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










Publication year range
1.
PLoS Comput Biol ; 16(6): e1007752, 2020 06.
Article in English | MEDLINE | ID: mdl-32479491

ABSTRACT

We study the pathogenesis of Francisella tularensis infection with an experimental mouse model, agent-based computation and mathematical analysis. Following inhalational exposure to Francisella tularensis SCHU S4, a small initial number of bacteria enter lung host cells and proliferate inside them, eventually destroying the host cell and releasing numerous copies that infect other cells. Our analysis of disease progression is based on a stochastic model of a population of infectious agents inside one host cell, extending the birth-and-death process by the occurrence of catastrophes: cell rupture events that affect all bacteria in a cell simultaneously. Closed expressions are obtained for the survival function of an infected cell, the number of bacteria released as a function of time after infection, and the total bacterial load. We compare our mathematical analysis with the results of agent-based computation and, making use of approximate Bayesian statistical inference, with experimental measurements carried out after murine aerosol infection with the virulent SCHU S4 strain of the bacterium Francisella tularensis, that infects alveolar macrophages. The posterior distribution of the rate of replication of intracellular bacteria is consistent with the estimate that the time between rounds of bacterial division is less than 6 hours in vivo.


Subject(s)
Francisella tularensis/cytology , Lung/microbiology , Tularemia/microbiology , Animals , Bayes Theorem , Computational Biology , Cytosol/metabolism , Disease Models, Animal , Female , Macrophages, Alveolar/microbiology , Mice , Mice, Inbred BALB C , Models, Theoretical , Phagosomes/metabolism , Probability , Stochastic Processes , Virulence
2.
Microbiol Mol Biol Rev ; 84(1)2020 05 20.
Article in English | MEDLINE | ID: mdl-32051235

ABSTRACT

Francisella tularensis is a tier 1 select agent responsible for tularemia in humans and a wide variety of animal species. Extensive research into understanding the virulence factors of the bacterium has been ongoing to develop an efficacious vaccine. At least two such virulence factors are described as capsules of F. tularensis: the O-antigen capsule and the capsule-like complex (CLC). These two separate entities aid in avoiding host immune defenses but have not been clearly differentiated. These components are distinct and differ in composition and genetic basis. The O-antigen capsule consists of a polysaccharide nearly identical to the lipopolysaccharide (LPS) O antigen, whereas the CLC is a heterogeneous complex of glycoproteins, proteins, and possibly outer membrane vesicles and tubes (OMV/Ts). In this review, the current understanding of these two capsules is summarized, and the historical references to "capsules" of F. tularensis are clarified. A significant amount of research has been invested into the composition of each capsule and the genes involved in synthesis of the polysaccharide portion of each capsule. Areas of future research include further exploration into the molecular regulation and pathways responsible for expression of each capsule and further elucidating the role that each capsule plays in virulence.


Subject(s)
Bacterial Capsules/metabolism , Francisella tularensis/cytology , Polysaccharides, Bacterial/metabolism , Animals , Francisella tularensis/genetics , Humans , Tularemia/microbiology , Virulence , Virulence Factors
3.
Lab Chip ; 14(13): 2327-33, 2014 Jul 07.
Article in English | MEDLINE | ID: mdl-24828279

ABSTRACT

Detection of pathogenic bacteria requires a sensitive, accurate, rapid, and portable device. Given that lethal microbes are of various sizes, bacterial sensors based on DC (direct current) impedance on chips should be equipped with channels with commensurate cross sections. When it comes to counting and interrogation of individual bacteria on a microfluidic chip, very narrow channels are required, which are neither easy nor cost-effective to fabricate. Here, we report a flow cytometry-based submicron-sized bacterial detection system using a movable virtual wall made of a non-conducting fluid. We show that the effective dimension of a microfluidic channel can be adjusted by varying the respective flow rates of a sample solution as well as the liquid wall therein. Using such a virtual wall, we have successfully controlled the channel width and detected submicron-sized Francisella tularensis, a lethal, tularemia-causing bacterium. Since the system is capable of monitoring changes in DC impedance and fluorescence simultaneously, we were also able to discriminate between different types of bacterial mixtures containing F. tularensis and E. coli BL21 that have different gamuts of size distributions. The proposed flow cytometry-based system represents a promising way to detect bacteria including, but not limited to, submicron-sized pathogenic microbes.


Subject(s)
Bacterial Typing Techniques , Escherichia coli/cytology , Flow Cytometry , Francisella tularensis/cytology , Microfluidic Analytical Techniques , Bacterial Typing Techniques/instrumentation , Bacterial Typing Techniques/methods , Escherichia coli/classification , Flow Cytometry/instrumentation , Flow Cytometry/methods , Francisella tularensis/classification , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods
4.
Infect Immun ; 82(7): 2935-48, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24778115

ABSTRACT

The Francisella FTT0831c/FTL_0325 gene encodes amino acid motifs to suggest it is a lipoprotein and that it may interact with the bacterial cell wall as a member of the OmpA-like protein family. Previous studies have suggested that FTT0831c is surface exposed and required for virulence of Francisella tularensis by subverting the host innate immune response (M. Mahawar et al., J. Biol. Chem. 287:25216-25229, 2012). We also found that FTT0831c is required for murine pathogenesis and intramacrophage growth of Schu S4, but we propose a different model to account for the proinflammatory nature of the resultant mutants. First, inactivation of FTL_0325 from live vaccine strain (LVS) or FTT0831c from Schu S4 resulted in temperature-dependent defects in cell viability and morphology. Loss of FTT0831c was also associated with an unusual defect in lipopolysaccharide O-antigen synthesis, but loss of FTL_0325 was not. Full restoration of these properties was observed in complemented strains expressing FTT0831c in trans, but not in strains lacking the OmpA motif, suggesting that cell wall contact is required. Finally, growth of the LVS FTL_0325 mutant in Mueller-Hinton broth at 37°C resulted in the appearance of membrane blebs at the poles and midpoint, prior to the formation of enlarged round cells that showed evidence of compromised cellular membranes. Taken together, these data are more consistent with the known structural role of OmpA-like proteins in linking the OM to the cell wall and, as such, maintenance of structural integrity preventing altered surface exposure or release of Toll-like receptor 2 agonists during rapid growth of Francisella in vitro and in vivo.


Subject(s)
Bacterial Proteins/metabolism , Francisella tularensis/cytology , Francisella tularensis/physiology , Gene Expression Regulation, Bacterial/physiology , Animals , Bacterial Proteins/genetics , Cell Shape , Female , Francisella tularensis/genetics , Gene Deletion , Genetic Complementation Test , Immunity, Innate , Mice , Mice, Inbred C3H , Tularemia/microbiology
5.
PLoS One ; 6(7): e22335, 2011.
Article in English | MEDLINE | ID: mdl-21799828

ABSTRACT

BACKGROUND: The gram-negative bacterium Francisella tularensis survives in arthropods, fresh water amoeba, and mammals with both intracellular and extracellular phases and could reasonably be expected to express distinct phenotypes in these environments. The presence of a capsule on this bacterium has been controversial with some groups finding such a structure while other groups report that no capsule could be identified. Previously we reported in vitro culture conditions for this bacterium which, in contrast to typical methods, yielded a bacterial phenotype that mimics that of the bacterium's mammalian, extracellular phase. METHODS/FINDINGS: SDS-PAGE and carbohydrate analysis of differentially-cultivated F. tularensis LVS revealed that bacteria displaying the host-adapted phenotype produce both longer polymers of LPS O-antigen (OAg) and additional HMW carbohydrates/glycoproteins that are reduced/absent in non-host-adapted bacteria. Analysis of wildtype and OAg-mutant bacteria indicated that the induced changes in surface carbohydrates involved both OAg and non-OAg species. To assess the impact of these HMW carbohydrates on the access of outer membrane constituents to antibody we used differentially-cultivated bacteria in vitro to immunoprecipitate antibodies directed against outer membrane moieties. We observed that the surface-carbohydrates induced during host-adaptation shield many outer membrane antigens from binding by antibody. Similar assays with normal mouse serum indicate that the induced HMW carbohydrates also impede complement deposition. Using an in vitro macrophage infection assay, we find that the bacterial HMW carbohydrate impedes TLR2-dependent, pro-inflammatory cytokine production by macrophages. Lastly we show that upon host-adaptation, the human-virulent strain, F. tularensis SchuS4 also induces capsule production with the effect of reducing macrophage-activation and accelerating tularemia pathogenesis in mice. CONCLUSION: F. tularensis undergoes host-adaptation which includes production of multiple capsular materials. These capsules impede recognition of bacterial outer membrane constituents by antibody, complement, and Toll-Like Receptor 2. These changes in the host-pathogen interface have profound implications for pathogenesis and vaccine development.


Subject(s)
Adaptation, Physiological/immunology , Adaptive Immunity , Bacterial Capsules/biosynthesis , Francisella tularensis/immunology , Francisella tularensis/metabolism , Immunity, Innate , Lipopolysaccharides/biosynthesis , Animals , Antibodies, Bacterial/immunology , Bacterial Capsules/chemistry , Bacterial Outer Membrane Proteins/metabolism , Cell Membrane/metabolism , Complement System Proteins/metabolism , Extracellular Space/metabolism , Francisella tularensis/cytology , Francisella tularensis/growth & development , Humans , Lipopolysaccharides/chemistry , Mice , Molecular Weight , O Antigens/biosynthesis , O Antigens/chemistry , Phenotype , Toll-Like Receptor 2/metabolism , Tularemia/immunology , Tularemia/microbiology
6.
Curr Protoc Immunol ; Chapter 19: Unit 19.14, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21462168

ABSTRACT

This unit describes the utility of various mouse models of infection for studying pathogenesis and adaptive immune responses to the facultative intracellular bacteria pathogen Francisella tularensis. By judicious use of different combinations of mouse and bacterial strains, as well as different routes of infection, murine tularemia models may be used to explore a complete picture of F. tularensis infection and immunity. Moreover, studies using Francisella, particularly the Live Vaccine Strain (LVS), serve as a convenient and tractable model system that appears to be representative of mammalian host responses to intracellular pathogens in general.


Subject(s)
Disease Models, Animal , Tularemia/immunology , Tularemia/microbiology , Animal Structures/immunology , Animal Structures/microbiology , Animals , Bacteremia/immunology , Bacteriological Techniques/methods , Ear Auricle/immunology , Ear Auricle/microbiology , Francisella tularensis/cytology , Francisella tularensis/immunology , Francisella tularensis/pathogenicity , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Immunologic Techniques , Mice , Mice, Inbred Strains , Muscle, Skeletal/immunology , Muscle, Skeletal/microbiology , Peritoneal Cavity/microbiology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , Skin Diseases, Bacterial/immunology , Skin Diseases, Bacterial/microbiology , Tularemia/etiology
7.
J Biol Chem ; 285(36): 27839-49, 2010 Sep 03.
Article in English | MEDLINE | ID: mdl-20605777

ABSTRACT

The O-antigen polymerase of gram-negative bacteria has been difficult to characterize. Herein we report the biochemical and functional characterization of the protein product (Wzy) of the gene annotated as the putative O-antigen polymerase, which is located in the O-antigen biosynthetic locus of Francisella tularensis. In silico analysis (homology searching, hydropathy plotting, and codon usage assessment) strongly suggested that Wzy is an O-antigen polymerase whose function is to catalyze the addition of newly synthesized O-antigen repeating units to a glycolipid consisting of lipid A, inner core polysaccharide, and one repeating unit of the O-polysaccharide (O-PS). To characterize the function of the Wzy protein, a non-polar deletion mutant of wzy was generated by allelic replacement, and the banding pattern of O-PS was observed by immunoblot analysis of whole-cell lysates obtained by SDS-PAGE and stained with an O-PS-specific monoclonal antibody. These immunoblot analyses showed that O-PS of the wzy mutant expresses only one repeating unit of O-antigen. Further biochemical characterization of the subcellular fractions of the wzy mutant demonstrated that (as is characteristic of O-antigen polymerase mutants) the low molecular weight O-antigen accumulates in the periplasm of the mutant. Site-directed mutagenesis based on protein homology and topology, which was carried out to locate a catalytic residue of the protein, showed that modification of specific residues (Gly(176), Asp(177), Gly(323), and Tyr(324)) leads to a loss of O-PS polymerization. Topology models indicate that these amino acids most likely lie in close proximity on the bacterial surface.


Subject(s)
Francisella tularensis/enzymology , Hexosyltransferases/metabolism , Amino Acid Sequence , Amino Acids/metabolism , Biocatalysis , Cell Membrane/metabolism , Francisella tularensis/cytology , Francisella tularensis/genetics , Gene Expression Regulation, Fungal , Genetic Complementation Test , Hexosyltransferases/chemistry , Hexosyltransferases/genetics , Lipopolysaccharides/isolation & purification , Lipopolysaccharides/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , O Antigens/metabolism , Vaccines, Attenuated
8.
Microsc Microanal ; 16(4): 478-87, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20569528

ABSTRACT

Cellular autofluorescence, though ubiquitous when imaging cells and tissues, is often assumed to be small in comparison to the signal of interest. Uniform estimates of autofluorescence intensity obtained from separate control specimens are commonly employed to correct for autofluorescence. While these may be sufficient for high signal-to-background applications, improvements in detector and probe technologies and introduction of spectral imaging microscopes have increased the sensitivity of fluorescence imaging methods, exposing the possibility of effectively probing the low signal-to-background regime. With spectral imaging, reliable monitoring of signals near or even below the noise levels of the microscope is possible if compensation for autofluorescence and background signals can be performed accurately. We demonstrate the importance of accurate autofluorescence modeling and the utility of spectral imaging and multivariate analysis methods using a case study focusing on fluorescence confocal spectral imaging of host-pathogen interactions. In this application fluorescent proteins are produced when Francisella novicida invade host macrophage cells. The resulting analyte signal is spectrally overlapped and typically weaker than the cellular autofluorescence. In addition to discussing the advantages of spectral imaging for following pathogen invasion, we present the spectral properties and cellular origin of macrophage autofluorescence.


Subject(s)
Francisella tularensis/cytology , Image Processing, Computer-Assisted/methods , Macrophages/microbiology , Microscopy, Confocal/methods , Animals , Cell Line , Mice
9.
Mol Immunol ; 45(12): 3428-37, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18514317

ABSTRACT

Tularemia is a zoonotic disease caused by the Gram-negative intracellular pathogen Francisella tularensis. These bacteria evade phagolysosomal fusion, escape from the phagosome and replicate in the host cell cytoplasm. IFNgamma has been shown to suppress the intra-macrophage growth of Francisella through both nitric oxide-dependent and -independent pathways. Since Francisella is known to subvert host immune responses, we hypothesized that this pathogen could interfere with IFNgamma signaling. Here, we report that infection with Francisella suppresses IFNgamma-induced STAT1 expression and phosphorylation in both human and murine mononuclear phagocytes. This suppressive effect of Francisella is independent of phagosomal escape or replication and is mediated by a heat-stable and constitutively expressed bacterial factor. An analysis of the molecular mechanism of STAT1 inhibition indicated that expression of SOCS3, an established negative regulator of IFNgamma signaling, is highly up-regulated during infection and suppresses STAT1 phosphorylation. Functional analyses revealed that this interference with IFNgamma signaling is accompanied by the suppression of IP-10 production and iNOS induction resulting in increased intracellular bacterial survival. Importantly, the suppressive effect on IFNgamma-mediated host cell protection is most effective when IFNgamma is added post infection, suggesting that the bacteria establish a permissive environment within the host cell.


Subject(s)
Francisella tularensis/cytology , Interferon-gamma/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/microbiology , Microbial Viability/immunology , Phagocytes/immunology , Phagocytes/microbiology , Animals , Cell Line , Cell Proliferation/drug effects , Francisella tularensis/drug effects , Humans , Interferon-gamma/pharmacology , Leukocytes, Mononuclear/drug effects , Macrophages/drug effects , Macrophages/enzymology , Macrophages/microbiology , Mice , Microbial Viability/drug effects , Models, Immunological , Monocytes/drug effects , Monocytes/immunology , Monocytes/microbiology , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/metabolism , Phagocytes/drug effects , Phagosomes/drug effects , Phagosomes/microbiology , Phosphorylation/drug effects , STAT1 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , Tularemia/immunology , Tularemia/microbiology
10.
J Leukoc Biol ; 84(1): 77-85, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18430786

ABSTRACT

Francisella tularensis (Ft) is resistant to serum but requires complement factor C3-derived opsonins for uptake by phagocytic cells and subsequent intracellular growth. In this study, we show that C3 fragments, deposited on Ft, are detected by anti-C3d and -iC3b mAb and that the classical and the alternative pathways are involved in this event. This was demonstrated using C2-depleted sera and specific inhibitors of the classical-versus-alternative pathways of complement activation. Further, we demonstrate that factor C4b, which is crucial for the classical pathway, is deposited on the surface of Ft. In contrast, the C5b-C9 membrane attack complex (MAC) is not assembled on the surface of Ft, which may explain its resistance to complement killing. Deposition of C3 opsonins leads to enhanced phagocytosis by human immature dendritic cells (DC), which leads to intracellular survival, growth, and DC death. Finally, we show that factor H (fH) can bind to the surface of Ft. We believe our data suggest that important virulence factors for Ft are its ability to bind fH and inactivate C3b to iC3b, which culminates in opsonin-induced uptake for subsequent intracellular growth. C3b inactivation also leads to inefficient MAC assembly, which contributes to the ability of this bacterium to resist complement lysis.


Subject(s)
Complement Activation/immunology , Francisella tularensis/cytology , Francisella tularensis/immunology , Opsonin Proteins/immunology , Phagocytosis/immunology , Serum/microbiology , Cell Membrane/metabolism , Complement C3b/immunology , Complement Factor H/immunology , Complement Membrane Attack Complex/metabolism , Complement Pathway, Alternative/immunology , Complement Pathway, Classical/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Microbial Viability
11.
Article in Russian | MEDLINE | ID: mdl-8701638

ABSTRACT

In this investigation isogenic avirulent variants obtained from F. tularensis standard virulent strain 503 were used. The capsule-deficient variants (cap-) were shown to have no species-specific capsular antigens and to be capable of producing R-LPS having no the polysaccharide part of the molecules. The capsule-defective forms (cap +/- ) were found to synthesize capsular antigens and S-LPS whose polysaccharide part essentially differed from the O-lateral chains of LPS of the virulent strain. The study of bacterial dissemination revealed that virulent bacteria rapidly spread in the macroorganism, and their subsequent proliferation shortly led to the death of animals. Avirulent mutants (cap- and cap +/- ) appeared in the organs of animals later and proliferated slower, parasitizing in the macroorganism without fatal outcome. The cap- variants were not capable of inducing the synthesis of antitularemic antibodies and possessed no protective properties. The cap +/- mutants were capable of inducing the synthesis of antitularemic antibodies in mice. These antibodies facilitated the elimination of avirulent strains from the macroorganism, but did not ensure protection from infection with virulent strains.


Subject(s)
Francisella tularensis/pathogenicity , Tularemia/microbiology , Animals , Antibodies, Bacterial/blood , Antibody Specificity , Antigens, Bacterial/analysis , Cell Division , Francisella tularensis/cytology , Francisella tularensis/immunology , Lipopolysaccharides/analysis , Mice , Mutation , Time Factors , Tularemia/immunology , Virulence
12.
Vaccine ; 12(9): 773-5, 1994 Jul.
Article in English | MEDLINE | ID: mdl-7975855

ABSTRACT

Francisella tularensis live vaccine strain (LVS), taken directly from lyophilized vials, did not appear to have a capsule and had low virulence for Balb/c mice. When this strain was subcultured on Chamberlain's synthetic medium (pH 6.5), it became extensively capsulated (1-5 micron in diameter) and its virulence for Balb/c mice increased about 1000-fold. We conclude that the virulence of the attenuated vaccine strain, F. tularensis LVS, may still be influenced by growth conditions.


Subject(s)
Bacterial Vaccines/immunology , Francisella tularensis/immunology , Amino Acids/chemistry , Animals , Bacterial Capsules/immunology , Bacterial Capsules/ultrastructure , Buffers , Culture Media , Francisella tularensis/cytology , Francisella tularensis/pathogenicity , Freeze Drying , Hydrogen-Ion Concentration , Mice , Mice, Inbred BALB C , Microscopy, Electron , Tularemia/prevention & control , Vaccines, Attenuated/immunology , Virulence/immunology
13.
Arch Immunol Ther Exp (Warsz) ; 40(3-4): 183-90, 1992.
Article in English | MEDLINE | ID: mdl-1300982

ABSTRACT

Primary F. tularensis infection in mice induces the production of macrophage activating factors (MAFs) by spleen cells. The stimulation of macrophage cytolytic activity (MAF-c) and hydrogen peroxide production (MAF-H2O2) dominates between days 7 and 10 in the course of tularemia. Three various pools of active fractions (10-11, 14-15, 25-28) were fractionated by two-step chromatography. Typical for 10-11 and 14-15 is MAF-c activity whereas in 25-28 prevails MAF-H2O2. Initial concentrated supernatant (day 7 of infection) and individual fractions have been used to raise antibodies KI (anti 10-11) and KII (anti 14-15). Neutralization reactions with specific antibodies indicate the presence of tumor necrosis factor alpha (TNF alpha) in 14-15 (44% inhibitable), interferon gamma (IFN gamma) and interleukin 2 (IL 2) in 25-28 (65% and 30% neutralization, respectively). Utilizing KI and KII, 99% and 90% inhibition of cytolytic activity is reached in 10-11 and 14-15, respectively, in spite of non-specific cross reaction. Western blot analysis of proteins in supernatant on day 7 detects, besides TNF alpha, further protein bands (13, 15.5, 52 and 72 kDa) that seem to be associated with macrophage activation. Significant protective effect against in vivo multiplication of tularemic microbes indicates a certain role of TNF alpha, however, cooperation of other molecules is worth to be taken into consideration.


Subject(s)
Macrophage-Activating Factors/biosynthesis , Tularemia/immunology , Animals , Cell Division , Female , Francisella tularensis/cytology , Francisella tularensis/immunology , Hydrogen Peroxide/metabolism , Immunity, Cellular , Interferon-gamma/biosynthesis , Interleukin-2/biosynthesis , Macrophage Activation/immunology , Mice , Mice, Inbred C3H , Tularemia/metabolism , Tularemia/microbiology , Tumor Necrosis Factor-alpha/biosynthesis
14.
Article in Russian | MEDLINE | ID: mdl-1455960

ABSTRACT

Instrumental methods of investigation were used for the demonstration of changes in the fatty acid composition of F. tularensis, strain 15 Gaiskii, during cultivation in solid culture medium, storage after lyophilization, as well as changes in the functioning of the system of membrane-dependent enzymes of the respiratory chain and in the permeability of cell wall membranes by water molecules and NADH after lyophilization. A relationship between the survival rate of F. tularensis cells after lyophilization and stimulation of their endogenic respiration with NADH and succinate was revealed. An increase in residual moisture from 6 to 10-12% was found to intensify the process of lipid peroxidation during the storage of lyophilized F. tularensis cells of strain 15 Gaiskii.


Subject(s)
Francisella tularensis/cytology , Bacterial Vaccines , Bacteriological Techniques , Cell Membrane/physiology , Cell Membrane/ultrastructure , Cell Membrane Permeability/drug effects , Culture Media , Francisella tularensis/immunology , Francisella tularensis/physiology , Freeze Drying/instrumentation , Freeze Drying/methods , NAD/pharmacokinetics , Time Factors , Water/metabolism
15.
Antibiot Khimioter ; 35(8): 25-8, 1990 Aug.
Article in Russian | MEDLINE | ID: mdl-2264748

ABSTRACT

Altered viable forms of F. tularensis with spheroplast specific damages of the surface structures were isolated after the culture exposure to lithium chloride (0.5 and 1%). Study of natural penicillin resistance in the spheroplasts and bacterial forms of F. tularensis revealed their difference: the spheroplasts of the strains tested had a lower resistance to beta-lactam antibiotics than the bacterial forms while the activity of spheroplast beta-lactamase did not differ from that of the enzyme of the bacterial form and equalled 224 to 252 U/ml of the cell suspension. Therefore, on the model of the lithium-induced spheroplasts it appeared possible to show that the damages of the surface structures of the cell walls of F. tularensis changed the penicillin resistance level which was indicative of involvement of the F. tularensis cell walls in the phenomenon of the natural resistance to beta-lactams.


Subject(s)
Francisella tularensis/drug effects , Penicillin Resistance , Penicillins/pharmacology , Spheroplasts/drug effects , Cell Membrane/drug effects , Cell Membrane/ultrastructure , Cell Wall/drug effects , Cell Wall/ultrastructure , Culture Media , Drug Resistance, Microbial , Francisella tularensis/cytology , In Vitro Techniques , Microscopy, Electron , Spheroplasts/ultrastructure
19.
Appl Microbiol ; 15(5): 1165-71, 1967 Sep.
Article in English | MEDLINE | ID: mdl-4169543

ABSTRACT

Fluorescein isothiocyanate-labeled beta-glucosidase was used as a simple staining reagent with selected gram-positive and gram-negative organisms. Staining in situ appeared to be dependent on the presence of accessible glycosidic-type linkages in the bacterial cell wall. Extensive wall damage or lysis did not occur when stained cells were suspended in washing and mounting solutions. The apparent specificity of labeled enzyme for wall substance was tested by blocking reactions, staining of isolated cell walls, and failure to stain substances lacking appropriate glycosidic linkages. Severe cell wall lesions were produced after prolonged contact with labeled enzyme, and this phenomenon may also be related to staining specificity. Gram-negative organisms and spores were poorly stained unless protected glycopeptide substrate was previously exposed by treatment of cells with thioglycolic acid or dilute alkaline sodium hypochlorite solution. A potential for staining tissues and cell lines may also exist. Some possible applications of labeled enzymes are briefly discussed.


Subject(s)
Bacteria/cytology , Fluoresceins , Glucosidases , Staining and Labeling , Bacillus anthracis/cytology , Bacillus megaterium/cytology , Bacillus subtilis/cytology , Brucella abortus/cytology , Escherichia coli/cytology , Francisella tularensis/cytology , Listeria monocytogenes/cytology , Microscopy, Electron , Salmonella typhi/cytology , Sarcina/cytology , Staphylococcus/cytology , Vibrio/cytology , Yersinia pestis/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...