Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Antibiotics (Basel) ; 9(4)2020 Apr 01.
Article in English | MEDLINE | ID: mdl-32244733

ABSTRACT

In the beginning of the 21st century, the frequency of antimicrobial resistance (AMR) has reached an apex, where even 4th and 5th generation antibiotics are becoming useless in clinical settings. In turn, patients are suffering from once-curable infections, with increases in morbidity and mortality. The root cause of many of these infections are the ESKAPEE pathogens (Enterococcus species, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacter species, and Escherichia coli), which thrive in the nosocomial environment and are the bacterial species that have seen the largest rise in the acquisition of antibiotic resistance genes. While traditional small-molecule development still dominates the antibacterial landscape for solutions to AMR, some researchers are now turning to biological approaches as potential game changers. Monoclonal antibodies (mAbs)-more specifically, human monoclonal antibodies (Hu-mAbs)-have been highly pursued in the anti-cancer, autoimmune, and antiviral fields with many success stories, but antibody development for bacterial infection is still just scratching the surface. The untapped potential for Hu-mAbs to be used as a prophylactic or therapeutic treatment for bacterial infection is exciting, as these biologics do not have the same toxicity hurdles of small molecules, could have less resistance as they often target virulence proteins rather than proteins required for survival, and are narrow spectrum (targeting just one pathogenic species), therefore avoiding the disruption of the microbiome. This mini-review will highlight the current antibacterial mAbs approved for patient use, the success stories for mAb development, and new Hu-mAb products in the antibacterial pipeline.

2.
PLoS One ; 6(4): e19003, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21544194

ABSTRACT

BACKGROUND: Francisella tularensis is a category-A select agent and is responsible for tularemia in humans and animals. The surface components of F. tularensis that contribute to virulence are not well characterized. An electron-dense capsule has been postulated to be present around F. tularensis based primarily on electron microscopy, but this specific antigen has not been isolated or characterized. METHODS AND FINDINGS: A capsule-like complex (CLC) was effectively extracted from the cell surface of an F. tularensis live vaccine strain (LVS) lacking O-antigen with 0.5% phenol after 10 passages in defined medium broth and growth on defined medium agar for 5 days at 32°C in 7% CO2. The large molecular size CLC was extracted by enzyme digestion, ethanol precipitation, and ultracentrifugation, and consisted of glucose, galactose, mannose, and Proteinase K-resistant protein. Quantitative reverse transcriptase PCR showed that expression of genes in a putative polysaccharide locus in the LVS genome (FTL_1432 through FTL_1421) was upregulated when CLC expression was enhanced. Open reading frames FTL_1423 and FLT_1422, which have homology to genes encoding for glycosyl transferases, were deleted by allelic exchange, and the resulting mutant after passage in broth (LVSΔ1423/1422_P10) lacked most or all of the CLC, as determined by electron microscopy, and CLC isolation and analysis. Complementation of LVSΔ1423/1422 and subsequent passage in broth restored CLC expression. LVSΔ1423/1422_P10 was attenuated in BALB/c mice inoculated intranasally (IN) and intraperitoneally with greater than 80 times and 270 times the LVS LD50, respectively. Following immunization, mice challenged IN with over 700 times the LD50 of LVS remained healthy and asymptomatic. CONCLUSIONS: Our results indicated that the CLC may be a glycoprotein, FTL_1422 and -FTL_1423 were involved in CLC biosynthesis, the CLC contributed to the virulence of F. tularensis LVS, and a CLC-deficient mutant of LVS can protect mice against challenge with the parent strain.


Subject(s)
Francisella tularensis/chemistry , Francisella tularensis/pathogenicity , Glycoproteins/genetics , Glycoproteins/isolation & purification , Tularemia/microbiology , Virulence Factors/genetics , Virulence Factors/isolation & purification , Animals , Francisella tularensis/ultrastructure , Gas Chromatography-Mass Spectrometry , Glycoproteins/physiology , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Microscopy, Electron , Polysaccharides, Bacterial/chemistry , Reverse Transcriptase Polymerase Chain Reaction , Virulence Factors/physiology
3.
PLoS One ; 5(7): e11060, 2010 Jul 06.
Article in English | MEDLINE | ID: mdl-20625403

ABSTRACT

Capsular polysaccharides are important factors in bacterial pathogenesis and have been the target of a number of successful vaccines. Francisella tularensis has been considered to express a capsular antigen but none has been isolated or characterized. We have developed a monoclonal antibody, 11B7, which recognizes the capsular polysaccharide of F. tularensis migrating on Western blot as a diffuse band between 100 kDa and 250 kDa. The capsule stains poorly on SDS-PAGE with silver stain but can be visualized using ProQ Emerald glycoprotein stain. The capsule appears to be highly conserved among strains of F. tularensis as antibody 11B7 bound to the capsule of 14 of 14 F. tularensis type A and B strains on Western blot. The capsular material can be isolated essentially free of LPS, is phenol and proteinase K resistant, ethanol precipitable and does not dissociate in sodium dodecyl sulfate. Immunoelectron microscopy with colloidal gold demonstrates 11B7 circumferentially staining the surface of F. tularensis which is typical of a polysaccharide capsule. Mass spectrometry, compositional analysis and NMR indicate that the capsule is composed of a polymer of the tetrasaccharide repeat, 4)-alpha-D-GalNAcAN-(1->4)-alpha-D-GalNAcAN-(1->3)-beta-D-QuiNAc-(1->2)-beta-D-Qui4NFm-(1-, which is identical to the previously described F. tularensis O-antigen subunit. This indicates that the F. tularensis capsule can be classified as an O-antigen capsular polysaccharide. Our studies indicate that F. tularensis O-antigen glycosyltransferase mutants do not make a capsule. An F. tularensis acyltransferase and an O-antigen polymerase mutant had no evidence of an O-antigen but expressed a capsular antigen. Passive immunization of BALB/c mice with 75 microg of 11B7 protected against a 150 fold lethal challenge of F. tularensis LVS. Active immunization of BALB/c mice with 10 microg of capsule showed a similar level of protection. These studies demonstrate that F. tularensis produces an O-antigen capsule that may be the basis of a future vaccine.


Subject(s)
Bacterial Capsules/immunology , Bacterial Capsules/metabolism , Francisella tularensis/immunology , Francisella tularensis/metabolism , O Antigens/immunology , O Antigens/metabolism , Animals , Antibodies, Monoclonal/immunology , Bacterial Capsules/ultrastructure , Blotting, Western , Cryoelectron Microscopy , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Francisella tularensis/ultrastructure , Gas Chromatography-Mass Spectrometry , Hexosyltransferases/genetics , Hexosyltransferases/metabolism , Magnetic Resonance Spectroscopy , Mass Spectrometry , Mice , Microscopy, Immunoelectron , O Antigens/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
4.
Appl Environ Microbiol ; 74(9): 2637-45, 2008 May.
Article in English | MEDLINE | ID: mdl-18344342

ABSTRACT

Francisella tularensis is the etiologic agent of an intracellular systemic infection of the lymphatic system in humans called tularemia. The organism has become the subject of considerable research interest due to its classification as a category A select agent by the CDC. To aid genetic analysis of this pathogen, we have constructed a temperature-sensitive Tn5-based transposon delivery system that is capable of generating chromosomal reporter fusions with lacZ or luxCDABE, enabling us to monitor gene expression. Transposition is catalyzed by the hyperactive Tn5 transposase, whose expression is driven by the Francisella groES promoter. When high-temperature selection (42 degrees C) is applied to a bacterial culture carrying the transposon delivery plasmid, approximately 0.1% of the population is recovered with Tn5 insertions in the chromosome. Nucleotide sequence analysis of a sample of mutants revealed that the insertions occur randomly throughout the chromosome. The kanamycin-selectable marker of the transposon is also flanked by FLP recombination target sequences that allow deletion of the antibiotic resistance gene when desired. This system has been used to generate transposon mutant libraries for the F. tularensis live vaccine strain as well as two different virulent F. tularensis strains. Chromosomal reporters delivered with the transposon were used to identify genes upregulated by growth in Chamberlain's defined medium. Genes in the fsl operon, reported to be involved in iron acquisition, as well as genes in the igl gene cluster were among those identified by the screen. Further experiments implicate the ferric uptake regulator (Fur) protein in the negative regulation of fsl but not igl reporters, which occurs in an iron-dependent manner. Our results indicate that we have created a valuable new transposon that can be used to identify and characterize virulence genes in F. tularensis strains.


Subject(s)
DNA Transposable Elements , Francisella tularensis/genetics , Mutagenesis, Insertional/methods , Artificial Gene Fusion/methods , Gene Expression Profiling , Genes, Bacterial , Hot Temperature , Kanamycin Resistance/genetics , Luciferases/genetics , Recombination, Genetic , Up-Regulation , beta-Galactosidase/genetics
5.
Infect Immun ; 75(11): 5518-31, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17724076

ABSTRACT

Lipopolysaccharide (LPS) is a major component of the outer membrane of gram-negative bacteria, and the lipid A region of LPS mediates stimulation of the immune system in a structure-dependent manner. Unlike the LPS of many other gram-negative bacteria, the LPS of Francisella tularensis isolated from in vitro cultures is not proinflammatory. This observed lack of proinflammatory prowess may reflect structural features of the lipid A, such as the number and length of the acyl chains and the single-phosphate group. To better understand this phenotype, we have begun to elucidate LPS biosynthesis in F. tularensis. We present complementation, mutational, and chemical data demonstrating that F. tularensis FTT0232c encodes a functional late acyltransferase enzyme with specificity similar to that of the Escherichia coli LpxL ortholog. Expression of this late acyltransferase complemented the temperature-sensitive and hypoacylated lipid A phenotypes of an E. coli lpxL mutant, expression of FTT0232c is increased during intracellular growth relative to that during in vitro growth, and finally, LPS obtained from a mutant of F. tularensis lacking FTT0232c showed an abundant triacyl lipid A species after mass spectrometric analysis, consistent with the loss of an LpxL late acyltransferase.


Subject(s)
Acyltransferases/genetics , Acyltransferases/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Francisella tularensis/enzymology , Escherichia coli/genetics , Escherichia coli/physiology , Francisella tularensis/chemistry , Francisella tularensis/genetics , Gene Deletion , Gene Expression Regulation, Bacterial , Genetic Complementation Test , Lipid A/chemistry , Mass Spectrometry
6.
J Leukoc Biol ; 82(4): 813-20, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17634278

ABSTRACT

Francisella tularensis, a designated Category A biological agent, can cause severe infection in humans. Previous studies have demonstrated a significant immunoprotective role for B lymphocytes in animal models, but the responses of human B lymphocytes to F. tularensis components are largely unknown. The LPS of F. tularensis is atypical and has been reported to lack biological activity on myeloid cells and mouse B cells. Our study characterized the immunological effects of highly purified LPS from different stains of F. tularensis on human B lymphocytes and compared these effects with those on mouse B cells and human monocyte-derived macrophages. Results indicate that marked differences exist between cell type and species in specific responses to this interesting bacterial component. In sharp contrast to responses of mouse splenic B cells or human macrophages, human peripheral B cells showed reproducibly elevated IL-6, TNF-alpha, and antibody production in response to F. tularensis LPS. Data also indicated that these activated human B lymphocytes may subsequently promote the activation of other immune cell types by direct cell-cell interaction. Further investigation into the potential usefulness of F. tularensis LPS as an adjuvant component of a more optimal subunit vaccine is warranted, as it is now clear that it is not biologically inactive, as assumed previously.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antibody Formation/drug effects , B-Lymphocytes/immunology , Francisella tularensis , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Adjuvants, Immunologic/chemistry , Animals , B-Lymphocytes/cytology , Biological Warfare , Cell Line , Francisella tularensis/chemistry , Humans , Interleukin-6/immunology , Lipopolysaccharides/chemistry , Lymphocyte Activation/immunology , Macrophages/cytology , Macrophages/immunology , Mice , Models, Immunological , Species Specificity , Tumor Necrosis Factor-alpha/immunology , Vaccines, Subunit/chemistry , Vaccines, Subunit/immunology
7.
Infect Immun ; 75(6): 3178-82, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17339345

ABSTRACT

In observing Francisella tularensis interactions with nonphagocytic cell lines in vitro, we noted significant adherence, invasion, and intracellular growth of the bacteria within these cells. F. tularensis live vaccine strain invasion of nonprofessional phagocytic cells is inhibited by cytochalasin D and nocodazole, suggesting that both the actin and microtubule cytoskeletons are important for invasion.


Subject(s)
Bacterial Adhesion/physiology , Bacterial Vaccines/immunology , Endothelial Cells/microbiology , Francisella tularensis/metabolism , Bacterial Vaccines/administration & dosage , Cell Line/microbiology , Cytochalasin D/pharmacology , Endothelial Cells/metabolism , Francisella tularensis/immunology , Humans , Models, Biological , Phagocytosis , Tularemia/prevention & control
8.
Anal Chem ; 79(3): 1034-42, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17263332

ABSTRACT

Lipopolysaccharide (LPS) is a major component of the outer membrane of Gram-negative bacteria. The lipid A region of LPS stimulates the immune system in a structure-dependent manner. We have previously identified the two major lipid A species from Francisella tularensis as asymmetric tetraacylated structures containing four long acyl chains (16 and 18 carbons) and a single phosphate group that is partially modified by galactosamine (Phillips, N. J.; Schilling B.; McLendon, M. K.; Apicella, M. A.; Gibson, B. W. Infect. Immun. 2004, 72, 5340-5348). In the current study, we used matrix-assisted laser desorption/ionization on an intermediate vacuum source (vMALDI) coupled to a linear ion trap (LIT) mass spectrometer in multiple-stage mass fragmentation mode (MSn) to determine the structures of several minor and low abundant lipid A species present in F. tularensis, Francisella novicida, and Francisella philomiragia LPS that have not been previously characterized. Comprehensive vMALDI-MSn fragmentation studies allowed us to deduce the composition and the position of the fatty acid substituents within the lipid A moieties. Unexpectedly, most of these minor lipid A species consisted of multiple isobaric species with acyl chains of various lengths. Moreover, we found that a small portion of these lipid A species may be modified by the addition of a hexose or hexosamine sugar, in addition to the galactosamine that was previously identified. Overall, we found that MSn analysis on the vMALDI-LIT-MS platform was highly efficient and sensitive, allowing for thorough analysis of very minor lipid A species.


Subject(s)
Francisella/chemistry , Lipid A/chemistry , Acylation , Francisella tularensis/chemistry , Hexosamines , Hexoses , Lipopolysaccharides , Molecular Structure , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Vacuum
9.
Annu Rev Microbiol ; 60: 167-85, 2006.
Article in English | MEDLINE | ID: mdl-16704343

ABSTRACT

Tularemia is a zoonosis of humans caused by infection with the facultative intracellular bacterium Francisella tularensis. Interest in F. tularensis has increased markedly in the past few years because of its potential use as an agent of bioterrorism. Five subspecies of this organism are found in the Northern hemisphere, but only F. tularensis subsp. tularensis and subsp. holarctica cause disease in humans. This review summarizes what is known about the pathogenesis of tularemia with a focus on bacterial surface components such as lipopolysaccharide and capsule as well as information obtained from the F. tularensis subsp. tularensis SCHU S4 genome. In particular, the mechanisms of action of recently identified virulence factors are discussed in the context of bacterial replication in macrophages and manipulation of the host inflammatory response. Throughout this report, shared and unique features of F. tularensis subsp. tularensis, subsp. holarctica, and subsp. novicida are discussed.


Subject(s)
Biological Warfare , Francisella tularensis/pathogenicity , Animals , Bacterial Capsules/chemistry , Cytokines/biosynthesis , Francisella tularensis/classification , Francisella tularensis/genetics , Francisella tularensis/immunology , Genome, Bacterial , Humans , Lipopolysaccharides/chemistry , Neutrophils/immunology , Phagocytosis
10.
Infect Immun ; 72(9): 5340-8, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15322031

ABSTRACT

We have investigated the lipid A of Francisella tularensis subsp. holarctica strain 1547-57, a type B strain, by using matrix-assisted laser desorption ionization-time-of-flight mass spectrometry, nanoelectrospray quadrupole ion-trap mass spectrometry, and chemical methods. In accordance with the previously published structures of the lipid A from F. tularensis live vaccine strain (LVS) (ATCC 29684) (E. Vinogradov et al., Eur. J. Biochem. 269:6112-6118, 2002), all of the major lipid A forms from strain 1547-57 were tetraacylated. As in the LVS strain, the major fatty acids detected in the F. tularensis 1547-57 lipid A sample included 3-hydroxyoctadecanoic acid, 3-hydroxyhexadecanoic acid, hexadecanoic acid, and tetradecanoic acid. However, several of the lipid A components present in strain 1547-57 were of higher molecular weight than the previously published structures. A major component with an M(r) of 1,666 was found to contain three C(18:0)(3-OH) fatty acids, one C(16:0) fatty acid, one phosphate group, and one 161-Da moiety. This 161-Da moiety could be removed from the lipid A by treatment with aqueous hydrofluoric acid and was identified as galactosamine following peracetylation and analysis by gas chromatography-mass spectrometry. Detailed investigations of the M(r)-1,666 species by ion-trap mass spectrometry with multiple stages of fragmentation suggested that the galactosamine-1-phosphate was linked to the reducing terminus of the lipid A. Similar to the modification of lipid A with arabinosamine, lipopolysaccharide species from F. tularensis containing a phosphate-linked galactosamine could potentially influence its intracellular survival by conferring resistance to antimicrobial peptides.


Subject(s)
Francisella tularensis/chemistry , Lipid A/analysis , Gas Chromatography-Mass Spectrometry , Humans , Lipid A/chemistry , Lipid A/isolation & purification , Mass Spectrometry/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
11.
BMC Infect Dis ; 3: 15, 2003 Jul 11.
Article in English | MEDLINE | ID: mdl-12857350

ABSTRACT

BACKGROUND: The differential expression of virulence genes is often used by microbial pathogens in adapting to the environment of their host. The differential expression of such sets of genes can be regulated by RNA polymerase sigma factors. Some sigma factors are differentially expressed, which can provide a means to identifying other differentially expressed genes such as those whose expression are controlled by the sigma factor. METHODS: To identify sigma factor-regulated genes, we developed a method, termed I-TRAP, for the identification of transcriptional regulator activated promoters. The I-TRAP method is based on the fact that some genes will be differentially expressed in the presence and absence of a transcriptional regulator. I-TRAP uses a DNA library in a promoter-trap vector that contains two reporter genes, one to allow the selection of active promoters in the presence of the transcriptional regulator and a second to allow screening for promoter activity in the absence of the transcriptional regulator. RESULTS: To illustrate the development and use of the I-TRAP approach, the construction of the vectors, host strains, and library necessary to identify SigmaE-regulated genes of Mycobacterium tuberculosis is described. CONCLUSION: The I-TRAP method should be a versatile and useful method for identifying and characterizing promoter activity under a variety of conditions and in response to various regulatory proteins. In our study, we isolated 360 clones that may contain plasmids carrying SigmaE-regulated promoters genes of M. tuberculosis.


Subject(s)
Molecular Biology/methods , Mycobacterium tuberculosis/genetics , Promoter Regions, Genetic , Sigma Factor/physiology , Transcription Factors/physiology , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...