Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Addiction ; 119(4): 772-783, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38105033

ABSTRACT

AIMS: The aims of this study were to present an enhanced cannabis timeline followback (EC-TLFB) enabling comprehensive assessment of cannabis use measures, including standard tetrahydrocannabinol (THC) units, and to validate these against objectively indexed urinary 11-nor-9-carboxy-tetrahydrocannabinol (THC-COOH) concentrations. DESIGN: We used cross-sectional baseline data from the 'CannTeen' observational longitudinal study. SETTING: The study was conducted in London, UK. PARTICIPANTS: A total of 147 participants who used cannabis regularly took part in the study (n = 71 female, n = 76 male; mean age = 21.90, standard deviation = 5.32). MEASUREMENTS: The EC-TLFB was used to calculate frequency of cannabis use, method of administration, including co-administration with tobacco, amount of cannabis used (measured with unaided self-report and also using pictorial aided self-report) and type of cannabis product (flower, hash) which was used to estimate THC concentration (both from published data on THC concentration of products and analysis of cannabis samples donated by participants in this study). We calculated total weekly standard THC units (i.e. 5 mg THC for all cannabis products and methods of administration) using the EC-TLFB. The outcome variable for validation of past week EC-TLFB assessments was creatinine-normalized carboxy-tetrahydrocannabinol (THC-COOH) in urine. FINDINGS: All measures of cannabis exposure included in this analysis were positively correlated with levels of THC-COOH in urine (r = 0.41-0.52). Standard THC units, calculated with average concentrations of THC in cannabis in the UK and unaided self-report measures of amount of cannabis used in grams showed the strongest correlation with THC-COOH in urine (r = 0.52, 95% bias-corrected and accelerated = 0.26-0.70). CONCLUSIONS: The enhanced cannabis timeline followback (EC-TLFB) can provide a valid assessment of a comprehensive set of cannabis use measures including standard tetrahydrocannabinol units as well as and traditional TLFB assessments (e.g. frequency of use and grams of cannabis use).


Subject(s)
Cannabis , Hallucinogens , Adult , Female , Humans , Male , Young Adult , Cannabinoid Receptor Agonists , Cross-Sectional Studies , Dronabinol , Longitudinal Studies , Observational Studies as Topic
2.
Addict Behav ; 51: 1-6, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26183442

ABSTRACT

PURPOSE: Retail tobacco access is an important determinant of youth smoking prevalence. This study examines perceptions of ease in obtaining cigarettes and how prevalence of self-reported retail tobacco access among youth smokers varies by province in Canada. Additionally, relevant retail experiences, such as being asked for identification by a store clerk, are described. METHODS: Data from grades 9-12 students who participated in the 2010/2011 Youth Smoking Survey, a nationally generalizable sample of Canadian students (n=31396) were used to examine retail tobacco access and related experiences. Logistic regression models were used to examine differences in retail tobacco access and retail tobacco experiences by sociodemographic and regional characteristics. RESULTS: 79% of students who never smoked thought it would be easy to get cigarettes. About one-quarter of smokers reported usually buying cigarettes from stores, and the percent of student smokers usually buying cigarettes in stores ranged from 16% in British Columbia to 36% in Quebec. Compared to grade 9 students, grade 12 students had higher odds of report being asked for identification (OR=6.3, 95% CI 1.9-21.5). CONCLUSIONS: Retail tobacco access appears to be a significant source of cigarette access among Canadian youth. Retail tobacco access varies significantly by province, which suggests provincial policies should be strengthened.


Subject(s)
Adolescent Behavior , Commerce/statistics & numerical data , Health Knowledge, Attitudes, Practice , Tobacco Products/statistics & numerical data , Adolescent , Canada , Female , Humans , Male , Self Report , Socioeconomic Factors , Tobacco Products/economics
3.
PLoS One ; 8(2): e56834, 2013.
Article in English | MEDLINE | ID: mdl-23457625

ABSTRACT

Different Francisella spp. produce five or six predicted acid phosphatases (AcpA, AcpB, AcpC, AcpD, HapA and HapB). The genes encoding the histidine acid phosphatases (hapA, hapB) and acpD of F. tularensis subsp. Schu S4 strain are truncated or disrupted. However, deletion of HapA (FTT1064) in F. tularensis Schu S4 resulted in a 33% reduction in acid phosphatase activity and loss of the four functional acid phosphatases in F. tularensis Schu S4 (ΔABCH) resulted in a>99% reduction in acid phosphatase activity compared to the wild type strain. All single, double and triple mutants tested, demonstrated a moderate decrease in mouse virulence and survival and growth within human and murine phagocytes, whereas the ΔABCH mutant showed >3.5-fold decrease in intramacrophage survival and 100% attenuation of virulence in mouse. While the Schu S4 ΔABCH strain was attenuated in the mouse model, it showed only limited protection against wild type challenge. F. tularensis Schu S4 failed to stimulate reactive oxygen species production in phagocytes, whereas infection by the ΔABCH strain stimulated 5- and 56-fold increase in reactive oxygen species production in neutrophils and human monocyte-derived macrophages, respectively. The ΔABCH mutant but not the wild type strain strongly co-localized with p47 (phox) and replicated in macrophages isolated from p47 (phox) knockout mice. Thus, F. tularensis Schu S4 acid phosphatases, including the truncated HapA, play a major role in intramacrophage survival and virulence of this human pathogen.


Subject(s)
Acid Phosphatase/metabolism , Francisella tularensis/enzymology , Francisella tularensis/physiology , Acid Phosphatase/deficiency , Acid Phosphatase/genetics , Animals , Female , Francisella tularensis/genetics , Gene Deletion , Humans , Macrophages/cytology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Monocytes/cytology , Monocytes/microbiology , NADPH Oxidases/metabolism , Neutrophils/cytology , Neutrophils/microbiology , Phagosomes/microbiology , Protein Transport , Reactive Oxygen Species/metabolism
4.
J Immunol ; 184(9): 5141-50, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20348422

ABSTRACT

Francisella tularensis contains four putative acid phosphatases that are conserved in Francisella novicida. An F. novicida quadruple mutant (AcpA, AcpB, AcpC, and Hap [DeltaABCH]) is unable to escape the phagosome or survive in macrophages and is attenuated in the mouse model. We explored whether reduced survival of the DeltaABCH mutant within phagocytes is related to the oxidative response by human neutrophils and macrophages. F. novicida and F. tularensis subspecies failed to stimulate reactive oxygen species production in the phagocytes, whereas the F. novicida DeltaABCH strain stimulated a significant level of reactive oxygen species. The DeltaABCH mutant, but not the wild-type strain, strongly colocalized with p47(phox) and replicated in phagocytes only in the presence of an NADPH oxidase inhibitor or within macrophages isolated from p47(phox) knockout mice. Finally, purified AcpA strongly dephosphorylated p47(phox) and p40(phox), but not p67(phox), in vitro. Thus, Francisella acid phosphatases play a major role in intramacrophage survival and virulence by regulating the generation of the oxidative burst in human phagocytes.


Subject(s)
Acid Phosphatase/physiology , Francisella tularensis/enzymology , Francisella tularensis/immunology , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Phagocytes/enzymology , Phagocytes/microbiology , Acid Phosphatase/genetics , Animals , Cells, Cultured , Francisella tularensis/growth & development , Humans , Intracellular Fluid/enzymology , Intracellular Fluid/immunology , Intracellular Fluid/microbiology , Isoenzymes/genetics , Isoenzymes/physiology , Macrophages/enzymology , Macrophages/immunology , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidases/biosynthesis , Neutrophils/enzymology , Neutrophils/immunology , Neutrophils/microbiology , Phagocytes/immunology , Phosphorylation/immunology , Reactive Oxygen Species/metabolism , Respiratory Burst/immunology
5.
Front Microbiol ; 1: 129, 2010.
Article in English | MEDLINE | ID: mdl-21687776

ABSTRACT

Francisella tularensis is a CDC Category A biological agent and a potential bioterrorist threat. There is no licensed vaccine against tularemia in the United States. A long-standing issue with potential Francisella vaccines is strain phase variation to a gray form that lacks protective capability in animal models. Comparisons of the parental strain (LVS) and a gray variant (LVSG) have identified lipopolysaccharide (LPS) alterations as a primary change. The LPS of the F. tularensis variant strain gains reactivity to F. novicida anti-LPS antibodies, suggesting structural alterations to the O-antigen. However, biochemical and structural analysis of the F. tularensis LVSG and LVS LPS demonstrated that LVSG has less O-antigen but no major O-antigen structural alterations. Additionally, LVSG possesses structural differences in both the core and lipid A regions, the latter being decreased galactosamine modification. Recent work has identified two genes important in adding galactosamine (flmF2 and flmK) to the lipid A. Quantitative real-time PCR showed reduced transcripts of both of these genes in the gray variant when compared to LVS. Loss of flmF2 or flmK caused less frequent phase conversion but did not alter intramacrophage survival or colony morphology. The LVSG strain demonstrated an intramacrophage survival defect in human and rat but not mouse macrophages. Consistent with this result, the LVSG variant demonstrated little change in LD(50) in the mouse model of infection. Furthermore, the LVSG strain lacks the protective capacity of F. tularensis LVS against virulent Type A challenge. These data suggest that the LPS of the F. tularensis LVSG phase variant is dramatically altered. Understanding the mechanism of blue to gray phase variation may lead to a way to inhibit this variation, thus making future F. tularensis vaccines more stable and efficacious.

6.
Antimicrob Agents Chemother ; 54(1): 244-53, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19917753

ABSTRACT

Francisella tularensis requires iron (Fe) for growth, but the biologic sources of Fe for this organism are largely unknown. We found that Francisella sp. growing in broth culture or within human macrophages can acquire Fe from the two major host Fe-binding proteins, lactoferrin (Lf) and transferrin (Tf). Fe acquisition is a potential target for novel therapies. Gallium (Ga) is a transition metal that interferes with cellular Fe metabolism by competing with Fe for uptake/utilization. Growth of either F. tularensis live vaccine strain (LVS) or Francisella novicida was inhibited by >or=2 microM Ga chelated to Tf or Lf, with GaLf being somewhat more potent. Francisella spp. express two Fe-containing antioxidant enzymes, catalase (KatG) and Fe cofactored superoxide dismutase (FeSOD). Growth of LVS with 10 muM GaTf or GaLf led to a dramatic decrease in bacterial catalase activity and in FeSOD activity that was associated with an increased susceptibility to H(2)O(2). Ga also protected mice from intranasal challenge with F. novicida. Whereas 100% of the F. novicida-infected mice died by day 9, 75% of the mice receiving Ga continued to survive to at least day 15. Thus, a single intranasal dose of Ga followed by daily intraperitoneal Ga at a dose tolerated by the animals resulted in prolonged survival. These data support the potential utility of Ga as a therapy for F. tularensis infection of the lung.


Subject(s)
Francisella/drug effects , Francisella/metabolism , Gallium/pharmacology , Gallium/therapeutic use , Gram-Negative Bacterial Infections/metabolism , Iron/metabolism , Lung Diseases/drug therapy , Lung Diseases/metabolism , Adult , Animals , Antioxidants/metabolism , Catalase/metabolism , Drug Resistance, Bacterial , Female , Francisella tularensis , Gram-Negative Bacterial Infections/microbiology , Humans , Hydrogen Peroxide/pharmacology , Lung Diseases/microbiology , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Superoxide Dismutase/metabolism , Tularemia/metabolism , Tularemia/microbiology
7.
J Biomed Sci ; 16: 110, 2009 Dec 09.
Article in English | MEDLINE | ID: mdl-20003180

ABSTRACT

BACKGROUND: Autophagy has been shown recently to play an important role in the intracellular survival of several pathogenic bacteria. In this study, we investigated the effect of a novel small-molecule autophagy-inducing agent, AR-12, on the survival of Francisella tularensis, the causative bacterium of tularemia in humans and a potential bioterrorism agent, in macrophages. METHODS AND RESULTS: Our results show that AR-12 induces autophagy in THP-1 macrophages, as indicated by increased autophagosome formation, and potently inhibits the intracellular survival of F. tularensis (type A strain, Schu S4) and F. novicida in macrophages in association with increased bacterial co-localization with autophagosomes. The effect of AR-12 on intracellular F. novicida was fully reversed in the presence of the autophagy inhibitor, 3-methyl adenine or the lysosome inhibitor, chloroquine. Intracellular F. novicida were not susceptible to the inhibitory activity of AR-12 added at 12 h post-infection in THP-1 macrophages, and this lack of susceptibility was independent of the intracellular location of bacteria. CONCLUSION: Together, AR-12 represents a proof-of-principle that intracellular F. tularensis can be eradicated by small-molecule agents that target innate immunity.


Subject(s)
Autophagy , Francisella tularensis/metabolism , Macrophages/microbiology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Tularemia/microbiology , Anti-Infective Agents/chemistry , Bacterial Vaccines/immunology , Cell Line , Chloroquine/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Lysosomes/metabolism , Microscopy, Fluorescence/methods , Time Factors
8.
Antimicrob Agents Chemother ; 53(12): 5236-44, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19805568

ABSTRACT

Eradication of intracellular pathogenic bacteria with host-directed chemical agents has been an anticipated innovation in the treatment of antibiotic-resistant bacteria. We previously synthesized and characterized a novel small-molecule agent, AR-12, that induces autophagy and inhibits the Akt kinase in cancer cells. As both autophagy and the Akt kinase have been shown recently to play roles in the intracellular survival of several intracellular bacteria, including Salmonella enterica serovar Typhimurium, we investigated the effect of AR-12 on the intracellular survival of Salmonella serovar Typhimurium in macrophages. Our results show that AR-12 induces autophagy in macrophages, as indicated by increased autophagosome formation, and potently inhibits the survival of serovar Typhimurium in macrophages in association with increased colocalization of intracellular bacteria with autophagosomes. Intracellular bacterial growth was partially rescued in the presence of AR-12 by the short hairpin RNA-mediated knockdown of Beclin-1 or Atg7 in macrophages. Moreover, AR-12 inhibits Akt kinase activity in infected macrophages, which we show to be important for its antibacterial effect as the enforced expression of constitutively activated Akt1 in these cells reverses the AR-12-induced inhibition of intracellular serovar Typhimurium survival. Finally, oral administration of AR-12 at 2.5 mg/kg/day to serovar Typhimurium-infected mice reduced hepatic and splenic bacterial burdens and significantly prolonged survival. These findings show that AR-12 represents a proof of principle that the survival of intracellular bacteria can be suppressed by small-molecule agents that target both innate immunity and host cell factors modulated by bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Salmonella Infections , Salmonella typhimurium/drug effects , Animals , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/therapeutic use , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/physiology , Autophagy/drug effects , Autophagy-Related Protein 7 , Beclin-1 , Cell Survival/drug effects , Female , Immunoblotting , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/physiology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/physiology , Salmonella Infections/drug therapy , Salmonella Infections/microbiology , Salmonella typhimurium/metabolism
9.
Antimicrob Agents Chemother ; 53(7): 2998-3002, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19398640

ABSTRACT

Francisella tularensis, a bacterium which causes tularemia in humans, is classified as a CDC category A bioterrorism agent. In this study, we demonstrate that celecoxib, an anti-inflammatory cyclooxygenase-2 inhibitor in clinical use, exhibits activity against a type A strain of F. tularensis (Schu S4), the live vaccine strain of F. tularensis (a type B strain), and F. novicida ("F. tularensis subsp. novicida") directly in growth medium. This bacterial killing, however, was not noted with rofecoxib, despite its higher potency than that of celecoxib in inhibiting cyclooxygenase-2. The unique ability of celecoxib to inhibit the proliferation of F. tularensis could be pharmacologically exploited to develop novel anti-Francisella therapeutic agents, of which the proof of principle is demonstrated by compound 20, a celecoxib derivative identified through the screening of a celecoxib-based focused compound library. Compound 20 inhibited the intracellular proliferation of Francisella in macrophages without causing appreciable toxicity to these host cells. Together, these data support the translational potential of compound 20 for the further development of novel, potent anti-Francisella agents.


Subject(s)
Cyclooxygenase 2 Inhibitors/pharmacology , Francisella tularensis/drug effects , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Animals , Celecoxib , Cell Line , Cell Survival/drug effects , Humans , Lactones/pharmacology , Mice , Microbial Sensitivity Tests , Sulfones/pharmacology
10.
J Immunol ; 181(8): 5568-78, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18832715

ABSTRACT

The bacterium Francisella tularensis (Ft) is a potential weapon of bioterrorism when aerosolized. Macrophage infection is necessary for disease progression and efficient phagocytosis by human macrophages requires serum opsonization by complement. Microbial complement activation leads to surface deposition of a highly regulated protein complex resulting in opsonization or membrane lysis. The nature of complement component C3 deposition, i.e., C3b (opsonization and lysis) or C3bi (opsonization only) fragment deposition, is central to the outcome of activation. In this study, we examine the mechanisms of Ft resistance to complement-mediated lysis, C3 component deposition on the Ft surface, and complement activation. Upon incubation in fresh nonimmune human serum, Schu S4 (Ft subsp. tularensis), Fn (Ft subsp. novicida), and LVS (Ft subsp. holarctica live vaccine strain) were resistant to complement-mediated lysis, but LVSG and LVSR (LVS strains altered in surface carbohydrate structures) were susceptible. C3 deposition, however, occurred on all strains. Complement-susceptible strains had markedly increased C3 fragment deposition, including the persistent presence of C3b compared with C3bi, which indicates that C3b inactivation results in survival of complement-resistant strains. C1q, an essential component of the classical activation pathway, was necessary for lysis of complement-susceptible strains and optimal C3 deposition on all strains. Finally, use of Francisella LPS mutants confirmed O Ag as a major regulator of complement resistance. These data provide evidence that pathogenic Francisella activate complement, but are resistant to complement-mediated lysis in part due to limited C3 deposition, rapid conversion of surface-bound C3b to C3bi, and the presence of LPS O Ag.


Subject(s)
Complement Activation/immunology , Complement C3/immunology , Francisella tularensis/immunology , Macrophages/immunology , O Antigens/immunology , Tularemia/immunology , Biological Warfare Agents , Bioterrorism , Cell Membrane/genetics , Cell Membrane/immunology , Complement Activation/genetics , Complement C1q/genetics , Complement C1q/immunology , Complement C3/genetics , Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Humans , Macrophages/microbiology , Mutation , O Antigens/genetics , Phagocytosis/genetics , Phagocytosis/immunology , Tularemia/genetics
11.
PLoS One ; 3(6): e2487, 2008 Jun 25.
Article in English | MEDLINE | ID: mdl-18575611

ABSTRACT

BACKGROUND: Francisella tularensis is a gram-negative coccobacillus that causes the febrile illness tularemia. Subspecies that are pathogenic for humans include those comprising the type A (subspecies tularensis) or type B (subspecies holarctica) biovars. An attenuated live vaccine strain (LVS) developed from a type B isolate has previously been used to vaccinate at-risk individuals, but offers limited protection against high dose (>1000 CFUs) challenge with type A strains delivered by the respiratory route. Due to differences between type A and type B F. tularensis strains at the genetic level, it has been speculated that utilization of an attenuated type A strain as a live vaccine might offer better protection against homologous respiratory challenge compared with LVS. Here, we report the construction and characterization of an unmarked Delta purMCD mutant in the highly virulent type A strain Schu S4. METHODOLOGY/PRINCIPAL FINDINGS: Growth of Schu S4 Delta purMCD was severely attenuated in primary human peripheral blood monocyte-derived macrophages and in the A549 human lung epithelial cell line. The Schu S4 Delta purMCD mutant was also highly attenuated in mice when delivered via either the intranasal or intradermal infection route. Mice vaccinated intranasally with Schu S4 Delta purMCD were well protected against high dose intradermal challenge with virulent type A or type B strains of F. tularensis. However, intranasal vaccination with Schu S4 Delta purMCD induced tissue damage in the lungs, and conferred only limited protection against high dose Schu S4 challenge delivered by the same route. The level of protection observed was similar to that conferred following vaccination with wild-type LVS or the analogous LVS Delta purMCD mutant. CONCLUSIONS/SIGNIFICANCE: Collectively, these results argue that development of the next generation live attenuated vaccine for Francisella should be based on use of the less pathogenic type B biovar rather than the more reactogenic type A biovar.


Subject(s)
Bacterial Vaccines/immunology , Francisella tularensis/immunology , Purines/immunology , Administration, Intranasal , Animals , Bacterial Vaccines/administration & dosage , Cell Line , Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Humans , Lung/microbiology , Macrophages/microbiology , Mice , Mutation , Virulence
12.
Infect Immun ; 76(8): 3690-9, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18490464

ABSTRACT

Francisella tularensis is a facultative intracellular pathogen and the etiologic agent of tularemia. It is capable of escape from macrophage phagosomes and replicates in the host cell cytosol. Bacterial acid phosphatases are thought to play a major role in the virulence and intracellular survival of a number of intracellular pathogens. The goal of this study was to delete the four primary acid phosphatases (Acps) from Francisella novicida and examine the interactions of mutant strains with macrophages, as well as the virulence of these strains in mice. We constructed F. novicida mutants with various combinations of acp deletions and showed that loss of the four Acps (AcpA, AcpB, AcpC, and histidine acid phosphatase [Hap]) in an F. novicida strain (DeltaABCH) resulted in a 90% reduction in acid phosphatase activity. The DeltaABCH mutant was defective for survival/growth within human and murine macrophage cell lines and was unable to escape from phagosome vacuoles. With accumulation of Acp deletions, a progressive loss of virulence in the mouse model was observed. The DeltaABCH strain was dramatically attenuated and was an effective single-dose vaccine against homologous challenge. Furthermore, both acpA and hap were induced when the bacteria were within host macrophages. Thus, the Francisella acid phosphatases cumulatively play an important role in intracellular trafficking and virulence.


Subject(s)
Acid Phosphatase/genetics , Bacterial Proteins/genetics , Francisella tularensis/enzymology , Francisella tularensis/pathogenicity , Macrophages/microbiology , Phagosomes/microbiology , Virulence Factors/genetics , Animals , Cell Line , Colony Count, Microbial , Female , Francisella tularensis/genetics , Francisella tularensis/growth & development , Gene Deletion , Gene Order , Humans , Liver/microbiology , Mice , Mice, Inbred BALB C , Spleen/microbiology , Survival Analysis , Virulence
13.
Infect Immun ; 75(7): 3305-14, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17452468

ABSTRACT

Francisella tularensis is a category A agent of biowarfare/biodefense. Little is known about the regulation of virulence gene expression in Francisella spp. Comparatively few regulatory factors exist in Francisella, including those belonging to two-component systems (TCS). However, orphan members of typical TCS can be identified. To determine if orphan TCS members affect Francisella gene expression, a gene encoding a product with high similarity to the Salmonella PmrA response regulator (FTT1557c/FNU0663.2) was deleted in Francisella novicida (a model organism for F. tularensis). The F. novicida pmrA mutant was defective in survival/growth within human and murine macrophage cell lines and was 100% defective in virulence in mice at a dose of up to 10(8) CFU. In addition, the mutant strain demonstrated increased susceptibility to antimicrobial peptide killing, but no differences were observed between the lipid A of the mutant and the parental strain, as has been observed with pmrA mutants of other microbes. The F. novicida pmrA mutant was 100% protective as a single-dose vaccine when challenge was with 10(6) CFU of F. novicida but did not protect against type A Schu S4 wild-type challenge. DNA microarray analysis identified 65 genes regulated by PmrA. The majority of these genes were located in the region surrounding pmrA or within the Francisella pathogenicity island (FPI). These FPI genes are also regulated by MglA, but MglA does not regulate pmrA, nor does PmrA regulate MglA. Thus, the orphan response regulator PmrA is an important factor in controlling virulence in F. novicida, and a pmrA mutant strain is an effective vaccine against homologous challenge.


Subject(s)
Bacterial Proteins/metabolism , Francisella/pathogenicity , Gene Expression Regulation, Bacterial , Genomic Islands/genetics , Signal Transduction , Animals , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Vaccines/administration & dosage , Cell Line , Female , Francisella/genetics , Francisella/immunology , Francisella/metabolism , Gene Expression Profiling , Genomic Islands/physiology , Gram-Negative Bacterial Infections/prevention & control , Humans , Macrophages , Mice , Mice, Inbred BALB C , Mutation , Oligonucleotide Array Sequence Analysis , Virulence
14.
Infect Immun ; 75(1): 390-6, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17060465

ABSTRACT

AcpA of Francisella spp. is a respiratory-burst-inhibiting acid phosphatase that also exhibits phospholipase C activity. To better understand the molecular basis of AcpA in virulence, a deletion of acpA was constructed in Francisella novicida. The phosphatase and lipase activities were reduced 10-fold and 8-fold, respectively, in the acpA mutant compared to the wild type and were found mostly associated with the outer membrane. The acpA mutant was more susceptible to intracellular killing than the wild-type strain in the THP-1 human macrophage-like cell line. In addition, mice infected with the acpA mutant survived longer than the wild-type strain and were less fit than the wild-type strain in competition infection assays. Transmission electron microscopy showed that the acpA mutant was delayed in escape from macrophage phagosomes, as more than 75% of acpA mutant bacteria could still be found inside phagosomes after 12 h of infection in THP-1 cells and human monocyte-derived macrophages, whereas most of the wild-type bacteria had escaped from the phagosome by 6 h postinfection. Thus, AcpA affects intracellular trafficking and the fate of Francisella within host macrophages.


Subject(s)
Acid Phosphatase/immunology , Bacterial Proteins/immunology , Francisella/pathogenicity , Macrophages/immunology , Macrophages/microbiology , Acid Phosphatase/metabolism , Animals , Bacterial Proteins/metabolism , Cell Survival , Female , Francisella/enzymology , Humans , Mice , Mice, Inbred BALB C , Microscopy, Electron, Transmission , Phagosomes/immunology , Phagosomes/microbiology , Polymerase Chain Reaction , Virulence
15.
Infect Immun ; 74(9): 5114-25, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16926403

ABSTRACT

Inhalational pneumonic tularemia, caused by Francisella tularensis, is lethal in humans. F. tularensis is phagocytosed by macrophages followed by escape from phagosomes into the cytoplasm. Little is known of the phagocytic mechanisms for Francisella, particularly as they relate to the lung and alveolar macrophages. Here we examined receptors on primary human monocytes and macrophages which mediate the phagocytosis and intracellular survival of F. novicida. F. novicida association with monocyte-derived macrophages (MDM) was greater than with monocytes. Bacteria were readily ingested, as shown by electron microscopy. Bacterial association was significantly increased in fresh serum and only partially decreased in heat-inactivated serum. A role for both complement receptor 3 (CR3) and Fcgamma receptors in uptake was supported by studies using a CR3-expressing cell line and by down-modulation of Fcgamma receptors on MDM, respectively. Consistent with Fcgamma receptor involvement, antibody in nonimmune human serum was detected on the surface of Francisella. In the absence of serum opsonins, competitive inhibition of mannose receptor (MR) activity on MDM with mannan decreased the association of F. novicida and opsonization of F. novicida with lung collectin surfactant protein A (SP-A) increased bacterial association and intracellular survival. This study demonstrates that human macrophages phagocytose more Francisella than monocytes with contributions from CR3, Fcgamma receptors, the MR, and SP-A present in lung alveoli.


Subject(s)
Francisella tularensis/immunology , Macrophages/microbiology , Pneumonia, Bacterial/immunology , Receptors, Cell Surface/physiology , Tularemia/immunology , Antibodies, Bacterial/blood , Francisella tularensis/ultrastructure , Humans , Lectins, C-Type/physiology , Ligands , Lung/immunology , Macrophage-1 Antigen/physiology , Macrophages/immunology , Mannose Receptor , Mannose-Binding Lectins/physiology , Microscopy, Electron, Transmission , Monocytes/immunology , Monocytes/microbiology , Phagocytosis , Pulmonary Surfactant-Associated Protein A/physiology , Receptors, IgG/physiology , Serum/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...