51
|
Asare R, Kwaik YA. Exploitation of host cell biology and evasion of immunity by francisella tularensis. Front Microbiol 2011; 1:145. [PMID: 21687747 PMCID: PMC3109322 DOI: 10.3389/fmicb.2010.00145] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Accepted: 12/21/2010] [Indexed: 12/13/2022] Open
Abstract
Francisella tularensis is an intracellular bacterium that infects humans and many small mammals. During infection, F. tularensis replicates predominantly in macrophages but also proliferate in other cell types. Entry into host cells is mediate by various receptors. Complement-opsonized F. tularensis enters into macrophages by looping phagocytosis. Uptake is mediated in part by Syk, which may activate actin rearrangement in the phagocytic cup resulting in the engulfment of F. tularensis in a lipid raft rich phagosome. Inside the host cells, F. tularensis resides transiently in an acidified late endosome-like compartment before disruption of the phagosomal membrane and escape into the cytosol, where bacterial proliferation occurs. Modulation of phagosome biogenesis and escape into the cytosol is mediated by the Francisella pathogenicity island-encoded type VI-like secretion system. Whilst inside the phagosome, F. tularensis temporarily induce proinflammatory cytokines in PI3K/Akt-dependent manner, which is counteracted by the induction of SHIP that negatively regulates PI3K/Akt activation and promotes bacterial escape into the cytosol. Interestingly, F. tularensis subverts CD4 T cells-mediated killing by inhibiting antigen presentation by activated macrophages through ubiquitin-dependent degradation of MHC II molecules on activated macrophages. In the cytosol, F. tularensis is recognized by the host cell inflammasome, which is down-regulated by F. tularensis that also inhibits caspase-1 and ASC activity. During late stages of intracellular proliferation, caspase-3 is activated but apoptosis is delayed through activation of NF-κB and Ras, which ensures cell viability.
Collapse
Affiliation(s)
- Rexford Asare
- Department of Microbiology and Immunology, School of Medicine, University of Louisville Louisville, KY, USA
| | | |
Collapse
|
52
|
Scientific Opinion on Geographic Distribution of Tick-borne Infections and their Vectors in Europe and the other Regions of the Mediterranean Basin. EFSA J 2010. [DOI: 10.2903/j.efsa.2010.1723] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
53
|
Asare R, Kwaik YA. Molecular complexity orchestrates modulation of phagosome biogenesis and escape to the cytosol of macrophages by Francisella tularensis. Environ Microbiol 2010; 12:2559-86. [PMID: 20482590 PMCID: PMC2957515 DOI: 10.1111/j.1462-2920.2010.02229.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Upon entry of Francisella tularensis to macrophages, the Francisella-containing phagosome (FCP) is trafficked into an acidified late endosome-like phagosome with limited fusion to the lysosomes followed by rapid escape into the cytosol where the organism replicates. Although the Francisella Pathogenicity Island (FPI), which encodes a type VI-like secretion apparatus, is required for modulation of phagosome biogenesis and escape into the cytosol, the mechanisms involved are not known. To decipher the molecular bases of modulation of biogenesis of the FCP and bacterial escape into the macrophage cytosol, we have screened a comprehensive mutant library of F. tularensis ssp. novicida for their defect in proliferation within human macrophages, followed by characterization of modulation of phagosome biogenesis and bacterial escape into the cytosol. Our data show that at least 202 genes are required for intracellular proliferation within macrophages. Among the 125 most defective mutants in intracellular proliferation, we show that the FCP of at least 91 mutants colocalize persistently with the late endosomal/lysosomal marker LAMP-1 and fail to escape into the cytosol, as determined by fluorescence-based phagosome integrity assays and transmission electron microscopy. At least 34 genes are required for proliferation within the cytosol but do not play a detectable role in modulation of phagosome biogenesis and bacterial escape into the cytosol. Our data indicate a tremendous adaptation and metabolic reprogramming by F. tularensis to adjust to the micro-environmental and nutritional cues within the FCP, and these adjustments play essential roles in modulation of phagosome biogenesis and escape into the cytosol of macrophages as well as proliferation in the cytosol. The plethora of the networks of genes that orchestrate F. tularensis-mediated modulation of phagosome biogenesis, phagosomal escape and bacterial proliferation within the cytosol is novel, complex and involves an unusually large portion of the genome of an intracellular pathogen.
Collapse
Affiliation(s)
- Rexford Asare
- Department of Microbiology and Immunology, University of Louisville College of Medicine, Louisville, KY 40292
| | - Yousef Abu Kwaik
- Department of Microbiology and Immunology, University of Louisville College of Medicine, Louisville, KY 40292
| |
Collapse
|
54
|
Asare R, Akimana C, Jones S, Kwaik YA. Molecular bases of proliferation of Francisella tularensis in arthropod vectors. Environ Microbiol 2010; 12:2587-612. [PMID: 20482589 PMCID: PMC2957557 DOI: 10.1111/j.1462-2920.2010.02230.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Arthropod vectors are important vehicles for transmission of Francisella tularensis between mammals, but very little is known about the F. tularensis-arthropod vector interaction. Drosophila melanogaster has been recently developed as an arthropod vector model for F. tularensis. We have shown that intracellular trafficking of F. tularensis within human monocytes-derived macrophages and D. melanogaster-derived S2 cells is very similar. Within both evolutionarily distant host cells, the Francisella-containing phagosome matures to a late endosome-like phagosome with limited fusion to lysosomes followed by rapid bacterial escape into the cytosol where the bacterial proliferate. To decipher the molecular bases of intracellular proliferation of F. tularensis within arthropod-derived cells, we screened a comprehensive library of mutants of F. tularensis ssp. novicida for their defect in intracellular proliferation within D. melanogaster-derived S2 cells. Our data show that 394 genes, representing 22% of the genome, are required for intracellular proliferation within D. melanogaster-derived S2 cells, including many of the Francisella Pathogenicity Island (FPI) genes that are also required for proliferation within mammalian macrophages. Functional gene classes that exhibit growth defect include metabolic (25%), FPI (2%), type IV pili (1%), transport (16%) and DNA modification (5%). Among 168 most defective mutants in intracellular proliferation in S2 cells, 80 are defective in lethality and proliferation within adult D. melanogaster. The observation that only 135 of the 394 mutants that are defective in S2 cells are also defective in human macrophages indicates that F. tularensis utilize common as well as distinct mechanisms to proliferate within mammalian and arthropod cells. Our studies will facilitate deciphering the molecular aspects of F. tularensis-arthropod vector interaction and its patho-adaptation to infect mammals.
Collapse
Affiliation(s)
| | | | - Snake Jones
- Department of Microbiology and Immunology, University of Louisville College of Medicine, Louisville, KY 40292
| | - Yousef Abu Kwaik
- Department of Microbiology and Immunology, University of Louisville College of Medicine, Louisville, KY 40292
| |
Collapse
|
55
|
Bublitz DC, Noah CE, Benach JL, Furie MB. Francisella tularensis suppresses the proinflammatory response of endothelial cells via the endothelial protein C receptor. THE JOURNAL OF IMMUNOLOGY 2010; 185:1124-31. [PMID: 20543103 DOI: 10.4049/jimmunol.0902429] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Various bacterial pathogens activate the endothelium to secrete proinflammatory cytokines and recruit circulating leukocytes. In contrast, there is a distinct lack of activation of these cells by Francisella tularensis, the causative agent of tularemia. Given the importance of endothelial cells in facilitating innate immunity, we investigated the ability of the attenuated live vaccine strain and virulent Schu S4 strain of F. tularensis to inhibit the proinflammatory response of HUVECs. Living F. tularensis live vaccine strain and Schu S4 did not stimulate secretion of the chemokine CCL2 by HUVECs, whereas material released from heat-killed bacteria did. Furthermore, the living bacteria suppressed secretion in response to heat-killed F. tularensis. This phenomenon was dose and contact dependent, and it occurred rapidly upon infection. The living bacteria did not inhibit the activation of HUVECs by Escherichia coli LPS, highlighting the specificity of this suppression. The endothelial protein C receptor (EPCR) confers anti-inflammatory properties when bound by activated protein C. When the EPCR was blocked, F. tularensis lost the ability to suppress activation of HUVECs. To our knowledge, this is the first report that a bacterial pathogen inhibits the host immune response via the EPCR. Endothelial cells are a critical component of the innate immune response to infection, and suppression of their activation by F. tularensis is likely a mechanism that aids in bacterial dissemination and evasion of host defenses.
Collapse
Affiliation(s)
- DeAnna C Bublitz
- Center for Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | | | | | | |
Collapse
|
56
|
Akimana C, Al-Khodor S, Abu Kwaik Y. Host factors required for modulation of phagosome biogenesis and proliferation of Francisella tularensis within the cytosol. PLoS One 2010; 5:e11025. [PMID: 20552012 PMCID: PMC2883998 DOI: 10.1371/journal.pone.0011025] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Accepted: 05/13/2010] [Indexed: 01/07/2023] Open
Abstract
Francisella tularensis is a highly infectious facultative intracellular bacterium that can be transmitted between mammals by arthropod vectors. Similar to many other intracellular bacteria that replicate within the cytosol, such as Listeria, Shigella, Burkholderia, and Rickettsia, the virulence of F. tularensis depends on its ability to modulate biogenesis of its phagosome and to escape into the host cell cytosol where it proliferates. Recent studies have identified the F. tularensis genes required for modulation of phagosome biogenesis and escape into the host cell cytosol within human and arthropod-derived cells. However, the arthropod and mammalian host factors required for intracellular proliferation of F. tularensis are not known. We have utilized a forward genetic approach employing genome-wide RNAi screen in Drosophila melanogaster-derived cells. Screening a library of approximately 21,300 RNAi, we have identified at least 186 host factors required for intracellular bacterial proliferation. We silenced twelve mammalian homologues by RNAi in HEK293T cells and identified three conserved factors, the PI4 kinase PI4KCA, the ubiquitin hydrolase USP22, and the ubiquitin ligase CDC27, which are also required for replication in human cells. The PI4KCA and USP22 mammalian factors are not required for modulation of phagosome biogenesis or phagosomal escape but are required for proliferation within the cytosol. In contrast, the CDC27 ubiquitin ligase is required for evading lysosomal fusion and for phagosomal escape into the cytosol. Although F. tularensis interacts with the autophagy pathway during late stages of proliferation in mouse macrophages, this does not occur in human cells. Our data suggest that F. tularensis utilizes host ubiquitin turnover in distinct mechanisms during the phagosomal and cytosolic phases and phosphoinositide metabolism is essential for cytosolic proliferation of F. tularensis. Our data will facilitate deciphering molecular ecology, patho-adaptation of F. tularensis to the arthropod vector and its role in bacterial ecology and patho-evolution to infect mammals.
Collapse
Affiliation(s)
- Christine Akimana
- Department of Microbiology and Immunology, College of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Souhaila Al-Khodor
- Department of Microbiology and Immunology, College of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Yousef Abu Kwaik
- Department of Microbiology and Immunology, College of Medicine, University of Louisville, Louisville, Kentucky, United States of America
- Department of Biology, University of Louisville, Louisville, Kentucky, United States of America
- * E-mail:
| |
Collapse
|
57
|
Abstract
Like for all microbes, the goal of every pathogen is to survive and replicate. However, to overcome the formidable defenses of their hosts, pathogens are also endowed with traits commonly associated with virulence, such as surface attachment, cell or tissue invasion, and transmission. Numerous pathogens couple their specific virulence pathways with more general adaptations, like stress resistance, by integrating dedicated regulators with global signaling networks. In particular, many of nature's most dreaded bacteria rely on nucleotide alarmones to cue metabolic disturbances and coordinate survival and virulence programs. Here we discuss how components of the stringent response contribute to the virulence of a wide variety of pathogenic bacteria.
Collapse
Affiliation(s)
- Zachary D. Dalebroux
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sarah L. Svensson
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Erin C. Gaynor
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michele S. Swanson
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
58
|
Regulation of virulence gene transcripts by the Francisella novicida orphan response regulator PmrA: role of phosphorylation and evidence of MglA/SspA interaction. Infect Immun 2010; 78:2189-98. [PMID: 20231408 DOI: 10.1128/iai.00021-10] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Francisella tularensis subsp. tularensis is the etiologic agent of tularemia and has been designated a category A biothreat agent by the CDC. Tularemia is characterized by replication and dissemination within host phagocytes. Intramacrophage growth is dependent upon the regulation of Francisella pathogenicity island (FPI) virulence genes, which is poorly understood. Two-component regulatory systems (TCS) are widely employed by Gram-negative bacteria to monitor and respond to environmental signals. Virulent strains of F. tularensis subsp. tularensis are devoid of classical, tandemly arranged TCS genes, but orphaned members, such as that encoding the response regulator PmrA, have been identified. In the F. novicida model system, previous work has shown that a pmrA mutant shows decreased expression of FPI genes, is deficient for intramacrophage growth, and is avirulent in the mouse model. Here, we determine that phosphorylation aids PmrA binding to regulated promoters pmrA and the FPI-encoded pdpD, and KdpD is the histidine kinase primarily responsible for phosphorylation of PmrA at the aspartic acid at position 51 (D51). A strain expressing PmrA D51A retains some DNA binding but exhibits reduced expression of the PmrA regulon, is deficient for intramacrophage replication, and is attenuated in the mouse model. With regard to virulence gene induction, PmrA coprecipitates with the FPI transcription factors MglA and SspA, which bind RNA polymerase. Together, these data suggest a model of Francisella gene regulation that includes a TCS consisting of KdpD and PmrA. Once phosphorylated, PmrA binds to regulated gene promoters recruiting free or RNA polymerase-bound MglA and SspA to initiate FPI gene transcription.
Collapse
|
59
|
Santic M, Al-Khodor S, Abu Kwaik Y. Cell biology and molecular ecology ofFrancisella tularensis. Cell Microbiol 2010; 12:129-39. [DOI: 10.1111/j.1462-5822.2009.01400.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
60
|
Holická M, Novosad J, Loudová M, Kudlová M, Krejsek J. The effect of interferon-gamma and lipopolysaccharide on the growth of Francisella tularensis LVS in murine macrophage-like cell line J774. ACTA MEDICA (HRADEC KRÁLOVÉ) 2010; 52:101-6. [PMID: 20073421 DOI: 10.14712/18059694.2016.113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Francisella tularensis, a causative agent of human tularemia, displaying the ability to proliferate inside the human cells. AIMS To evaluate the growth potential of F. tularensis LVS strain in macrophage-like cell line J774 modulated by recombinant interferon gamma and E. coli derived lipopolysaccharide. RESULTS Stimulation of J774 cells either by interferon-gamma or lipopolysaccharide alone, or especially in combination before infection F. tularensis, revealed protective effects. Higher concentrations of stimulating agents were needed to inhibit ongoing F. tularensis infection. CONCLUSIONS Stimulation of J774 cell line by combination of interferon-gamma with lipopolysaccharide inhibits the intracellular growth of F. tularensis.
Collapse
Affiliation(s)
- Monika Holická
- Department of Clinical Immunology and Allergology, Charles University in Prague, Faculty of Medicine and University Hospital, Hradec Králové, Czech Republic
| | | | | | | | | |
Collapse
|
61
|
Rajaram MVS, Butchar JP, Parsa KVL, Cremer TJ, Amer A, Schlesinger LS, Tridandapani S. Akt and SHIP modulate Francisella escape from the phagosome and induction of the Fas-mediated death pathway. PLoS One 2009; 4:e7919. [PMID: 19936232 PMCID: PMC2775408 DOI: 10.1371/journal.pone.0007919] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Accepted: 10/27/2009] [Indexed: 12/13/2022] Open
Abstract
Francisella tularensis infects macrophages and escapes phago-lysosomal fusion to replicate within the host cytosol, resulting in host cell apoptosis. Here we show that the Fas-mediated death pathway is activated in infected cells and correlates with escape of the bacterium from the phagosome and the bacterial burden. Our studies also demonstrate that constitutive activation of Akt, or deletion of SHIP, promotes phago-lysosomal fusion and limits bacterial burden in the host cytosol, and the subsequent induction of Fas expression and cell death. Finally, we show that phagosomal escape/intracellular bacterial burden regulate activation of the transcription factors sp1/sp3, leading to Fas expression and cell death. These data identify for the first time host cell signaling pathways that regulate the phagosomal escape of Francisella, leading to the induction of Fas and subsequent host cell death.
Collapse
Affiliation(s)
- Murugesan V. S. Rajaram
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Jonathan P. Butchar
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Kishore V. L. Parsa
- The Ohio State Biochemistry Program, The Ohio State University, Columbus, Ohio, United States of America
| | - Thomas J. Cremer
- Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, Ohio, United States of America
| | - Amal Amer
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
- Center for Microbial Interface Biology, The Ohio State University, Columbus, Ohio, United States of America
| | - Larry S. Schlesinger
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
- Center for Microbial Interface Biology, The Ohio State University, Columbus, Ohio, United States of America
| | - Susheela Tridandapani
- Department of Internal Medicine, The Ohio State University, Columbus, Ohio, United States of America
- The Ohio State Biochemistry Program, The Ohio State University, Columbus, Ohio, United States of America
- Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, Ohio, United States of America
- Center for Microbial Interface Biology, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
62
|
Santic M, Pavokovic G, Jones S, Asare R, Kwaik YA. Regulation of apoptosis and anti-apoptosis signalling by Francisella tularensis. Microbes Infect 2009; 12:126-34. [PMID: 19925880 DOI: 10.1016/j.micinf.2009.11.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Revised: 11/05/2009] [Accepted: 11/05/2009] [Indexed: 02/06/2023]
Abstract
Francisella tularensis induces apoptosis within macrophages but the temporal and spatial modulation through activation of caspase-1, caspase-3, and the anti-apoptosis nuclear transcription factor B (NF-kappaB) is not known. Whether escape of the bacteria into the cytosol is sufficient and/or essential for activation of NF-kappaB is not known. Our results show that F. tularensis subsp. novicida induces sustained nuclear translocation of NF-kappaB at early time points after infection of human monocytes derived macrophages (hMDMs). The sustained nuclear translocation of NF-kappaB is defective in the iglC mutant that fails to escape into the cytosol of macrophages. Nuclear translocation of NF-kappaB by the wild type strain is abolished upon treatment with the NF-kappaB inhibitor caffein acid phenyl ester. While the wild type strain triggers caspase-3 and caspase-1 activation by 6 h post-infection the iglC mutant is defective in triggering both caspases. In hMDMs treated with the apoptosis-inducing agent, staurosporin, there is an induction of cell death in the iglC mutant-infected macrophages despite reduced frequency of caspase-1 and caspase-3 activity. The wt-infected macrophages are resistant to cell death-induced agent. We conclude that although caspase-1 and capsase-3 are triggered within F. tularensis-infected hMDMs during early stages of infection, cell death is delayed, which is correlated with simultaneous activation of NF-kappaB.
Collapse
Affiliation(s)
- Marina Santic
- Department of Microbiology and Parasitology, University of Rijeka, Medical Faculty, Brace Branchetta 20, 51000 Rijeka, Croatia.
| | | | | | | | | |
Collapse
|
63
|
Meibom KL, Barel M, Charbit A. Loops and networks in control of Francisella tularensis virulence. Future Microbiol 2009; 4:713-29. [PMID: 19659427 DOI: 10.2217/fmb.09.37] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Francisella tularensis is a highly infectious, Gram-negative bacterium responsible for the disease tularemia in a broad variety of animals, including humans. F. tularensis intracellular multiplication occurs mainly in macrophages. However, F. tularensis is able to infect many other cell types, including other phagocytic (dendritic cells, polymorphonuclear leukocytes) and nonphagocytic (alveolar epithelial cells, hepatocytes, endothelial cells and fibroblasts) cells. The ability of professional phagocytic cells to engulf and kill microbes is an essential component of innate defense. The ability of F. tularensis to impair phagocyte function and survive in the cytosol of infected cells thus constitutes a central aspect of its virulence. The F. tularensis intracellular lifecycle relies on the tightly regulated expression of a series of genes. The unraveling secrets of the regulatory cascades governing the regulation of virulence of F. tularensis will be discussed along with future challenges yet to be solved.
Collapse
Affiliation(s)
- Karin L Meibom
- INSERM U570, Université Paris Descartes, Faculté de Médecine Necker Enfants-Malades, 75730, Paris Cedex 15, France.
| | | | | |
Collapse
|
64
|
Abstract
Tularemia is a potentially fatal multi-systemic disease of humans and other animals caused by the bacterial pathogen Francisella tularensis. The disease can be transmitted by ticks, biting flies, water exposure, food, and aerosols and occurs around the northern hemisphere including North America, Europe, and Asia. There are several defined species and subspecies, including F. tularensis subsp. tularensis (Jellison Type A) which is pathogenic for rabbits and occurs in North America, F. tularensis subsp. holarctica (Type B) and mediaasiatica which are less pathogenic for rabbits, and F. tularensis subsp. novicida which has been regarded sometimes as the separate species F. novicida. Because it can have a high aerosol-related infection rate, low infectious dose, and ability to induce fatal disease, F. tularensis is considered a potential agent of biological warfare and is classified by the US Department of Health and Human Services as a List A select agent. We discuss microbiological, clinicopathological, epidemiological, and ecological aspects of tularemia.
Collapse
|
65
|
Mahawar M, Kirimanjeswara GS, Metzger DW, Bakshi CS. Contribution of citrulline ureidase to Francisella tularensis strain Schu S4 pathogenesis. J Bacteriol 2009; 191:4798-806. [PMID: 19502406 PMCID: PMC2715709 DOI: 10.1128/jb.00212-09] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2009] [Accepted: 05/21/2009] [Indexed: 12/25/2022] Open
Abstract
The citrulline ureidase (CTU) activity has been shown to be associated with highly virulent Francisella tularensis strains, including Schu S4, while it is absent in avirulent or less virulent strains. A definitive role of the ctu gene in virulence and pathogenesis of F. tularensis Schu S4 has not been assessed; thus, an understanding of the significance of this phenotype is long overdue. CTU is a carbon-nitrogen hydrolase encoded by the citrulline ureidase (ctu) gene (FTT0435) on the F. tularensis Schu S4 genome. In the present study, we evaluated the contribution of the ctu gene in the virulence of category A agent F. tularensis Schu S4 by generating a nonpolar deletion mutant, the Deltactu mutant. The deletion of the ctu gene resulted in loss of CTU activity, which was restored by transcomplementing the ctu gene. The Deltactu mutant did not exhibit any growth defect under acellular growth conditions; however, it was impaired for intramacrophage growth in resting as well as gamma interferon-stimulated macrophages. The Deltactu mutant was further tested for its virulence attributes in a mouse model of respiratory tularemia. Mice infected intranasally with the Deltactu mutant showed significantly reduced bacterial burden in the lungs, liver, and spleen compared to wild-type (WT) Schu S4-infected mice. The reduced bacterial burden in mice infected with the Deltactu mutant was also associated with significantly lower histopathological scores in the lungs. Mice infected with the Deltactu mutant succumbed to infection, but they survived longer and showed significantly extended median time to death compared to that shown by WT Schu S4-infected mice. To conclude, this study demonstrates that ctu contributes to intracellular survival, in vivo growth, and pathogenesis. However, ctu is not an absolute requirement for the virulence of F. tularensis Schu S4 in mice.
Collapse
Affiliation(s)
- Manish Mahawar
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, NY 12208-3479, USA
| | | | | | | |
Collapse
|
66
|
Moro WB, Yang Z, Kane TA, Zhou Q, Harville S, Brouillette CG, Brouillette WJ. SAR studies for a new class of antibacterial NAD biosynthesis inhibitors. JOURNAL OF COMBINATORIAL CHEMISTRY 2009; 11:617-25. [PMID: 19408950 PMCID: PMC2888690 DOI: 10.1021/cc9000357] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
A new lead class of antibacterial drug-like NAD synthetase (NADs) inhibitors was previously identified from a virtual screening study. Here a solution-phase synthetic library of 76 compounds, analogs of the urea-sulfonamide 5838, was synthesized in parallel to explore SAR on the sulfonamide aryl group. All library members were tested for enzyme inhibition against NADs and nicotinic acid mononucleotide adenylyltransferase (NaMNAT), the last two enzymes in the biosynthesis of NAD, and for growth inhibition in a Bacillus anthracis antibacterial assay. Most compounds that inhibited bacterial growth also showed inhibition against one of the enzymes tested. While only modest enhancements in the enzyme inhibition potency against NADs were observed, of significance was the observation that the antibacterial urea-sulfonamides more consistently inhibited NaMNAT.
Collapse
Affiliation(s)
- Whitney Beysselance Moro
- Department of Chemistry, University of Alabama at Birmingham, 901 14 Street South, Birmingham, Alabama 35294, USA
- Center for Biophysical Sciences & Engineering, University of Alabama at Birmingham, 1025 18 Street South, Birmingham, Alabama 35294, USA
| | - Zhengrong Yang
- Department of Chemistry, University of Alabama at Birmingham, 901 14 Street South, Birmingham, Alabama 35294, USA
- Center for Biophysical Sciences & Engineering, University of Alabama at Birmingham, 1025 18 Street South, Birmingham, Alabama 35294, USA
| | - Tasha A. Kane
- Center for Biophysical Sciences & Engineering, University of Alabama at Birmingham, 1025 18 Street South, Birmingham, Alabama 35294, USA
| | - Qingxian Zhou
- Center for Biophysical Sciences & Engineering, University of Alabama at Birmingham, 1025 18 Street South, Birmingham, Alabama 35294, USA
| | - Steve Harville
- Department of Chemistry, University of Alabama at Birmingham, 901 14 Street South, Birmingham, Alabama 35294, USA
| | - Christie G. Brouillette
- Department of Chemistry, University of Alabama at Birmingham, 901 14 Street South, Birmingham, Alabama 35294, USA
- Center for Biophysical Sciences & Engineering, University of Alabama at Birmingham, 1025 18 Street South, Birmingham, Alabama 35294, USA
| | - Wayne J. Brouillette
- Department of Chemistry, University of Alabama at Birmingham, 901 14 Street South, Birmingham, Alabama 35294, USA
- Center for Biophysical Sciences & Engineering, University of Alabama at Birmingham, 1025 18 Street South, Birmingham, Alabama 35294, USA
| |
Collapse
|
67
|
Buchan BW, McCaffrey RL, Lindemann SR, Allen LAH, Jones BD. Identification of migR, a regulatory element of the Francisella tularensis live vaccine strain iglABCD virulence operon required for normal replication and trafficking in macrophages. Infect Immun 2009; 77:2517-29. [PMID: 19349423 PMCID: PMC2687360 DOI: 10.1128/iai.00229-09] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2009] [Revised: 03/23/2009] [Accepted: 03/25/2009] [Indexed: 01/05/2023] Open
Abstract
Francisella tularensis, the etiological agent of tularemia, is capable of infecting a wide range of animals and causes a severe, lethal disease in humans. The pathogen evades killing by cells of the innate immune system utilizing genes encoding a pathogenicity island, including iglABCD, and instead utilizes these cells as a niche for replication and dissemination to other organs within the host. Regulators of the igl genes (e.g., MglA, SspA, FevR and PmrA) have been identified, but environmental stimuli and mechanisms of regulation are as yet unknown and are likely to involve additional gene products. In this work, we more closely examine the roles that environmental iron and the ferric uptake repressor protein (Fur) play in the regulation of the iglABCD operon. We also used a genetic approach to identify and characterize a new regulator of the igl operon, designated migR (macrophage intracellular growth regulator; FTL_1542). Quantitative real-time reverse transcription-PCR in a site-directed migR mutant confirmed the reduction in the number of iglC transcripts in this strain and also demonstrated reduced expression of fevR. Comparison of the migR and fevR mutants in monocyte-derived macrophages (MDMs) and epithelial cell lines revealed a reduced ability for each mutant to grow in MDMs, yet only the fevR mutant exhibited impaired replication in epithelial cell lines. Confocal analysis of infected MDMs revealed that although neither mutant reached the MDM cytosol, the fevR mutant was trapped in lamp-1-positive phagosomes, whereas the migR mutant resided in mature phagolysosomes enriched with both lamp-1 and cathepsin D. Disruption of migR and fevR also impaired the ability of F. tularensis to prevent neutrophil oxidant production. Thus, we have identified migR, a gene that regulates expression of the iglABCD operon and is essential for bacterial growth in MDMs and also contributes to the blockade of neutrophil NADPH oxidase activity.
Collapse
Affiliation(s)
- Blake W Buchan
- Department of Microbiology, Roy J. and Lucille A. Carver School of Medicine, University of Iowa, Iowa City, IA 52242-1109, USA
| | | | | | | | | |
Collapse
|
68
|
Ray K, Marteyn B, Sansonetti PJ, Tang CM. Life on the inside: the intracellular lifestyle of cytosolic bacteria. Nat Rev Microbiol 2009; 7:333-40. [PMID: 19369949 DOI: 10.1038/nrmicro2112] [Citation(s) in RCA: 324] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bacterial pathogens exploit a huge range of niches within their hosts. Many pathogens can invade non-phagocytic cells and survive within a membrane-bound compartment. However, only a small number of bacteria, including Listeria monocytogenes, Shigella flexneri, Burkholderia pseudomallei, Francisella tularensis and Rickettsia spp., can gain access to and proliferate within the host cell cytosol. Here, we discuss the mechanisms by which these cytosolic pathogens escape into the cytosol, obtain nutrients to replicate and subvert host immune responses.
Collapse
Affiliation(s)
- Katrina Ray
- Department of Microbiology, Centre for Molecular Microbiology and Infection, Imperial College London, London, UK
| | | | | | | |
Collapse
|
69
|
Woolard MD, Frelinger JA. Outsmarting the host: bacteria modulating the immune response. Immunol Res 2009; 41:188-202. [PMID: 18592144 DOI: 10.1007/s12026-008-8021-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Pathogenic bacteria and their hosts have had a two-way conversation for millions of years. This interaction has led to many measure/counter-measure responses by the host and bacteria. The host immune response has developed many mechanisms to neutralize and remove pathogen bacteria. In turn pathogenic bacteria have developed mechanisms to alter and evade the host immune response. We will review some of the mechanisms utilized by bacteria to accomplish this goal. We will also examine the current state of understanding of Francisella tularensis mediated immune evasion.
Collapse
Affiliation(s)
- Matthew D Woolard
- Department of Microbiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7290, USA
| | | |
Collapse
|
70
|
Schulert GS, McCaffrey RL, Buchan BW, Lindemann SR, Hollenback C, Jones BD, Allen LAH. Francisella tularensis genes required for inhibition of the neutrophil respiratory burst and intramacrophage growth identified by random transposon mutagenesis of strain LVS. Infect Immun 2009; 77:1324-36. [PMID: 19204089 PMCID: PMC2663180 DOI: 10.1128/iai.01318-08] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2008] [Revised: 12/03/2008] [Accepted: 02/02/2009] [Indexed: 12/29/2022] Open
Abstract
Francisella tularensis is a facultative intracellular pathogen and the causative agent of tularemia. We have shown that F. tularensis subspecies holarctica strain LVS prevents NADPH oxidase assembly and activation in human neutrophils, but how this is achieved is unclear. Herein, we used random transposon mutagenesis to identify LVS genes that affect neutrophil activation. Our initial screen identified carA, carB, and pyrB, which encode the small and large subunits of carbamoylphosphate synthase and aspartate carbamoyl transferase, respectively. These strains are uracil auxotrophs, and their growth was attenuated on cysteine heart agar augmented with sheep blood (CHAB) or in modified Mueller-Hinton broth. Phagocytosis of the uracil auxotrophic mutants triggered a respiratory burst in neutrophils, and ingested bacteria were killed and fragmented in phagosomes that contained superoxide. Conversely, phagocytosis did not trigger a respiratory burst in blood monocytes or monocyte-derived macrophages (MDM), and phagosomes containing wild-type or mutant bacteria lacked NADPH oxidase subunits. Nevertheless, the viability of mutant bacteria declined in MDM, and ultrastructural analysis revealed that phagosome egress was significantly inhibited despite synthesis of the virulence factor IglC. Other aspects of infection, such as interleukin-1beta (IL-1beta) and IL-8 secretion, were unaffected. The cultivation of carA, carB, or pyrB on uracil-supplemented CHAB was sufficient to prevent neutrophil activation and intramacrophage killing and supported escape from MDM phagosomes, but intracellular growth was not restored unless uracil was added to the tissue culture medium. Finally, all mutants tested grew normally in both HepG2 and J774A.1 cells. Collectively, our data demonstrate that uracil auxotrophy has cell type-specific effects on the fate of Francisella bacteria.
Collapse
Affiliation(s)
- Grant S Schulert
- Inflammation Program, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | | | | | | | |
Collapse
|
71
|
Moro WB, Yang Z, Kane T, Brouillette CG, Brouillette WJ. Virtual screening to identify lead inhibitors for bacterial NAD synthetase (NADs). Bioorg Med Chem Lett 2009; 19:2001-5. [PMID: 19249205 PMCID: PMC2666046 DOI: 10.1016/j.bmcl.2009.02.034] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2008] [Revised: 02/05/2009] [Accepted: 02/09/2009] [Indexed: 11/24/2022]
Abstract
Virtual screening was employed to identify new drug-like inhibitors of NAD synthetase (NADs) as antibacterial agents. Four databases of commercially available compounds were docked against three subsites of the NADs active site using FlexX in conjunction with CScore. Over 200 commercial compounds were purchased and evaluated in enzyme inhibition and antibacterial assays. 18 compounds inhibited NADs at or below 100 microM (7.6% hit rate), and two were selected for future SAR studies.
Collapse
Affiliation(s)
- Whitney Beysselance Moro
- Center for Biophysical Sciences and Engineering, University of Alabama at Birmingham, 1025 18th Street South, Birmingham, AL 35294
- Department of Chemistry, University of Alabama at Birmingham, 901 14th Street South, Birmingham, AL 35294
| | - Zhengrong Yang
- Center for Biophysical Sciences and Engineering, University of Alabama at Birmingham, 1025 18th Street South, Birmingham, AL 35294
- Department of Chemistry, University of Alabama at Birmingham, 901 14th Street South, Birmingham, AL 35294
| | - Tasha Kane
- Center for Biophysical Sciences and Engineering, University of Alabama at Birmingham, 1025 18th Street South, Birmingham, AL 35294
| | - Christie G. Brouillette
- Center for Biophysical Sciences and Engineering, University of Alabama at Birmingham, 1025 18th Street South, Birmingham, AL 35294
- Department of Chemistry, University of Alabama at Birmingham, 901 14th Street South, Birmingham, AL 35294
| | - Wayne J. Brouillette
- Center for Biophysical Sciences and Engineering, University of Alabama at Birmingham, 1025 18th Street South, Birmingham, AL 35294
- Department of Chemistry, University of Alabama at Birmingham, 901 14th Street South, Birmingham, AL 35294
| |
Collapse
|
72
|
Abstract
Recognition of bacterial infection is the first key step to the initiation of an inflammatory response and host defense. Transmembrane proteins of the Toll-like receptor family have long been recognized as key detectors of the extracellular presence of pathogens. Recently, much research has identified a variety of intracellular detectors that also mediate innate immune responses following bacterial infection. These cannot only recognize bacteria that invade the cell cytoplasm, but also a variety of bacterial products that are introduced into cells by specialized secretion systems or are secreted toxins. This article will focus on these intracellular detectors and the bacterial components that they recognize. These detectors are particularly well adapted to recognize the presence of pathogenic bacteria as opposed to commensal organisms. Their growing importance suggests that targeting such intracellular pathways may be important in the future for manipulating the immune response to infection as an aid to augmenting host defense and providing more effective vaccines.
Collapse
Affiliation(s)
- Thomas John Evans
- Division of Immunology, Infection & Inflammation, University of Glasgow, 120 University Place, Glasgow, G12 8TA, UK.
| |
Collapse
|
73
|
Santic M, Akimana C, Asare R, Kouokam JC, Atay S, Kwaik YA. Intracellular fate of Francisella tularensis within arthropod-derived cells. Environ Microbiol 2009; 11:1473-81. [PMID: 19220402 DOI: 10.1111/j.1462-2920.2009.01875.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Since transmission of Francisella tularensis into the mammalian host occurs via arthropod vectors such as ticks, mosquitoes, horseflies and deerflies, recent studies have established Drosophila melanogaster as an arthropod vector model system. Nothing is known about the intracellular fate of F. tularensis within arthropod-derived cells, and the role of this host-parasite adaptation in the evolution of this pathogen to infect mammals. In this report, we explored intracellular trafficking of F. tularensis ssp. novicida in D. melanogaster-derived S2 cells. First, we show that similar to the F. tularensis ssp. holarctica-derived LVS strain, F. tularensis ssp. novicida is highly infectious, replicates exponentially within S2 cells and within adult flies, and is fatal to adult fruit flies in a dose-dependent manner, while the iglC, iglD and mglA mutants are defective. Using electron and fluorescence microscopy-based phagosome integrity assays, we show that the wild-type strain escapes into the cytosol of S2 cells within 30-60 min post infection and by 6 h, 90% were cytosolic. In contrast, approximately 40-50% of the iglC and iglD mutants escape into the cytosol by 6 h while the other subpopulation becomes enclosed within multilamellar vesicles (MLVs). Pre-treatment of S2 cells with the autophagy inhibitor methyl adenine blocks formation of the MLVs and all the vacuolar subpopulation of the iglC and iglD mutant bacteria become enclosed within single membrane-surrounded vacuoles. Endocytic trafficking studies of F. tularensis within S2 cells show transient colocalization of the bacterial phagosome with D. melanogaster LAMP2-GFP fusion but not with lysosomes pre-loaded with fluorescent dextran. Our data show that MLVs harbouring the iglC mutant acquire Lamp2 and dextran while MLVs harbouring the iglD mutant exclude these late endosomal and lysosomal markers. Our data indicate crucial differences in the role of the pathogenicity island-encoded proteins in modulating intracellular trafficking within human macrophages and arthropod vector-derived cells.
Collapse
Affiliation(s)
- Marina Santic
- Department of Microbiology and Immunology, University of Louisville, College of Medicine, Louisville, KY 40202, USA
| | | | | | | | | | | |
Collapse
|
74
|
Zhang P, Katz J, Michalek SM. Glycogen synthase kinase-3beta (GSK3beta) inhibition suppresses the inflammatory response to Francisella infection and protects against tularemia in mice. Mol Immunol 2009; 46:677-87. [PMID: 18929413 PMCID: PMC3033759 DOI: 10.1016/j.molimm.2008.08.281] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2008] [Accepted: 08/27/2008] [Indexed: 12/01/2022]
Abstract
Francisella tularensis, the causative agent of tularemia, is currently considered a category A bioterrorism agent due to its high virulence. Infection with F. tularensis results in an inflammatory response that plays an important role in the pathogenesis of the disease; however, the cellular mechanisms regulating this response are poorly understood. Glycogen synthase kinase-3beta (GSK3beta) is a serine/threonine protein kinase that has recently emerged as a key regulatory switch in the modulation of the inflammatory response. In this study, we investigated the effect of GSK3beta inhibition in regulating F. tularensis LVS-induced inflammatory responses. F. tularensis LVS infection of murine peritoneal macrophages induced a TLR2 dependent phosphorylation of GSK3beta. Inhibition of GSK3beta resulted in a significant decrease in the production of pro-inflammatory cytokine IL-6, IL-12p40 and TNF-alpha, as well as a significant increase in the production of the anti-inflammatory cytokine IL-10. GSK3beta regulated the F. tularensis LVS-induced cytokine response by differentially affecting the activation of transcription factors NF-kappaB and CREB. Inhibition of GSK3beta by lithium in vivo suppressed the inflammatory response in mice infected with F. tularensis LVS and conferred a survival advantage. In addition, we show that the production of IFN-gamma contributed to the development of tularemia and to the fatal outcome of the infected animals, depending on the timing and the relative level of the IFN-gamma produced. IFN-gamma potentiated F. tularensis LVS-induced cytokine production by increasing GSK3beta activity and the nuclear translocation of NF-kappaB. Taken together, these results demonstrate a regulatory function of GSK3beta in modulating inflammatory responses that can be detrimental to the host during an F. tularensis LVS infection, and suggest that inhibition of GSK3beta may represent a novel therapeutic approach in the treatment of tularemia.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Pediatric Dentistry, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Jenny Katz
- Department of Pediatric Dentistry, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Suzanne M. Michalek
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| |
Collapse
|
75
|
Cremer TJ, Amer A, Tridandapani S, Butchar JP. Francisella tularensis regulates autophagy-related host cell signaling pathways. Autophagy 2009; 5:125-8. [PMID: 19029814 PMCID: PMC2774735 DOI: 10.4161/auto.5.1.7305] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The Gram-negative intracellular pathogen Francisella tularensis is known for its ability to dampen host immune responses. We recently performed a microarray analysis comparing human monocyte responses to the highly virulent F. tularensis tularensis Schu S4 strain (F.t.) versus the less virulent F. tularensis novicida (F.n.).(1) Many groups of genes were affected, including those involved with autophagy and with the regulation of autophagy. Here, we discuss the implications in the context of Francisella virulence and host cell response, then conclude with potential future experiments.
Collapse
Affiliation(s)
- Thomas J. Cremer
- Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, OH 43210
| | - Amal Amer
- The Center of Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
- Division of Pulmonary and Critical Care, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Susheela Tridandapani
- Molecular, Cellular, and Developmental Biology Program, The Ohio State University, Columbus, OH 43210
- The Center of Microbial Interface Biology, The Ohio State University, Columbus, OH 43210
- Division of Pulmonary and Critical Care, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | - Jonathan P. Butchar
- Division of Pulmonary and Critical Care, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
76
|
Bliska JB, Casadevall A. Intracellular pathogenic bacteria and fungi--a case of convergent evolution? Nat Rev Microbiol 2008; 7:165-71. [PMID: 19098923 DOI: 10.1038/nrmicro2049] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The bacterium Yersinia pestis and the fungus Cryptococcus neoformans are the causative agents of human plague and cryptococcosis, respectively. Both microorganisms are facultatively intracellular pathogens. A comparison of their pathogenic strategies reveals similar tactics for intracellular survival in Y. pestis and C. neoformans despite their genetic unrelatedness. Both organisms can survive in environments where they are vulnerable to predation by amoeboid protozoal hosts. Here, we propose that the overall similarities in their pathogenic strategies are an example of convergent evolution that has solved the problem of intracellular survival.
Collapse
Affiliation(s)
- James B Bliska
- Department of Molecular Genetics and Microbiology and Center for Infectious Diseases, School of Medicine, Stony Brook University, Stony Brook, New York, New York 11794, USA
| | | |
Collapse
|
77
|
Identification of an essential Francisella tularensis subsp. tularensis virulence factor. Infect Immun 2008; 77:152-61. [PMID: 18981253 DOI: 10.1128/iai.01113-08] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Francisella tularensis, the highly virulent etiologic agent of tularemia, is a low-dose intracellular pathogen that is able to escape from the phagosome and replicate in the cytosol. Although there has been progress in identifying loci involved in the pathogenicity of this organism, analysis of the genome sequence has revealed few obvious virulence factors. We previously reported isolation of an F. tularensis subsp. tularensis strain Schu S4 transposon insertion mutant with a mutation in a predicted hypothetical lipoprotein, FTT1103, that was deficient in intracellular replication in HepG2 cells. In this study, a mutant with a defined nonpolar deletion in FTT1103 was created, and its phenotype, virulence, and vaccine potential were characterized. A phagosomal integrity assay and lysosome-associated membrane protein 1 colocalization revealed that DeltaFTT1103 mutant bacteria were defective in phagosomal escape. FTT1103 mutant bacteria were maximally attenuated in the mouse model; mice survived, without visible signs of illness, challenge by more than 10(10) CFU when the intranasal route was used and challenge by 10(6) CFU when the intraperitoneal, subcutaneous, or intravenous route was used. The FTT1103 mutant bacteria exhibited dissemination defects. Mice that were infected by the intranasal route had low levels of bacteria in their livers and spleens, and these bacteria were cleared by 3 days postinfection. Mutant bacteria inoculated by the subcutaneous route failed to disseminate to the lungs. BALB/c or C57BL/6 mice that were intranasally vaccinated with 10(8) CFU of FTT1103 mutant bacteria were protected against subsequent challenge with wild-type strain Schu S4. These experiments identified the FTT1103 protein as an essential virulence factor and also demonstrated the feasibility of creating defined attenuated vaccines based on a type A strain.
Collapse
|
78
|
Genome-wide screen in Francisella novicida for genes required for pulmonary and systemic infection in mice. Infect Immun 2008; 77:232-44. [PMID: 18955478 DOI: 10.1128/iai.00978-08] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Francisella tularensis is a gram-negative, highly infectious, aerosolizable facultative intracellular pathogen that causes the potentially life-threatening disease tularemia. To date there is no approved vaccine available, and little is known about the molecular mechanisms important for infection, survival, and dissemination at different times of infection. We report the first whole-genome screen using an inhalation mouse model to monitor infection in the lung and dissemination to the liver and spleen. We queried a comprehensive library of 2,998 sequence-defined transposon insertion mutants in Francisella novicida strain U112 using a microarray-based negative-selection screen. We were able to track the behavior of 1,029 annotated genes, equivalent to a detection rate of 75% and corresponding to approximately 57% of the entire F. novicida genome. As expected, most transposon mutants retained the ability to colonize, but 125 candidate virulence genes (12%) could not be detected in at least one of the three organs. They fell into a variety of functional categories, with one-third having no annotated function and a statistically significant enrichment of genes involved in transcription. Based on the observation that behavior during complex pool infections correlated with the degree of attenuation during single-strain infection we identified nine genes expected to strongly contribute to infection. These included two genes, those for ATP synthase C (FTN_1645) and thioredoxin (FTN_1415), that when mutated allowed increased host survival and conferred protection in vaccination experiments.
Collapse
|
79
|
Kirimanjeswara GS, Olmos S, Bakshi CS, Metzger DW. Humoral and cell-mediated immunity to the intracellular pathogen Francisella tularensis. Immunol Rev 2008; 225:244-55. [PMID: 18837786 PMCID: PMC4871322 DOI: 10.1111/j.1600-065x.2008.00689.x] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
SUMMARY Francisella tularensis can cause fatal respiratory tularemia in humans and animals and is increasingly being isolated in the United States and several European countries. The correlates of protective immunity against this intracellular bacterium are not known, and currently there are no licensed vaccines available for human use. Cell-mediated immunity has long been believed to be critical for protection, and the importance of humoral immunity is also now recognized. Furthermore, synergy between antibodies, T cell-derived cytokines, and phagocytes appears to be critical to achieve sterilizing immunity against F. tularensis. Thus, novel vaccine approaches should be designed to induce robust antibody and cell-mediated immune responses to this pathogen.
Collapse
Affiliation(s)
| | - Sofia Olmos
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, 12208, NY
| | - Chandra S. Bakshi
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, 12208, NY
| | - Dennis W. Metzger
- Center for Immunology and Microbial Disease, Albany Medical College, Albany, 12208, NY
| |
Collapse
|
80
|
Tamilselvam B, Daefler S. Francisella targets cholesterol-rich host cell membrane domains for entry into macrophages. THE JOURNAL OF IMMUNOLOGY 2008; 180:8262-71. [PMID: 18523292 DOI: 10.4049/jimmunol.180.12.8262] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Francisella tularensis is a pathogen optimally adapted to efficiently invade its respective host cell and to proliferate intracellularly. We investigated the role of host cell membrane microdomains in the entry of F. tularensis subspecies holarctica vaccine strain (F. tularensis live vaccine strain) into murine macrophages. F. tularensis live vaccine strain recruits cholesterol-rich lipid domains ("lipid rafts") with caveolin-1 for successful entry into macrophages. Interference with lipid rafts through the depletion of plasma membrane cholesterol, through induction of raft internalization with choleratoxin, or through removal of raft-associated GPI-anchored proteins by treatment with phosphatidylinositol phospholipase C significantly inhibited entry of Francisella and its intracellular proliferation. Lipid raft-associated components such as cholesterol and caveolin-1 were incorporated into Francisella-containing vesicles during entry and the initial phase of intracellular trafficking inside the host cell. These findings demonstrate that Francisella requires cholesterol-rich membrane domains for entry into and proliferation inside macrophages.
Collapse
|
81
|
Francisella tularensis subsp. tularensis Schu S4 disulfide bond formation protein B, but not an RND-type efflux pump, is required for virulence. Infect Immun 2008; 76:3086-92. [PMID: 18458069 DOI: 10.1128/iai.00363-08] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Francisella tularensis subsp. tularensis is a highly virulent bacterium that is a CDC select agent. Despite advancements in the understanding of its biology, details pertaining to virulence are poorly understood. In previous work, we identified a transposon insertion mutant in the FTT0107c locus that was defective in intracellular survival in HepG2 and J77A.1 cells. Here, we report that this mutant was also highly attenuated in vivo. The FTT0107c locus is predicted to encode an ortholog of the disulfide bond formation B protein (DsbB). This designation was confirmed by complementation of an Escherichia coli dsbB mutant. This dsbB mutant of Schu S4 was highly attenuated in mice, but unlike what has been reported for Francisella novicida, intranasal immunization with a sublethal dose did not induce protection against wild-type challenge. dsbB was found to be transcribed in an operon with acrA and acrB, which encode an RND-type efflux pump. However, this pump did not make a significant contribution to virulence because strains with nonpolar deletions in acrA and acrB behaved like wild-type strain Schu S4 with respect to intracellular growth and in vivo virulence. This result is in contrast to a report that an acrB mutant of a live vaccine strain of F. tularensis has decreased virulence in mice. Overall, these results demonstrate key differences between the virulence requirements of Schu S4 and less virulent subspecies of Francisella. We have shown that DsbB is a key participant in intracellular growth and virulence, and our results suggest that there are critical virulence factors that contain disulfide bonds.
Collapse
|
82
|
Type IV pili in Francisella tularensis: roles of pilF and pilT in fiber assembly, host cell adherence, and virulence. Infect Immun 2008; 76:2852-61. [PMID: 18426883 DOI: 10.1128/iai.01726-07] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Francisella tularensis, a highly virulent facultative intracellular bacterium, is the causative agent of tularemia. Genome sequencing of all F. tularensis subspecies revealed the presence of genes that could encode type IV pili (Tfp). The live vaccine strain (LVS) expresses surface fibers resembling Tfp, but it was not established whether these fibers were indeed Tfp encoded by the pil genes. We show here that deletion of the pilF putative Tfp assembly ATPase in the LVS resulted in a complete loss of surface fibers. Disruption of the pilT putative disassembly ATPase also caused a complete loss of pili, indicating that pilT functions differently in F. tularensis than in model Tfp systems such as those found in Pseudomonas aeruginosa and Neisseria spp. The LVS pilF and pilT mutants were attenuated for virulence in a mouse model of tularemia by the intradermal route. Furthermore, although absence of pili had no effect on the ability of the LVS to replicate intracellularly, the pilF and pilT mutants were defective for adherence to macrophages, pneumocytes, and hepatocytes. This work confirms that the surface fibers expressed by the LVS are encoded by the pil genes and provides evidence that the Francisella pili contribute to host cell adhesion and virulence.
Collapse
|
83
|
Acquisition of the vacuolar ATPase proton pump and phagosome acidification are essential for escape of Francisella tularensis into the macrophage cytosol. Infect Immun 2008; 76:2671-7. [PMID: 18390995 DOI: 10.1128/iai.00185-08] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The Francisella tularensis-containing phagosome (FCP) matures to a late-endosome-like phagosome prior to bacterial escape into the cytosols of macrophages, where bacterial proliferation occurs. Our data show that within the first 15 min after infection of primary human monocyte-derived macrophages (hMDMs), approximately 90% of the FCPs acquire the proton vacuolar ATPase (vATPase) pump and the lysomotropic dye LysoTracker, which concentrates in acidic compartments, similar to phagosomes harboring the Listeria monocytogenes control. The acquired proton vATPase pump and lysomotropic dye are gradually lost by 30 to 60 min postinfection, which coincides with bacterial escape into the cytosols of hMDMs. Colocalization of phagosomes harboring the iglD mutant with the vATPase pump and the LysoTracker dye was also transient, and the loss of colocalization was faster than that observed for the wild-type strain, which is consistent with the faster escape of the iglD mutant into the macrophage cytosol. In contrast, colocalization of both makers with phagosomes harboring the iglC mutant was persistent, which is consistent with fusion to the lysosomes and failure of the iglC mutant to escape into the macrophage cytosol. We have utilized a fluorescence microscopy-based phagosome integrity assay for differential labeling of vacuolar versus cytosolic bacteria, using antibacterial antibodies loaded into the cytosols of live hMDMs. We show that specific inhibition of the proton vATPase pump by bafilomycin A1 (BFA) blocks rapid bacterial escape into the cytosols of hMDMs, but 30% to 50% of the bacteria escape into the cytosol by 6 to 12 h after BFA treatment. The effect of BFA on the blocking of bacterial escape into the cytosol is completely reversible, as the bacteria escape after removal of BFA. We also show that the limited fusion of the FCP to lysosomes is not due to failure to recruit the late-endosomal fusion regulator Rab7. Therefore, within few minutes of its biogenesis, the FCP transiently acquires the proton vATPase pump to acidify the phagosome, and this transient acidification is essential for subsequent bacterial escape into the macrophage cytosol.
Collapse
|
84
|
Allen LAH, McCaffrey RL. To activate or not to activate: distinct strategies used by Helicobacter pylori and Francisella tularensis to modulate the NADPH oxidase and survive in human neutrophils. Immunol Rev 2007; 219:103-17. [PMID: 17850485 DOI: 10.1111/j.1600-065x.2007.00544.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Neutrophils accumulate rapidly at sites of infection, and the ability of these cells to phagocytose and kill microorganisms is an essential component of the innate immune response. Relatively few microbial pathogens are able to evade neutrophil killing. Herein, we describe the novel strategies used by Helicobacter pylori and Francisella tularensis to disrupt neutrophil function, with a focus on assembly and activation of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase.
Collapse
Affiliation(s)
- Lee-Ann H Allen
- Inflammation Program, Department of Internal Medicine, University of Iowa, VA Medical Center, Iowa City, IA 52241, USA.
| | | |
Collapse
|
85
|
Sun P, Austin BP, Schubot FD, Waugh DS. New protein fold revealed by a 1.65 A resolution crystal structure of Francisella tularensis pathogenicity island protein IglC. Protein Sci 2007; 16:2560-3. [PMID: 17905833 PMCID: PMC2211698 DOI: 10.1110/ps.073177307] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Francisella tularensis is a highly infectious Gram-negative intracellular pathogen that causes the fulminating disease tularemia and is considered to be a potential bioweapon. F. tularensis pathogenicity island proteins play a key role in modulating phagosome biogenesis and subsequent bacterial escape into the cytoplasm of macrophages. The 23 kDa pathogenicity island protein IglC is essential for the survival and proliferation of F. tularensis in macrophages. Seeking to gain some insight into its function, we determined the crystal structure of IglC at 1.65 A resolution. IglC adopts a beta-sandwich conformation that exhibits no similarity with any known protein structure.
Collapse
Affiliation(s)
- Ping Sun
- Macromolecular Crystallography Laboratory, Center for Cancer Research, National Cancer Institute at Frederick, MD 21702-1201, USA
| | | | | | | |
Collapse
|
86
|
Butchar JP, Rajaram MVS, Ganesan LP, Parsa KVL, Clay CD, Schlesinger LS, Tridandapani S. Francisella tularensis induces IL-23 production in human monocytes. THE JOURNAL OF IMMUNOLOGY 2007; 178:4445-54. [PMID: 17372002 DOI: 10.4049/jimmunol.178.7.4445] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Francisella tularensis, the causative agent of tularemia, is phagocytosed by immune cells such as monocytes and macrophages. Instead of being destroyed in the phagolysosome, the bacterium escapes the phagosome and replicates within the host cytosol. Recent studies indicate that phagosomal escape may have a major impact on the nature of the inflammatory cytokine response to infection. To better understand the host cell response to Francisella infection, we exposed human peripheral blood monocytes to Francisella novicida and analyzed transcriptional changes using high-density oligonucleotide microarrays. Results showed a nearly 300-fold up-regulation of transcripts for the p19 subunit of IL-23, and a nearly 18-fold up-regulation for the p40 subunit of IL-12. IL-23 is formed by the heterodimerization of p19 and p40, and is an important cytokine of the innate immune response. Up-regulation of p19 and p40 was confirmed at the protein level by Western blotting and ELISA analyses, and was found to be largely dependent on PI3K and NF-kappaB activity. Studies using medium from infected monocytes with or without a p19 blocking Ab showed that the secreted IL-23 induced IFN-gamma production from NK cells, suggesting a potential biologically important role for IL-23 in host defense. Finally, infection of human monocytes by the highly virulent Francisella SCHU S4 strain likewise led to IL-23 production, suggesting that the IL-23 response may be relevant during tularemia.
Collapse
Affiliation(s)
- Jonathan P Butchar
- Department of Internal Medicine, Division of Infectious Diseases, Ohio State University, 473 West 12th Avenue, Columbus, OH 43210, USA
| | | | | | | | | | | | | |
Collapse
|
87
|
Shaffer SA, Harvey MD, Goodlett DR, Ernst RK. Structural heterogeneity and environmentally regulated remodeling of Francisella tularensis subspecies novicida lipid A characterized by tandem mass spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2007; 18:1080-92. [PMID: 17446084 PMCID: PMC2743246 DOI: 10.1016/j.jasms.2007.03.008] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/21/2006] [Revised: 03/16/2007] [Accepted: 03/20/2007] [Indexed: 05/15/2023]
Abstract
The structural characterization of environmentally-regulated lipid A derived from Francisella tularensis subspecies novicida (Fn) U112 is described using negative electrospray ionization with a linear ion trap Fourier transform ion cyclotron resonance (IT-FT-ICR) hybrid mass spectrometer. The results indicate that a unique profile of lipid A molecular structures are synthesized in response to Fn growth at 25 degrees C versus 37 degrees C. Molecular species were found to be tetra-acylated, sharing a conserved glucosamine disaccharide backbone, a galactosamine-1-phosphate linked to the reducing glucosamine, and multiple O- and N-linked fatty acyl groups. Deprotonated molecules were interrogated by MS(n) scanning techniques at both high and nominal mass resolution and were found to be complex heterogeneous mixtures where structures differed based on the positions and identities of the O- and N-linked fatty acyl substituents. For the dominant ion series, which consisted of five peaks, 30 unique lipid A structures were identified. Estimates for the relative abundance of each structure were derived from MS relative abundance ratios and fragment ion ratios from comparable dissociation pathways from MS(2) through MS(4) experiments. The results suggest a remodeling pathway in which the amide linked fatty acid of the reducing glucosamine favors a 3-hydroxyhexadecanoic acid substituent for growth conditions at 25 degrees C versus a 3-hydroxyoctadecanoic acid substituent for growth conditions at 37 degrees C.
Collapse
Affiliation(s)
- Scott A Shaffer
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195, USA
| | | | | | | |
Collapse
|
88
|
Santic M, Molmeret M, Barker JR, Klose KE, Dekanic A, Doric M, Abu Kwaik Y. A Francisella tularensis pathogenicity island protein essential for bacterial proliferation within the host cell cytosol. Cell Microbiol 2007; 9:2391-403. [PMID: 17517064 DOI: 10.1111/j.1462-5822.2007.00968.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Francisella tularensis is an intracellular bacterial pathogen, and is a category A bioterrorism agent. Within quiescent human macrophages, the F. tularensis pathogenicity island (FPI) is essential for bacterial growth within quiescent macrophages. The F. tularensis-containing phagosome matures to a late endosome-like stage that does not fuse to lysosomes for 1-8 h, followed by gradual bacterial escape into the macrophage cytosol. Here we show that the FPI protein IglD is essential for intracellular replication in primary human monocyte-derived macrophages (hMDMs). While the parental strain replicates robustly in pulmonary, hepatic and splenic tissues of BALB/c mice associated with severe immunopathologies, the isogenic iglD mutant is severely defective. Within hMDMs, the iglD mutant-containing phagosomes mature to either a late endosome-like phagosome, similar to the parental strain, or to a phagolysosome, similar to phagosomes harbouring the iglC mutant control. Despite heterogeneity and alterations in phagosome biogenesis, the iglD mutant bacteria escape into the cytosol faster than the parental strain within hMDMs and pulmonary cells of BALB/c mice. Co-infections of hMDMs with the wild-type strain and the iglD mutant, or super-infection of iglD mutant-infected hMDMs with the wild-type strain show that the mutant strain replicates robustly within the cytosol of hMDMs coinhabited by the wild strain. However, when the wild-type strain-infected hMDMs are super-infected by the iglD mutant, the mutant fails to replicate in the cytosol of communal macrophages. This is the first demonstration of a F. tularensis novel protein essential for proliferation in the macrophage cytosol. Our data indicate that F. tularensis transduces signals to the macrophage cytosol to remodel it into a proliferative niche, and IglD is essential for transduction of these signals.
Collapse
Affiliation(s)
- Marina Santic
- Department of Microbiology and Immunology, University of Louisville, College of Medicine, Louisville, KY 40202, USA
| | | | | | | | | | | | | |
Collapse
|
89
|
Hsu YM, Shieh HK, Chen WH, Sun TY, Shiang JH. Antimicrobial susceptibility, plasmid profiles and haemocin activities of Avibacterium paragallinarum strains. Vet Microbiol 2007; 124:209-18. [PMID: 17485180 DOI: 10.1016/j.vetmic.2007.04.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2006] [Revised: 03/30/2007] [Accepted: 04/04/2007] [Indexed: 11/26/2022]
Abstract
In this study, 18 Avibacterium paragallinarum isolates collected in Taiwan from 1990 to 2003 were serotyped and tested for resistance to antimicrobial agents. Serotyping revealed that 13 isolates were Page serovar A and 5 isolates were Page serovar C. More than 75% of the isolates were resistant to neomycin, streptomycin and erythromycin. The most common resistance pattern (15 isolates, 83.3%) was neomycin-streptomycin. Furthermore, 88.9% of the isolates were resistant to two or more antibiotics. About 72% of isolates contained plasmids (pYMH5 and/or pA14). Plasmid pYMH5 encoded functional streptomycin, sulfonamide, kanamycin and neomycin resistance genes and revealed significant homology to a broad host-range plasmid, pLS88. Plasmid pA14 encoded a putative MglA protein and RNase II, both of which might be associated with virulence. Furthermore, seven isolates showed haemocin activity. Plasmid pYMH5 is the first multidrug-resistance plasmid reported in A. paragallinarum and it may facilitate the spread of antibiotic-resistance genes between bacteria. The putative virulence plasmid pA14 and haemocin-like activity in A. paragallinarum indicate two possible mechanisms which might be responsible for the pathogenesis.
Collapse
Affiliation(s)
- Yuan-Man Hsu
- Department of Biological Science and Technology, China Medical University, 91, Hsueh-Shih Road, Taichung 404, Taiwan.
| | | | | | | | | |
Collapse
|
90
|
Su J, Yang J, Zhao D, Kawula TH, Banas JA, Zhang JR. Genome-wide identification of Francisella tularensis virulence determinants. Infect Immun 2007; 75:3089-101. [PMID: 17420240 PMCID: PMC1932872 DOI: 10.1128/iai.01865-06] [Citation(s) in RCA: 150] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Francisella tularensis is a gram-negative pathogen that causes life-threatening infections in humans and has potential for use as a biological weapon. The genetic basis of the F. tularensis virulence is poorly understood. This study screened a total of 3,936 transposon mutants of the live vaccine strain for infection in a mouse model of respiratory tularemia by signature-tagged mutagenesis. We identified 341 mutants attenuated for infection in the lungs. The transposon disruptions were mapped to 95 different genes, virtually all of which are also present in the genomes of other F. tularensis strains, including human pathogenic F. tularensis strain Schu S4. A small subset of these attenuated mutants carried insertions in the genes encoding previously known virulence factors, but the majority of the identified genes have not been previously linked to F. tularensis virulence. Among these are genes encoding putative membrane proteins, proteins associated with stress responses, metabolic proteins, transporter proteins, and proteins with unknown functions. Several attenuated mutants contained disruptions in a putative capsule locus which partially resembles the poly-gamma-glutamate capsule biosynthesis locus of Bacillus anthracis, the anthrax agent. Deletional mutation analysis confirmed that this locus is essential for F. tularensis virulence.
Collapse
Affiliation(s)
- Jingliang Su
- Center for Immunology and Microbial Disease, Albany Medical College, 47 New Scotland Avenue, Albany, NY 12208, USA
| | | | | | | | | | | |
Collapse
|
91
|
Woolard MD, Wilson JE, Hensley LL, Jania LA, Kawula TH, Drake JR, Frelinger JA. Francisella tularensis-infected macrophages release prostaglandin E2 that blocks T cell proliferation and promotes a Th2-like response. THE JOURNAL OF IMMUNOLOGY 2007; 178:2065-74. [PMID: 17277110 DOI: 10.4049/jimmunol.178.4.2065] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Francisella tularensis is a highly infectious bacterial pathogen, and is likely to have evolved strategies to evade and subvert the host immune response. In this study, we show that F. tularensis infection of macrophages alters T cell responses in vitro, by blocking T cell proliferation and promoting a Th2-like response. We demonstrate that a soluble mediator is responsible for this effect and identify it as PGE(2). Supernatants from F. tularensis-infected macrophages inhibited IL-2 secretion from both MHC class I and MHC class II-restricted T cell hybridomas, as well as enhanced a Th2-like response by inducing increased production of IL-5. Furthermore, the soluble mediator blocked proliferation of naive MHC class I-restricted T cells when stimulated with cognate tetramer. Indomethacin treatment partially restored T cell proliferation and lowered IL-5 production to wild-type levels. Macrophages produced PGE(2) when infected with F. tularensis, and treatment of infected macrophages with indomethacin, a cyclooxygenase-1/cyclooxygenase-2 inhibitor, blocked PGE(2) production. To further demonstrate that PGE(2) was responsible for skewing of T cell responses, we infected macrophages from membrane PGE synthase 1 knockout mice (mPGES1(-/-)) that cannot produce PGE(2). Supernatants from F. tularensis-infected membrane PGE synthase 1(-/-) macrophages did not inhibit T cell proliferation. Furthermore, treatment of T cells with PGE(2) recreated the effects seen with infected supernatant. From these data, we conclude that F. tularensis can alter host T cell responses by causing macrophages to produce PGE(2). This study defines a previously unknown mechanism used by F. tularensis to modulate adaptive immunity.
Collapse
Affiliation(s)
- Matthew D Woolard
- Department of Microbiology and Immunology, University of North Carolina Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | |
Collapse
|
92
|
Lindemann SR, McLendon MK, Apicella MA, Jones BD. An in vitro model system used to study adherence and invasion of Francisella tularensis live vaccine strain in nonphagocytic cells. Infect Immun 2007; 75:3178-82. [PMID: 17339345 PMCID: PMC1932879 DOI: 10.1128/iai.01811-06] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
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.
Collapse
Affiliation(s)
- Stephen R Lindemann
- Department of Microbiology, University of Iowa School of Medicine, 51 Newton Road, Iowa City, IA 52242-1109, USA
| | | | | | | |
Collapse
|
93
|
Gallagher LA, Ramage E, Jacobs MA, Kaul R, Brittnacher M, Manoil C. A comprehensive transposon mutant library of Francisella novicida, a bioweapon surrogate. Proc Natl Acad Sci U S A 2007; 104:1009-14. [PMID: 17215359 PMCID: PMC1783355 DOI: 10.1073/pnas.0606713104] [Citation(s) in RCA: 220] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Francisella tularensis, the causative agent of tularemia, is one of the most infectious bacterial pathogens known and is a category A select agent. We created a sequence-defined, near-saturation transposon mutant library of F. tularensis novicida, a subspecies that causes a tularemia-like disease in rodents. The library consists of 16,508 unique insertions, an average of >9 insertions per gene, which is a coverage nearly twice that of the greatest previously achieved for any bacterial species. Insertions were recovered in 84% (1,490) of the predicted genes. To achieve high coverage, it was necessary to construct transposons carrying an endogenous Francisella promoter to drive expression of antibiotic resistance. An analysis of genes lacking (or with few) insertions identified nearly 400 candidate essential genes, most of which are likely to be required for growth on rich medium and which represent potential therapeutic targets. To facilitate genome-scale screening using the mutant collection, we assembled a sublibrary made up of two purified mutants per gene. The library provides a resource for virtually complete identification of genes involved in virulence and other nonessential processes.
Collapse
Affiliation(s)
- Larry A. Gallagher
- *Department of Genome Sciences, University of Washington, Campus Box 355065, 1705 NE Pacific Street, Seattle, WA 98195; and
| | - Elizabeth Ramage
- *Department of Genome Sciences, University of Washington, Campus Box 355065, 1705 NE Pacific Street, Seattle, WA 98195; and
| | - Michael A. Jacobs
- Department of Medicine, University of Washington, Campus Box 352145, 1705 NE Pacific Street, Seattle, WA 98195
| | - Rajinder Kaul
- Department of Medicine, University of Washington, Campus Box 352145, 1705 NE Pacific Street, Seattle, WA 98195
| | - Mitchell Brittnacher
- *Department of Genome Sciences, University of Washington, Campus Box 355065, 1705 NE Pacific Street, Seattle, WA 98195; and
| | - Colin Manoil
- *Department of Genome Sciences, University of Washington, Campus Box 355065, 1705 NE Pacific Street, Seattle, WA 98195; and
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|
94
|
Rohmer L, Fong C, Abmayr S, Wasnick M, Larson Freeman TJ, Radey M, Guina T, Svensson K, Hayden HS, Jacobs M, Gallagher LA, Manoil C, Ernst RK, Drees B, Buckley D, Haugen E, Bovee D, Zhou Y, Chang J, Levy R, Lim R, Gillett W, Guenthener D, Kang A, Shaffer SA, Taylor G, Chen J, Gallis B, D'Argenio DA, Forsman M, Olson MV, Goodlett DR, Kaul R, Miller SI, Brittnacher MJ. Comparison of Francisella tularensis genomes reveals evolutionary events associated with the emergence of human pathogenic strains. Genome Biol 2007; 8:R102. [PMID: 17550600 PMCID: PMC2394750 DOI: 10.1186/gb-2007-8-6-r102] [Citation(s) in RCA: 194] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2006] [Revised: 03/02/2007] [Accepted: 06/05/2007] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Francisella tularensis subspecies tularensis and holarctica are pathogenic to humans, whereas the two other subspecies, novicida and mediasiatica, rarely cause disease. To uncover the factors that allow subspecies tularensis and holarctica to be pathogenic to humans, we compared their genome sequences with the genome sequence of Francisella tularensis subspecies novicida U112, which is nonpathogenic to humans. RESULTS Comparison of the genomes of human pathogenic Francisella strains with the genome of U112 identifies genes specific to the human pathogenic strains and reveals pseudogenes that previously were unidentified. In addition, this analysis provides a coarse chronology of the evolutionary events that took place during the emergence of the human pathogenic strains. Genomic rearrangements at the level of insertion sequences (IS elements), point mutations, and small indels took place in the human pathogenic strains during and after differentiation from the nonpathogenic strain, resulting in gene inactivation. CONCLUSION The chronology of events suggests a substantial role for genetic drift in the formation of pseudogenes in Francisella genomes. Mutations that occurred early in the evolution, however, might have been fixed in the population either because of evolutionary bottlenecks or because they were pathoadaptive (beneficial in the context of infection). Because the structure of Francisella genomes is similar to that of the genomes of other emerging or highly pathogenic bacteria, this evolutionary scenario may be shared by pathogens from other species.
Collapse
Affiliation(s)
- Laurence Rohmer
- Department of Genome Sciences, University of Washington, Campus Box 357710, 1705 NE Pacific street Seattle, Washington 98195, USA
| | - Christine Fong
- Department of Genome Sciences, University of Washington, Campus Box 357710, 1705 NE Pacific street Seattle, Washington 98195, USA
| | - Simone Abmayr
- Department of Genome Sciences, University of Washington, Campus Box 357710, 1705 NE Pacific street Seattle, Washington 98195, USA
| | - Michael Wasnick
- Department of Genome Sciences, University of Washington, Campus Box 357710, 1705 NE Pacific street Seattle, Washington 98195, USA
| | - Theodore J Larson Freeman
- Department of Genome Sciences, University of Washington, Campus Box 357710, 1705 NE Pacific street Seattle, Washington 98195, USA
| | - Matthew Radey
- Department of Genome Sciences, University of Washington, Campus Box 357710, 1705 NE Pacific street Seattle, Washington 98195, USA
| | - Tina Guina
- Department of Pediatrics, Division of Infectious Diseases, University of Washington, Campus Box 357710, 1720 NE Pacific street, Seattle, Washington 98195, USA
| | - Kerstin Svensson
- NBC Analysis, Division of NBC Defence, Swedish Defence Research Agency, SE-901 82 Umeå, Sweden
- Department of Clinical Microbiology, Infectious Diseases, Umeå University, SE-901 85 Umeå, Sweden
| | - Hillary S Hayden
- University of Washington Genome Center, University of Washington, Campus Box 352145, Mason Road, Seattle, Washington 98195, USA
| | - Michael Jacobs
- University of Washington Genome Center, University of Washington, Campus Box 352145, Mason Road, Seattle, Washington 98195, USA
| | - Larry A Gallagher
- Department of Genome Sciences, University of Washington, Campus Box 357710, 1705 NE Pacific street Seattle, Washington 98195, USA
| | - Colin Manoil
- Department of Genome Sciences, University of Washington, Campus Box 357710, 1705 NE Pacific street Seattle, Washington 98195, USA
| | - Robert K Ernst
- Department Medicine, University of Washington, Seattle, Washington 98195, USA
| | - Becky Drees
- Department of Microbiology, University of Washington, Box 357242, 1720 NE Pacific street, Seattle, Washington 98195, USA
| | - Danielle Buckley
- University of Washington Genome Center, University of Washington, Campus Box 352145, Mason Road, Seattle, Washington 98195, USA
| | - Eric Haugen
- University of Washington Genome Center, University of Washington, Campus Box 352145, Mason Road, Seattle, Washington 98195, USA
| | - Donald Bovee
- University of Washington Genome Center, University of Washington, Campus Box 352145, Mason Road, Seattle, Washington 98195, USA
| | - Yang Zhou
- University of Washington Genome Center, University of Washington, Campus Box 352145, Mason Road, Seattle, Washington 98195, USA
| | - Jean Chang
- University of Washington Genome Center, University of Washington, Campus Box 352145, Mason Road, Seattle, Washington 98195, USA
| | - Ruth Levy
- University of Washington Genome Center, University of Washington, Campus Box 352145, Mason Road, Seattle, Washington 98195, USA
| | - Regina Lim
- University of Washington Genome Center, University of Washington, Campus Box 352145, Mason Road, Seattle, Washington 98195, USA
| | - Will Gillett
- University of Washington Genome Center, University of Washington, Campus Box 352145, Mason Road, Seattle, Washington 98195, USA
| | - Don Guenthener
- University of Washington Genome Center, University of Washington, Campus Box 352145, Mason Road, Seattle, Washington 98195, USA
| | - Allison Kang
- University of Washington Genome Center, University of Washington, Campus Box 352145, Mason Road, Seattle, Washington 98195, USA
| | - Scott A Shaffer
- Department of Medicinal Chemistry, Box 357610, University of Washington, Seattle, Washington 98195, USA
| | - Greg Taylor
- Department of Medicinal Chemistry, Box 357610, University of Washington, Seattle, Washington 98195, USA
| | - Jinzhi Chen
- Department of Medicinal Chemistry, Box 357610, University of Washington, Seattle, Washington 98195, USA
| | - Byron Gallis
- Department of Medicinal Chemistry, Box 357610, University of Washington, Seattle, Washington 98195, USA
| | - David A D'Argenio
- Department of Microbiology, University of Washington, Box 357242, 1720 NE Pacific street, Seattle, Washington 98195, USA
| | - Mats Forsman
- NBC Analysis, Division of NBC Defence, Swedish Defence Research Agency, SE-901 82 Umeå, Sweden
| | - Maynard V Olson
- Department of Genome Sciences, University of Washington, Campus Box 357710, 1705 NE Pacific street Seattle, Washington 98195, USA
- University of Washington Genome Center, University of Washington, Campus Box 352145, Mason Road, Seattle, Washington 98195, USA
- Department Medicine, University of Washington, Seattle, Washington 98195, USA
| | - David R Goodlett
- Department of Medicinal Chemistry, Box 357610, University of Washington, Seattle, Washington 98195, USA
| | - Rajinder Kaul
- University of Washington Genome Center, University of Washington, Campus Box 352145, Mason Road, Seattle, Washington 98195, USA
- Department Medicine, University of Washington, Seattle, Washington 98195, USA
| | - Samuel I Miller
- Department of Genome Sciences, University of Washington, Campus Box 357710, 1705 NE Pacific street Seattle, Washington 98195, USA
- Department Medicine, University of Washington, Seattle, Washington 98195, USA
- Department of Microbiology, University of Washington, Box 357242, 1720 NE Pacific street, Seattle, Washington 98195, USA
| | - Mitchell J Brittnacher
- Department of Genome Sciences, University of Washington, Campus Box 357710, 1705 NE Pacific street Seattle, Washington 98195, USA
| |
Collapse
|
95
|
Thomas RM, Titball RW, Oyston PCF, Griffin K, Waters E, Hitchen PG, Michell SL, Grice ID, Wilson JC, Prior JL. The immunologically distinct O antigens from Francisella tularensis subspecies tularensis and Francisella novicida are both virulence determinants and protective antigens. Infect Immun 2006; 75:371-8. [PMID: 17074846 PMCID: PMC1828428 DOI: 10.1128/iai.01241-06] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We have determined the sequence of the gene cluster encoding the O antigen in Francisella novicida and compared it to the previously reported O-antigen cluster in Francisella tularensis subsp. tularensis. Immunization with purified lipopolysaccharide (LPS) from F. tularensis subsp. tularensis or F. novicida protected against challenge with Francisella tularensis subsp. holarctica and F. novicida, respectively. The LPS from F. tularensis subsp. tularensis did not confer protection against challenge with F. novicida, and the LPS from F. novicida did not confer protection against challenge with F. tularensis subsp. holarctica. Allelic replacement mutants of F. tularensis subsp. tularensis or F. novicida which failed to produce O antigen were attenuated, but exposure to these mutants did not induce a protective immune response. The O antigen of F. tularensis subsp. tularensis appeared to be important for intracellular survival whereas the O antigen of F. novicida appeared to be critical for serum resistance and less important for intracellular survival.
Collapse
Affiliation(s)
- Rebecca M Thomas
- Defence Science and Technology Laboratory, Porton Down, Salisbury, Wiltshire, SP4 0JQ, United Kingdom.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Raynaud C, Meibom KL, Lety MA, Dubail I, Candela T, Frapy E, Charbit A. Role of the wbt locus of Francisella tularensis in lipopolysaccharide O-antigen biogenesis and pathogenicity. Infect Immun 2006; 75:536-41. [PMID: 17030571 PMCID: PMC1828372 DOI: 10.1128/iai.01429-06] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Francisella tularensis is a highly infectious bacterial pathogen, responsible for the zoonotic disease tularemia. We screened a bank of transposon insertion mutants of F. tularensis subsp. holarctica LVS for colony morphology alterations and selected a mutant with a transposon insertion in wbtA, the first gene of the predicted lipopolysaccharide O-antigen gene cluster. Inactivation of wbtA led to the complete loss of O antigen, conferred serum sensitivity, impaired intracellular replication, and severely attenuated virulence in the mouse model. Notably, this mutant afforded protection against a challenge against virulent LVS.
Collapse
Affiliation(s)
- Catherine Raynaud
- Université Paris Descartes, Faculté de Médecine René Descartes, Paris F-75015, France
| | | | | | | | | | | | | |
Collapse
|
97
|
McCaffrey RL, Allen LAH. Francisella tularensis LVS evades killing by human neutrophils via inhibition of the respiratory burst and phagosome escape. J Leukoc Biol 2006; 80:1224-30. [PMID: 16908516 PMCID: PMC1828114 DOI: 10.1189/jlb.0406287] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Francisella tularensis is a Gram-negative bacterium and the causative agent of tularemia. Recent data indicate that F. tularensis replicates inside macrophages, but its fate in other cell types, including human neutrophils, is unclear. We now show that F. tularensis live vaccine strain (LVS), opsonized with normal human serum, was rapidly ingested by neutrophils but was not eliminated. Moreover, evasion of intracellular killing can be explained, in part, by disruption of the respiratory burst. As judged by luminol-enhanced chemiluminescence and nitroblue tetrazolium staining, neutrophils infected with live F. tularensis did not generate reactive oxygen species. Confocal microscopy demonstrated that NADPH oxidase assembly was disrupted, and LVS phagosomes did not acquire gp91/p22(phox) or p47/p67(phox). At the same time, F. tularensis also impaired neutrophil activation by heterologous stimuli such as phorbol esters and opsonized zymosan particles. Later in infection, LVS escaped the phagosome, and live organisms persisted in the neutrophil cytosol for at least 12 h. To our knowledge, our data are the first demonstration of a facultative intracellular pathogen, which disrupts the oxidative burst and escapes the phagosome to evade elimination inside neutrophils, and as such, our data define a novel mechanism of virulence.
Collapse
Affiliation(s)
- Ramona L. McCaffrey
- Inflammation Program, University of Iowa and the VA Medical Center, Iowa City, Iowa, USA
- Departments of Medicine, University of Iowa and the VA Medical Center, Iowa City, Iowa, USA
| | - Lee-Ann H. Allen
- Inflammation Program, University of Iowa and the VA Medical Center, Iowa City, Iowa, USA
- Departments of Medicine, University of Iowa and the VA Medical Center, Iowa City, Iowa, USA
- Microbiology, University of Iowa and the VA Medical Center, Iowa City, Iowa, USA
- Correspondence: Inflammation Program, University of Iowa, 2501 Crosspark Rd., MTF-D154, Coralville, IA 52241, USA. E-mail:
| |
Collapse
|
98
|
Qin A, Mann BJ. Identification of transposon insertion mutants of Francisella tularensis tularensis strain Schu S4 deficient in intracellular replication in the hepatic cell line HepG2. BMC Microbiol 2006; 6:69. [PMID: 16879747 PMCID: PMC1557513 DOI: 10.1186/1471-2180-6-69] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2006] [Accepted: 07/31/2006] [Indexed: 11/10/2022] Open
Abstract
Background Francisella tularensis is a zoonotic intracellular bacterial pathogen that causes tularemia. The subspecies tularensis is highly virulent and is classified as a category A agent of biological warfare because of its low infectious dose by an aerosol route, and its ability to cause severe disease. In macrophages F. tularensis exhibits a rather novel intracellular lifestyle; after invasion it remains in a phagosome for three to six hours before escaping to, and replicating in the cytoplasm. The molecular mechanisms that allow F. tularensis to invade and replicate within a host cell have not been well defined. Methods We constructed a stable transposon mutagenesis library of virulent strain Schu S4 using a derivative of the EZ::TN transposon system®. Approximately 2000 mutants were screened for the inability to invade, and replicate in the hepatic carcinoma cell line HepG2. These mutants were also tested for replication within the J774.1 macrophage-like cell line. Results Eighteen mutants defective in intracellular replication in HepG2 cells were identified. Eight of these mutants were auxotrophs; seven had mutations in nucleotide biosynthesis pathways. The remaining mutants had insertions in genes that were predicted to encode putative transporters, enzymes involved in protein modification and turnover, and hypothetical proteins. A time course of the intracellular growth of a pyrB mutant revealed that this mutant was only able to grow at low levels within HepG2 cells but grew like wild-type bacteria in J774.1 cells. This pyrB mutant was also attenuated in mice. Conclusion This is the first reported large-scale mutagenesis of a type A strain of F. tularensis and the first identification of mutants specifically defective in intracellular growth in a hepatic cell line. We have identified several genes and pathways that are key for the survival and growth of F. tularensis in a hepatic cell line, and a number of novel intracellular growth-defective mutants that have not been previously characterized in other pathogens. Further characterization of these mutants will help provide a better understanding of the pathogenicity of F. tularensis, and may have practical applications as targets for drugs or attenuated vaccines.
Collapse
Affiliation(s)
- Aiping Qin
- Department of Internal Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA 22908, USA
| | - Barbara J Mann
- Department of Internal Medicine, Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA 22908, USA
- Department of Microbiology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
99
|
Schulert GS, Allen LAH. Differential infection of mononuclear phagocytes by Francisella tularensis: role of the macrophage mannose receptor. J Leukoc Biol 2006; 80:563-71. [PMID: 16816147 PMCID: PMC1865506 DOI: 10.1189/jlb.0306219] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Francisella tularensis (Ft) is a Gram-negative bacterium and the causative agent of tularemia. It is well established that this organism replicates inside macrophages, but we are only beginning to understand this interface at the molecular level. Herein, we compared directly the ability of Ft subspecies holarctica live-vaccine strain to infect freshly isolated human peripheral blood monocytes, monocyte-derived macrophages (MDM), and cells of the murine macrophage cell line J774A.1 (J774). We now show that unopsonized bacteria infected human MDM fivefold more efficiently than monocytes or J774 cells in standard media. Moreover, enhanced infection of MDM was mediated, in part, by the macrophage mannose receptor (MR). Forming Ft phagosomes accumulated MR, and infection was inhibited by MR-blocking antibody or soluble mannan but not by the dectin-1 ligand laminarin. Up-regulation of MR in MDM (by exposure to interleukin-4) increased Ft phagocytosis, as did expression of MR in J774 cells. Conversely, opsonized Ft were ingested readily by monocytes and MDM. Medium supplementation with 2.5% fresh autologous serum was sufficient to confer opsonophagocytosis and CD11b accumulated in the membrane at sites of Ft engulfment. Infection of monocytes by opsonized Ft was nearly ablated by complement receptor 3 (CR3) blockade. Conversely, MDM used MR and CD11b/CD18 to ingest opsonized organisms. Altogether, our data demonstrate differential infection of mononuclear phagocytes by Ft and define distinct roles for MR and CR3 in phagocytosis.
Collapse
Affiliation(s)
- Grant S. Schulert
- Inflammation Program, University of Iowa and the VA Medical Center, Iowa City
- Department of Microbiology, University of Iowa and the VA Medical Center, Iowa City
| | - Lee-Ann H. Allen
- Inflammation Program, University of Iowa and the VA Medical Center, Iowa City
- Department of Microbiology, University of Iowa and the VA Medical Center, Iowa City
- Department of Internal Medicine, University of Iowa and the VA Medical Center, Iowa City
- Correspondence: Inflammation Program, University of Iowa, 2501 Crosspark Rd., MTF D154, Coralville, IA 52241. E-mail:
| |
Collapse
|