phase in the blood of the host. It is unknown, however, whether F. tularensis can infect erythrocytes; thus, we examined this possibility in vivo and in vitro. In the murine model of pulmonary type A tularemia, we showed the presence of intraerythrocytic bacteria by double-immunofluorescence microscopy and ex vivo gentamicin protection of the purified erythrocyte fraction. In vitro, F. tularensis invaded human erythrocytes, as shown in the gentamicin protection assays, double-immunofluorescence microscopy, flow cytometry, scanning electron microscopy, and transmission electron microscopy with immunogold labeling of the bacteria. Additional in vitro tests indicated that serum complement-dependent and complement-independent mechanisms contribute to erythrocyte invasion. Our results reveal a novel intraerythrocytic phase during F. tularensis infection.


Subject(s)
Erythrocytes/microbiology , Francisella tularensis/pathogenicity , Tularemia/microbiology , Tularemia/pathology , Animals , Complement System Proteins/immunology , Disease Models, Animal , Female , Flow Cytometry , Francisella tularensis/growth & development , Humans , Mice , Mice, Inbred C57BL , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Tularemia/epidemiology
10.
Infect Immun ; 79(9): 3665-76, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21670171

ABSTRACT

Tularemia is a debilitating febrile illness caused by the category A biodefense agent Francisella tularensis. This pathogen infects over 250 different hosts, has a low infectious dose, and causes high morbidity and mortality. Our understanding of the mechanisms by which F. tularensis senses and adapts to host environments is incomplete. Polyamines, including spermine, regulate the interactions of F. tularensis with host cells. However, it is not known whether responsiveness to polyamines is necessary for the virulence of the organism. Through transposon mutagenesis of F. tularensis subsp. holarctica live vaccine strain (LVS), we identified FTL_0883 as a gene important for spermine responsiveness. In-frame deletion mutants of FTL_0883 and FTT_0615c, the homologue of FTL_0883 in F. tularensis subsp. tularensis Schu S4 (Schu S4), elicited higher levels of cytokines from human and murine macrophages compared to wild-type strains. Although deletion of FTL_0883 attenuated LVS replication within macrophages in vitro, the Schu S4 mutant with a deletion in FTT_0615c replicated similarly to wild-type Schu S4. Nevertheless, both the LVS and the Schu S4 mutants were significantly attenuated in vivo. Growth and dissemination of the Schu S4 mutant was severely reduced in the murine model of pneumonic tularemia. This attenuation depended on host responses to elevated levels of proinflammatory cytokines. These data associate responsiveness to polyamines with tularemia pathogenesis and define FTL_0883/FTT_0615c as an F. tularensis gene important for virulence and evasion of the host immune response.


Subject(s)
Bacterial Proteins/genetics , Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Spermine/metabolism , Tularemia/microbiology , Animals , Bacterial Proteins/physiology , Cells, Cultured , Chemokines/biosynthesis , Chemokines/immunology , Cytokines/biosynthesis , Cytokines/immunology , Enzyme-Linked Immunosorbent Assay , Female , Francisella tularensis/growth & development , Francisella tularensis/immunology , Host-Pathogen Interactions , Humans , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mutagenesis , Polymerase Chain Reaction , Sequence Deletion , Tularemia/immunology
11.
Infect Immun ; 78(6): 2607-19, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20385757

ABSTRACT

The pathogenesis of Francisella tularensis has been associated with this bacterium's ability to replicate within macrophages. F. tularensis can also invade and replicate in a variety of nonphagocytic host cells, including lung and kidney epithelial cells and hepatocytes. As uracil biosynthesis is a central metabolic pathway usually necessary for pathogens, we characterized DeltapyrF mutants of both F. tularensis LVS and Schu S4 to investigate the role of these mutants in intracellular growth. As expected, these mutant strains were deficient in de novo pyrimidine biosynthesis and were resistant to 5-fluoroorotic acid, which is converted to a toxic product by functional PyrF. The F. tularensis DeltapyrF mutants could not replicate in primary human macrophages. The inability to replicate in macrophages suggested that the F. tularensis DeltapyrF strains would be attenuated in animal infection models. Surprisingly, these mutants retained virulence during infection of chicken embryos and in the murine model of pneumonic tularemia. We hypothesized that the F. tularensis DeltapyrF strains may replicate in cells other than macrophages to account for their virulence. In support of this, F. tularensis DeltapyrF mutants replicated in HEK-293 cells and normal human fibroblasts in vitro. Moreover, immunofluorescence microscopy showed abundant staining of wild-type and mutant bacteria in nonmacrophage cells in the lungs of infected mice. These findings indicate that replication in nonmacrophages contributes to the pathogenesis of F. tularensis.


Subject(s)
Bacterial Proteins/physiology , Francisella tularensis/pathogenicity , Gene Deletion , Virulence Factors/physiology , Animals , Bacterial Proteins/genetics , Cell Line , Cells, Cultured , Chick Embryo , Chickens , Disease Models, Animal , Epithelial Cells/microbiology , Female , Fibroblasts/microbiology , Humans , Lung/microbiology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Tularemia/microbiology , Tularemia/pathology , Virulence Factors/genetics
12.
J Innate Immun ; 2(1): 56-65, 2010.
Article in English | MEDLINE | ID: mdl-20375623

ABSTRACT

Interferon (IFN)-gamma is important to the immune defense against intracellular pathogens and specifically the ability of macrophages to control Mycobacterium tuberculosis (MTB). Increasing evidence has accumulated to support the idea that macrophages produce IFN-gamma. We describe here the cytokine interactions that determine IFN-gamma expression and secretion during MTB infection of human macrophages. Detection of biologically important IFN-gamma levels in culture supernatants of MTB-infected human macrophages requires the addition of interleukin (IL)-12. IL-18 augmented IFN-gamma production from human macrophages in response to the combination of MTB and supplemental IL-12. Although IL-18 gene expression was generally unchanged, IL-18 protein secretion was enhanced by the combination of MTB and IL-12, and functioned primarily to stimulate IFN-gamma release. Importantly, IL-27 induced by MTB infection opposed IFN-gamma production by antagonizing IL-18 activity in human macrophages. Neutralization of IL-27 increased the expression of the IL-18 receptor beta-chain. Additionally, IL-27 blocked NF-kappaB activation in response to IL-18. These results define the signals required for IFN-gamma production by human macrophages and highlight the interactions between cytokines produced during MTB infection. Together, they identify a novel role for IL-27 in regulating macrophage function by disrupting IL-18 activity.


Subject(s)
Interferon-gamma/immunology , Interleukins/immunology , Macrophages/immunology , Gene Expression Regulation , Humans , Interferon-gamma/biosynthesis , Interleukin-12/immunology , Interleukin-18/immunology , Macrophages/metabolism , Mycobacterium tuberculosis/immunology , NF-kappa B/immunology , Receptors, Interleukin-18/immunology , Receptors, Interleukin-18/metabolism
13.
J Microbiol Methods ; 80(1): 106-8, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19879904

ABSTRACT

We engineered an efficient system to make Francisella tularensis deletion mutations using an unstable, poorly maintained plasmid to enhance the likelihood of homologous recombination. For counterselection, we adapted a strategy using I-SceI, which causes a double-stranded break in the integrated suicide vector, forcing a second recombination to mediate allelic replacement.


Subject(s)
Deoxyribonuclease I/metabolism , Francisella tularensis/genetics , Genetic Engineering/methods , Plasmids/genetics , Sequence Deletion , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line , Deoxyribonuclease I/genetics , Recombination, Genetic , Tularemia/microbiology
14.
J Bacteriol ; 191(22): 6855-64, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19749055

ABSTRACT

Tularemia is caused by the category A biodefense agent Francisella tularensis. This bacterium is associated with diverse environments and a plethora of arthropod and mammalian hosts. How F. tularensis adapts to these different conditions, particularly the eukaryotic intracellular environment in which it replicates, is poorly understood. Here, we demonstrate that the polyamines spermine and spermidine are environmental signals that alter bacterial stimulation of host cells. Genomewide analysis showed that F. tularensis LVS undergoes considerable changes in gene expression in response to spermine. Unexpectedly, analysis of gene expression showed that multiple members of two classes of Francisella insertion sequence (IS) elements, ISFtu1 and ISFtu2, and the genes adjacent to these elements were induced by spermine. Spermine was sufficient to activate transcription of these IS elements and of nearby genes in broth culture and in macrophages. Importantly, the virulent strain of F. tularensis, Schu S4, exhibited similar phenotypes of cytokine induction and gene regulation in response to spermine. Distinctions in gene expression changes between Schu S4 and LVS at one orthologous locus, however, correlated with differences in IS element location. Our results indicate that spermine and spermidine are novel triggers to alert F. tularensis of its eukaryotic host environment. The results reported here also identify an unexpected mechanism of gene regulation controlled by a spermine-responsive promoter contained within IS elements. Different arrangements of these mobile genetic elements among Francisella strains may contribute to virulence by conveying new expression patterns for genes from different strains.


Subject(s)
DNA Transposable Elements/physiology , Francisella tularensis/drug effects , Francisella tularensis/genetics , Gene Expression Regulation, Bacterial , Spermine/pharmacology , DNA Transposable Elements/genetics , Enzyme-Linked Immunosorbent Assay , Francisella tularensis/pathogenicity , Gene Expression Regulation, Bacterial/drug effects , Gene Expression Regulation, Bacterial/genetics , Macrophages/microbiology , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Promoter Regions, Genetic/genetics , Tularemia/genetics , Tularemia/microbiology , Virulence
15.
Genes Chromosomes Cancer ; 48(7): 569-82, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19396866

ABSTRACT

Altered microRNA (miRNA) expression profiles have been observed in numerous malignancies, including oral squamous cell carcinoma (OSCC). However, their role in disease is not entirely clear. Several genetic aberrations are characteristic of OSCC, with amplification of chromosomal band 11q13 and loss of distal 11q being among the most prevalent. It is not known if the expression levels of miRNAs in these regions are altered or whether they play a role in disease. We hypothesize that the expression of miRNAs mapping to 11q are altered in OSCC because of loss or amplification of chromosomal material, and that this contributes to the development and progression of OSCC. We found that miR-125b and miR-100 are down-regulated in OSCC tumor and cell lines, and that transfecting cells with exogenous miR-125b and miR-100 significantly reduced cell proliferation and modified the expression of target and nontarget genes, including some that are overexpressed in radioresistant OSCC cells. In conclusion, the down-regulation of miR-125b and miR-100 in OSCC appears to play an important role in the development and/or progression of disease and may contribute to the loss of sensitivity to ionizing radiation.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/biosynthesis , Mouth Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Blotting, Western , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Chromosomes, Human, Pair 11 , Down-Regulation , Female , Humans , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Mouth Neoplasms/genetics , Oligonucleotide Array Sequence Analysis , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Young Adult
16.
BMC Microbiol ; 8: 172, 2008 Oct 08.
Article in English | MEDLINE | ID: mdl-18842136

ABSTRACT

BACKGROUND: After infecting a mammalian host, the facultative intracellular bacterium, Francisella tularensis, encounters an elevated environmental temperature. We hypothesized that this temperature change may regulate genes essential for infection. RESULTS: Microarray analysis of F. tularensis LVS shifted from 26 degrees C (environmental) to 37 degrees C (mammalian) showed approximately 11% of this bacterium's genes were differentially-regulated. Importantly, 40% of the protein-coding genes that were induced at 37 degrees C have been previously implicated in virulence or intracellular growth of Francisella in other studies, associating the bacterial response to this temperature shift with pathogenesis. Forty-four percent of the genes induced at 37 degrees C encode proteins of unknown function, suggesting novel Francisella virulence traits are regulated by mammalian temperature. To explore this possibility, we generated two mutants of loci induced at 37 degrees C [FTL_1581 and FTL_1664 (deoB)]. The FTL_1581 mutant was attenuated in a chicken embryo infection model, which was likely attributable to a defect in survival within macrophages. FTL_1581 encodes a novel hypothetical protein that we suggest naming temperature-induced, virulence-associated locus A, tivA. Interestingly, the deoB mutant showed diminished entry into mammalian cells compared to wild-type LVS, including primary human macrophages and dendritic cells, the macrophage-like RAW 264.7 line, and non-phagocytic HEK-293 cells. This is the first study identifying a Francisella gene that contributes to uptake into both phagocytic and non-phagocytic host cells. CONCLUSION: Our results provide new insight into mechanisms of Francisella virulence regulation and pathogenesis. F. tularensis LVS undergoes considerable gene expression changes in response to mammalian body temperature. This temperature shift is important for the regulation of genes that are critical for the pathogenesis of Francisella. Importantly, the compilation of temperature-regulated genes also defines a rich collection of novel candidate virulence determinants, including tivA (FTL_1581). An analysis of tivA and deoB (FTL_1664) revealed that these genes contribute to intracellular survival and entry into mammalian cells, respectively.


Subject(s)
Body Temperature , Francisella tularensis/genetics , Transcription, Genetic , Tularemia/microbiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cells, Cultured , Chick Embryo , Dendritic Cells/microbiology , Francisella tularensis/metabolism , Francisella tularensis/pathogenicity , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Genes, Bacterial , Humans , Macrophages/microbiology , Mice , Mutation , Oligonucleotide Array Sequence Analysis , Plasmids , RNA, Bacterial/genetics , Reverse Transcriptase Polymerase Chain Reaction
17.
Res Microbiol ; 159(7-8): 562-8, 2008.
Article in English | MEDLINE | ID: mdl-18718529

ABSTRACT

Serratia marcescens is an emerging opportunistic pathogen with a remarkably broad host range. The cAMP-regulated catabolite repression system of S. marcescens has recently been identified and demonstrated to regulate biofilm formation through the production of surface adhesions. Here we report that mutations in components of the catabolite repression system (cyaA and crp) eliminate flagellum production and swimming motility. Exogenous cAMP was able to restore flagellum production to adenylate cyclase mutants, as determined by transmission electron microscopy and PAGE analysis. A transposon-generated suppressor mutation of the crp motility defect mapped to upstream of the flhDC operon. This suppressor mutation resulted in an upregulation of flhD expression and flagellum production, indicating that flhDC expression is sufficient to restore flagellum production to crp mutants. Lastly, and contrary to a previous report, we found that flhD expression is controlled by the catabolite repression system using quantitative RT-PCR. Together, these data indicate that flagellum production is regulated by the cAMP-dependent catabolite repression system. Given the role of flagella in bacterial pathogenicity, the regulatory pathway described here may assist us in better understanding the putative role of motility in dissemination and virulence of this opportunistic pathogen.


Subject(s)
Bacterial Proteins/metabolism , Cyclic AMP Receptor Protein/metabolism , Down-Regulation , Flagella/genetics , Serratia marcescens/genetics , Bacterial Proteins/genetics , Cyclic AMP Receptor Protein/genetics , Flagella/physiology , Flagella/ultrastructure , Gene Expression Regulation, Bacterial , Humans , Microscopy, Electron, Transmission , Mutation , Serratia Infections/microbiology , Serratia marcescens/physiology , Serratia marcescens/ultrastructure
18.
Appl Environ Microbiol ; 74(11): 3461-70, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18424546

ABSTRACT

The mechanisms by which environmental carbon sources regulate biofilm formation are poorly understood. This study investigates the roles of glucose and the catabolite repression system in Serratia marcescens biofilm formation. The abilities of this opportunistic pathogen to proliferate in a wide range of environments, to cause disease, and to resist antimicrobials are linked to its ability to form biofilms. We observed that growth of S. marcescens in glucose-rich medium strongly stimulated biofilm formation, which contrasts with previous studies showing that biofilm formation is inhibited by glucose in Escherichia coli and other enteric bacteria. Glucose uptake is known to inversely mediate intracellular cyclic AMP (cAMP) synthesis through regulation of adenylate cyclase (cyaA) activity, which in turn controls fundamental processes such as motility, carbon utilization and storage, pathogenesis, and cell division in many bacteria. Here, we demonstrate that mutation of catabolite repression genes that regulate cAMP levels (crr and cyaA) or the ability to respond to cAMP (crp) confers a large increase in biofilm formation. Suppressor analysis revealed that phenotypes of a cAMP receptor protein (crp) mutant require the fimABCD operon, which is responsible for type 1 fimbria production. Consistently, fimA transcription and fimbria production were determined to be upregulated in a cyaA mutant background by using quantitative real-time reverse transcription-PCR and transmission electron microscopy analysis. The regulatory pathway by which environmental carbon sources influence cAMP concentrations to alter production of type 1 fimbrial adhesins establishes a novel mechanism by which bacteria control biofilm development.


Subject(s)
Biofilms/growth & development , Cyclic AMP/metabolism , Fimbriae, Bacterial/metabolism , Gene Expression Regulation, Bacterial , Serratia marcescens/physiology , Bacterial Proteins/genetics , Cyclic AMP Receptor Protein/genetics , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/ultrastructure , Gene Deletion , Gene Expression Profiling , Glucose/metabolism , Microscopy, Electron, Transmission , Molecular Sequence Data , Mutagenesis, Insertional , Phosphoenolpyruvate Sugar Phosphotransferase System/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Serratia marcescens/genetics , Serratia marcescens/ultrastructure
19.
J Bacteriol ; 189(20): 7262-72, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17675374

ABSTRACT

OxyR is a conserved bacterial transcription factor with a regulatory role in oxidative stress response. From a genetic screen for genes that modulate biofilm formation in the opportunistic pathogen Serratia marcescens, mutations in an oxyR homolog and predicted fimbria structural genes were identified. S. marcescens oxyR mutants were severely impaired in biofilm formation, in contrast to the hyperbiofilm phenotype exhibited by oxyR mutants of Escherichia coli and Burkholderia pseudomallei. Further analysis revealed that OxyR plays a role in the primary attachment of cells to a surface. Similar to what is observed in other bacterial species, S. marcescens OxyR is required for oxidative stress resistance. Mutations in oxyR and type I fimbrial genes resulted in severe defects in fimbria-associated phenotypes, revealing roles in cell-cell and cell-biotic surface interactions. Transmission electron microscopy revealed the absence of fimbria-like surface structures on an OxyR-deficient strain and an enhanced fimbrial phenotype in strains bearing oxyR on a multicopy plasmid. The hyperfimbriated phenotype conferred by the multicopy oxyR plasmid was absent in a type I fimbrial mutant background. Real-time reverse transcriptase PCR indicated an absence of transcripts from a fimbrial operon in an oxyR mutant that were present in the wild type and a complemented oxyR mutant strain. Lastly, chromosomal P(lac)-mediated expression of fimABCD was sufficient to restore wild-type levels of yeast agglutination and biofilm formation to an oxyR mutant. Together, these data support a model in which OxyR contributes to early stages of S. marcescens biofilm formation by influencing fimbrial gene expression.


Subject(s)
Bacterial Adhesion/physiology , Biofilms/growth & development , Serratia marcescens/physiology , Transcription Factors/physiology , Bacterial Adhesion/genetics , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Burkholderia pseudomallei/genetics , DNA Transposable Elements , Escherichia coli/genetics , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/ultrastructure , Gene Deletion , Gene Dosage , Gene Expression Regulation, Bacterial , Genetic Complementation Test , Microscopy, Electron, Transmission , Mutagenesis, Insertional , Oxidative Stress/genetics , RNA, Bacterial/biosynthesis , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Serratia marcescens/genetics , Transcription Factors/genetics
20.
Microb Pathog ; 42(5-6): 204-14, 2007.
Article in English | MEDLINE | ID: mdl-17369012

ABSTRACT

Francisella tularensis, the causative agent of tularemia and Category A biodefense agent, is known to replicate within host macrophages, though the pathogenesis of this organism is incompletely understood. We have isolated a variant of F. tularensis live vaccine strain (LVS) based on colony morphology and its effect on macrophages. Human monocyte-derived macrophages produced more tumor necrosis factor alpha (TNFalpha), interleukin (IL)-1beta, IL-6, and IL-12 p40 following exposure to the variant, designated the activating variant (ACV). The immunoreactivity of the lipopolysaccharide (LPS) from both LVS and ACV was comparable to the previously described blue variant and was distinct from the gray variant of LVS. We found, however, the soluble protein fractions of LVS and ACV differed. Further investigation using two-dimensional gel electrophoresis demonstrated higher levels of several proteins in the parental LVS isolate. The differentially expressed proteins featured several associated with virulence in F. tularensis and other pathogens, including intracellular growth locus C (IglC), a sigma(54)-modulation protein family member (YhbH), and aconitase. ACV reverted to the LVS phenotype, indicated by low cytokine induction and high IglC expression, after growth in a chemically defined medium. These data provide evidence that the levels of virulence factors in F. tularensis are modulated based on culture conditions and that this modulation impacts host responses. This work provides a basis for investigation of Francisella virulence factor regulation and the identification of additional factors, co-regulated with IglC, that affect macrophage responses.


Subject(s)
Francisella tularensis/immunology , Macrophages/microbiology , Virulence Factors/biosynthesis , Bacterial Vaccines/microbiology , Cytokines/biosynthesis , Cytokines/immunology , Francisella tularensis/genetics , Francisella tularensis/isolation & purification , Francisella tularensis/pathogenicity , Humans , Macrophages/immunology , Virulence Factors/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...