198
Views
14
CrossRef citations to date
0
Altmetric
Review

Host–pathogen interactions and immune evasion strategies in Francisella tularensis pathogenicity

, &
Pages 239-251 | Published online: 18 Sep 2014

Abstract

Francisella tularensis is an intracellular Gram-negative bacterium that causes life-threatening tularemia. Although the prevalence of natural infection is low, F. tularensis remains a tier I priority pathogen due to its extreme virulence and ease of aerosol dissemination. F. tularensis can infect a host through multiple routes, including the intradermal and respiratory routes. Respiratory infection can result from a very small inoculum (ten organisms or fewer) and is the most lethal form of infection. Following infection, F. tularensis employs strategies for immune evasion that delay the immune response, permitting systemic distribution and induction of sepsis. In this review we summarize the current knowledge of F. tularensis in an immunological context, with emphasis on the host response and bacterial evasion of that response.

Introduction

The bacterium Francisella tularensis is the causative agent of the disease tularemia. Originally discovered in ground squirrels in Tulare County, CA, USA, F. tularensis has previously been termed Bacterium tularense and Pasteurella tularensis.Citation1Citation3 The first known infection of a human with F. tularensis was identified in 1913.Citation2 The bacterium has a coccobacillus shape, stains Gram-negative, and is nonmotile.Citation4,Citation5

F. tularensis exists as multiple subspecies, including the “type A” subspecies F. tularensis tularensis and the “type B” subspecies F. tularensis holarctica and F. tularensis mediasiatica. Additionally, there exists a related species, Francisella novicida.Citation6Citation8 Subspecies F. tularensis tularensis, holarctica, and mediasiatica can all cause infection in humans, although only subspecies F. tularensis tularensis is appreciably lethal. By contrast, F. novicida has been reported to cause infection only in immunocompromised individuals.Citation7

Humans can contract infection with F. tularensis through several routes, including arthropod bites, contact with infected animals or animal carcasses, ingestion of contaminated materials, or inhalation.Citation6,Citation9 Infection typically produces a febrile illness, although specific pathology and mortality rates are highly dependent upon the route of infection.Citation1,Citation2,Citation6,Citation10 The most common presentation is glandular or ulceroglandular tularemia, in which infection occurs through the skin and causes localized inflammation and infection of local lymph nodes.Citation6 Ulcers produced by ulceroglandular tularemia are persistent, but the infection is rarely lethal and can often resolve without treatment.Citation6 F. tularensis can also cause typhoidal tularemia, which is characterized by systemic infection without lymphadenopathy or development of ulcers.Citation6 Typhoidal tularemia is more serious, with a mortality rate potentially as high as 60%.Citation6 Less commonly, infection can present as oropharyngeal or gastrointestinal tularemia resulting from ingestion of contaminated materials, or as oculoglandular tularemia as a result of infection of the conjunctiva of the eyes.Citation6 The most serious form of Francisella infection, pneumonic tularemia, is typically caused by inhalation of bacterial aerosols or airborne suspensions of infectious material. Type A F. tularensis is extraordinarily infectious and highly lethal when infection occurs through the respiratory route, with a case fatality rate in excess of 60% and a minimum infectious dose of fewer than 10 colony-forming units (CFU).Citation10

The extreme respiratory infectivity of type A F. tularensis and its high survivability as an aerosol, combined with the high lethality of the pneumonic form of infection, have made the organism an attractive candidate for weaponization, and several governments produced large quantities of F. tularensis for that purpose during the 20th century.Citation8,Citation11Citation14 Today, F. tularensis remains a potential agent of biological terrorism.Citation14 To date, there is no licensed vaccine against F. tularensis. An attenuated strain of type B F. tularensis, the live vaccine strain (LVS), was developed in the mid-20th century by serial passage of F. tularensis holarctica through mice.Citation15,Citation16 LVS is attenuated in humans but retains virulence in mice. Salomonsson et alCitation17 found that complementation of LVS with the genes pilA and FTT0918 restored virulence to the level of virulent type B strains in subcutaneous infection. However, the mechanisms responsible for attenuation of LVS, particularly in models of respiratory infection, remain incompletely understood, leading to fears that LVS may revert to virulence or may cause disease in immunocompromised individuals.

Virulence determinants

Several virulence determinants have been proposed for F. tularensis, many of which are expressed as components of the Francisella pathogenicity island.Citation18 These bacterial products, as well as others, augment virulence through several mechanisms, including suppression of the host immune response and facilitation of phagosomal escape and intracellular survival.

The Francisella pathogenicity island was first described in 2004.Citation19 It contains 17 open reading frames of varying size, several of which have been identified as essential for pathogenesis.Citation19 Intriguingly, the Francisella pathogenicity island is characterized by a lower percentage of guanine and cytosine nucleotides compared with the rest of the Francisella genome, which itself has a fairly low guanine and cytosine content.Citation18,Citation19 In the study that identified the Francisella pathogenicity island, the pdpA gene was identified as being essential for virulence, but its function and the functions of the other pdp genes are not known.Citation19,Citation20 The pdpD gene was found to be present in F. novicida and in type A subspecies of F. tularensis but not in type B subspecies, and it has been implicated in intramacrophage survival.Citation18,Citation19 Several genes within the Francisella pathogenicity island, including vgrG, dotU, and many of the Igl genes, share sequence homology as well as biochemical and structural characteristics with type VI secretion genes of other bacteria.Citation21,Citation22 It is therefore not surprising that the products of vgrG and IglI have been found to be secreted by the bacterium during intracellular infection.Citation23 Several Francisella pathogenicity island proteins, including the IglC protein, have been shown to inhibit phagosome maturation.Citation24,Citation25 These proteins also appear to be involved in escape from the phagosome and intracellular survival. IglC and clpB have also been implicated in evasion of immunity, as F. novicida mutants with defects in these genes fail to induce secretion of immunosuppressive prostaglandin E2 (PGE2).Citation26 It is worth noting that most of the proteins secreted by the Francisella type VI secretion system, with the exception of vgrG, are unique to the Francisella genus.Citation27

Control of reactive oxygen species (ROS) and nitrogen species is also an essential component of Francisella virulence. The enzyme KatG possesses both catalase and peroxidase properties, allowing it to detoxify hydrogen peroxide as well as reactive nitrogen species.Citation28,Citation29 KatG plays a significant role in the virulence of F. tularensis LVS, as mutants lacking a functional katG gene failed to kill mice after intraperitoneal inoculation.Citation29 In contrast, katG mutants of Schu S4 were sensitive to hydrogen peroxide and reactive nitrogen species in vitro but retained lethality in mice, indicating that KatG is a sufficient but not necessary virulence factor.Citation29 Similarly, the superoxide dismutases SodB and SodC are essential for bacterial resistance to superoxide radicals, as F. tularensis LVS mutants of either enzyme are highly attenuated in mice challenged intranasally.Citation30,Citation31

Host–pathogen interactions

Bacterial survival and replication

F. tularensis has long been considered to be an intracellular pathogen. The first evidence that F. tularensis could replicate intracellularly came from studies of infected chick embryos and, later, HeLa cells.Citation32,Citation33 Notably, these early investigations showed that bacteria were present in the cytoplasm of infected cells.

Later work demonstrated that F. tularensis is also capable of replicating within macrophages. In 1995, Fortier et alCitation34 showed that F. tularensis LVS grew to high concentrations in the presence of peritoneal macrophages but not in the presence of lysed macrophages or in macrophage-conditioned media. F. tularensis is taken up by macrophages through a unique form of phagocytosis called “looping phagocytosis”, in which extensions of the cell membrane engulf a large volume of space surrounding the bacterium in an actin-dependent manner.Citation35,Citation36 The large space within the loop does not result from the presence of a bacterial capsule, as the volume of the vacuole is reduced rapidly after phagocytosis.Citation36 Entry into the cell can involve a variety of surface receptors, including mannose receptors, Fc receptors, and complement receptors.Citation35,Citation37Citation40 Both virulent and attenuated strains of F. tularensis survive phagocytosis by preventing acidification of the phagosome and arresting its maturation, and the organism ultimately escapes into the cytoplasm.Citation41Citation43 This process is summarized in . There is evidence that following escape of F. tularensis LVS into the cytoplasm, a proportion of cytoplasmic bacteria re-enters the endocytic pathway through autophagy.Citation44 It is not certain whether this process is an adaptive strategy by the bacterium or a part of the cellular defense against infection; however, evidence suggests that autophagy provides intracellular bacteria with nutrients required for replication.Citation45 The replication process ends in the destruction of the host cell. Lai et alCitation46 have shown that infection of the macrophage-like J774 cell line with LVS results in apoptosis. This apoptosis is not a self-sacrificing defense mechanism on the part of the cell, as apoptosis was not observed to reduce bacterial numbers.

Figure 1 Francisella tularensis (brown) binds to the cell surface using receptors such as the mannose receptor (1) or, in the case of opsonized bacteria, Fc receptors (2) or complement receptors. Bacteria enter the cell through looping phagocytosis (3) but survive by preventing maturation of the phagosome (4). F. tularensis escapes the phagosome to replicate in the cytoplasm (5), ultimately inducing apoptosis and escaping the cell (6). When macrophages are simulated by exposure to IFN-γ (7), they can restrict intracellular replication (8). Stimulation by IFN-γ and stimulation of Toll-like receptor 2 (9) can also lead to secretion of inflammatory cytokines and chemoattractants (10).

Abbreviations: IFN, interferon; TNF, tumor necrosis factor.

Figure 1 Francisella tularensis (brown) binds to the cell surface using receptors such as the mannose receptor (1) or, in the case of opsonized bacteria, Fc receptors (2) or complement receptors. Bacteria enter the cell through looping phagocytosis (3) but survive by preventing maturation of the phagosome (4). F. tularensis escapes the phagosome to replicate in the cytoplasm (5), ultimately inducing apoptosis and escaping the cell (6). When macrophages are simulated by exposure to IFN-γ (7), they can restrict intracellular replication (8). Stimulation by IFN-γ and stimulation of Toll-like receptor 2 (9) can also lead to secretion of inflammatory cytokines and chemoattractants (10).Abbreviations: IFN, interferon; TNF, tumor necrosis factor.

F. tularensis may also be able to use nonmacrophage cells as hosts. LVS has been observed to replicate within alveolar type II (ATII) epithelial cells in vitro, and experiments using green fluorescent protein-expressing F. tularensis of both type A and type B strains have detected labeled bacteria within neutrophils after intranasal infection.Citation47,Citation48 Experiments employing LVS and Schu S4 ΔpyrF mutants, which fail to replicate within primary macrophages, showed that such mutants were fully virulent in mice infected intranasally, suggesting that F. tularensis is not dependent upon replication within macrophages for survival and virulence.Citation49 Despite the proclivity of F. tularensis for intracellular replication, the majority of bloodborne bacteria of both type A and type B strains are extracellular.Citation50,Citation51

Iron is a key requirement for intracellular growth of F. tularensis.Citation34,Citation52,Citation53 Investigators have found that iron concentrations in growth media can affect expression of the Francisella pathogenicity island genes and can induce the organism to adopt a “host-adapted” phenotype.Citation18,Citation54 There is evidence that F. tularensis actively modulates expression of transferrin receptors in order to promote delivery of iron to the phagosome during the early stages of intracellular growth.Citation55 However, iron uptake may paradoxically render bacteria more susceptible to intracellular killing by ROS.Citation56 The highly virulent type A strain Schu S4 has been shown to have a lower iron content than the attenuated type B strain LVS, which may be a factor in the latter’s reduced virulence.Citation56

Pathogenesis

The earliest descriptions of tularemia included animals as varied as rodents, lagomorphs, and monkeys. These studies indicated that infection produced a febrile illness and generated foci of infection and necrotic inflammation in the lymph nodes as well as in dispersed organ sites.Citation1,Citation57 Morphological changes to the liver and spleen, particularly splenomegaly, have also been observed.Citation57,Citation58 Pneumonia is a frequent occurrence, even when the initial infection occurs through a route other than the respiratory route.Citation10,Citation59,Citation60 Following intravenous infection with F. tularensis LVS, infectious foci were detected in liver tissue after 16 hours, accompanied by infiltration of both neutrophils and monocytes.Citation61 Systemic dissemination also occurs following respiratory infection. Following aerosol infection with F. tularensis LVS, bacteria can be isolated from livers and spleens as early as 48 hours later.Citation62 In humans, infection is known to produce fever, persistent malaise, and ulcerous lesions or buboes.Citation2,Citation10,Citation59,Citation60

A major factor in F. tularensis pathogenicity is its capacity for intracellular replication. It has long been recognized that F. tularensis can replicate within host cells,Citation32,Citation33 and particularly macrophages.Citation46,Citation63,Citation64 This intracellular replication strategy contributes to pathogenesis as infected cells either apoptose or become lysed by uninfected immune cells.Citation46,Citation61 Intracellular replication may also play a role in dissemination of the infection to distant sites. In 2008, Bar-Haim et alCitation65 showed that following intranasal F. tularensis LVS infection, dendritic cells trafficked to the mediastinal lymph nodes, which quickly became severely infected.

Death in F. tularensis infection appears to result from widespread sepsis and inflammation rather than bacterial pneumonia, even when infection occurs through the respiratory route.Citation66Citation69 In a study of pneumonic F. tularensis LVS infection, high levels of inflammatory cytokines and chemokines in the lungs and spleen, including interleukin (IL)-6, macrophage inflammatory protein 2, and chemokine ligand 2, were correlated with mortality.Citation58 Investigations involving respiratory infection with F. tularensis Schu S4 and F. novicida showed that lethal infection was associated with hypercytokinemia and biochemical markers for sepsis.Citation66,Citation69 This sepsis may result from the fact that in F. tularensis infection, upregulation of cytokine production and inflammatory cell recruitment is delayed,Citation66 and the high degree of apoptotic cell debris associated with F. tularensis infection leads to alternative activation of macrophages, impeding bacterial clearance.Citation70 In lung infection, excessive levels of neutrophil recruitment may also contribute to pathology. Although neutrophil recruitment is delayed in Francisella infection,Citation66 high levels of neutrophil recruitment are associated with increased lung histopathology and, somewhat paradoxically, high bacterial burden.Citation71 These findings may be explained in part by observations that human neutrophils infected with F. tularensis LVS or Schu S4 exhibit a delay in time to apoptosis, preventing resolution of inflammation.Citation72

Innate immunity

The type 1-associated immune pathway is known to be an important factor in protection from F. tularensis infection. After cutaneous infection with F. tularensis LVS, immunized mice produced large quantities of IL-12 within 24 hours of infection.Citation73 Another study found that IL-12 p40-knockout mice failed to clear LVS administered intraperitoneally, even after immunization by sublethal intradermal infection.Citation74 These mice also had significantly lower levels of serum interferon (IFN)-γ, suggesting a connection between secretion of IL-12 and the IFN-γ response. The p40 subunit of IL-12 was also shown to induce migration of dendritic cells from the lungs to the draining lymph nodes after LVS infection.Citation75

After intradermal infection with LVS, IFN-γ messenger ribonucleic acid expression can be detected within 48 hours in naïve mice and within 24 hours in immunized mice.Citation73 In F. tularensis infection, both natural killer (NK) cells and T-cells are important producers of IFN-γ, although NK cells are the dominant source at early time points.Citation76,Citation77 Following intranasal F. tularensis LVS infection, NK cells are among the first cells recruited to the lungs, and can be observed to secrete IFN-γ within 72 hours of infection.Citation76,Citation78 NK cells primed with bacterial deoxyribonucleic acid can reduce replication of F. tularensis LVS in macrophages, even in the absence of T- or B-cells, and this effect is dependent upon IFN-γ and TNF-α.Citation79 IFN-γ is essential to host survival following pneumonic infection, as IFN-γ knockout mice challenged by the respiratory route exhibited greater rates of mortality and higher lung bacterial burden than wild-type (WT) mice.Citation80,Citation81 Although exogenous treatment with IL-12 was shown to be protective against intranasal infection, this protection was not evident in IFN-γ knockout mice, indicating that the protective effect of IL-12 is dependent upon IFN-γ expression.Citation81

Despite the capacity of F. tularensis to replicate intracellularly in vitro, it has been known for some time that IFN-γ-stimulated macrophages play an important role in defense against this infection. In 1992, Fortier et alCitation64 showed that exposure of macrophages to IFN-γ caused activation of these cells and allowed controlled replication of F. tularensis LVS in vitro. The authors attributed this to production of nitric oxide. A later paper by the same laboratory, however, showed that although IFN-γ-stimulated alveolar macrophages produced nitric oxide, their antimicrobial activity against F. tularensis was independent of nitric oxide production.Citation82 In other macrophage populations, reactive nitrogen species appear to take on greater importance. A 2005 study indicated that IFN-γ-stimulated peritoneal exudate cells secrete nitric oxide and control bacterial replication, but inducible nitric oxide synthase (iNOS) knockout mice or pharmacological inhibition of iNOS abrogated this bactericidal effect.Citation83 In contrast, knockout of p47, a component of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex, which induces the respiratory burst, reduced but did not eliminate bactericidal activity.Citation83 IFN-γ-treated alveolar macrophages produced large quantities of tumor necrosis factor (TNF)-α, but TNF-α is not essential for protection in vitro when IFN-γ is present. Alveolar macrophages treated with recombinant IFN-γ produced reactive nitrogen species and controlled LVS burden equally well in the presence or absence of neutralizing monoclonal antibodies against TNF-α.Citation82 In vivo, both IFN-γ and TNF-α were found to be essential for survival of primary intravenous infection in mice infected with F. tularensis LVS.Citation84 Neutralization of TNF-α or IFN-γ in vivo rendered naïve mice unable to control bacterial replication after F. tularensis LVS infection. Immunized mice fared better at low infectious doses but proved unable to control infection against an intravenous dose exceeding 106 CFU.Citation84

Macrophages can also respond to infection through means other than the production of nitric oxide or the respiratory burst. In 2007, it was found that infection of murine macrophages with F. novicida resulted in activation of the inflammasome in a manner dependent upon type I interferons.Citation85 Cole et alCitation86 found that macrophages detect F. tularensis LVS through both Toll-like receptor 2 (TLR2) and cytosolic signaling, resulting in secretion of IFN-β. TLR2 knockout mice were found to be impaired in their ability to facilitate TNF secretion from macrophages, although this effect did not specifically require a complex of TLR2 with either TLR1 or TLR6.Citation87 Although IFN-γ was found to restrict replication of the virulent type A strain Schu S4 within both murine bone marrow-derived macrophages and human blood monocyte-derived macrophages, this effect was found to be independent of either NADPH oxidase or iNOS.Citation88 The roles of IFN-γ and TLR2 are summarized in .

The role of neutrophils in F. tularensis infection is somewhat controversial and may be dependent upon the site of infection. Early research using the neutrophil-depleting monoclonal antibody RB6-8C5 indicated that neutrophils were essential for survival and control of bacterial replication when mice were infected intradermally or intravenously with F. tularensis LVS.Citation89 Intranasal infection resulted in recruitment of neutrophils to the lungs within 72 hours of infection.Citation90 However, the authors observed that cell depletion or increased recruitment of neutrophils to the lungs failed to affect either bacterial burden or time to death. Mice deficient in gp91phox were slightly more susceptible to low infectious doses, indicating a role for NADPH oxidase.Citation90 Some investigators have suggested that neutrophil recruitment results in harmful levels of inflammation after respiratory infection. Expression of matrix metalloprotease 9 was associated with production of the neutrophil chemoattractant tripeptide proline–glycine–proline and with increased neutrophil recruitment and severe histopathology after F. tularensis infection.Citation71 Knockout of IL-10 resulted in increased expression of IL-17, which led to increased neutrophil recruitment into the lungs after intranasal infection.Citation91 However, although IL-10 knockout mice were protected against cutaneous infection, they were more susceptible than WT mice to pneumonic infection.

Other cells present in the lung are also involved in F. tularensis infection. Following infection of mice with green fluorescent protein-expressing F. tularensis LVS and Schu S4, bacteria were detected by flow cytometry within ATII epithelial cells.Citation47,Citation48 However, Gentry et alCitation92 showed that human ATII cells in an in vitro transwell system responded to both LVS and Schu S4 with NF-κB-dependent upregulation of IL-8. Mast cells have been found to inhibit replication of F. tularensis LVS within mouse macrophages through direct contact and via secretion of IL-4.Citation93,Citation94

Cell-mediated immunity

As F. tularensis spends much of its existence within host cells, cell-mediated immunity likely plays a major role in control of tularemia. Early work indicated that splenocytes adoptively transferred from mice immunized with F. tularensis LVS could protect naïve mice from the virulent strain Schu S4.Citation95 However, this work failed to make a distinction between the potential roles of T-cells and B-cells. It was not until 1991 that experiments were conducted to determine the relative contributions of different splenocyte populations to adaptive immunity.Citation96 In those experiments, Fortier et alCitation96 inoculated splenocytes from LVS-immunized mice into naïve mice. Although immune splenocytes protected mice from an otherwise lethal intradermal LVS infection, depletion of T-cells removed the protective effect, implicating T-cells as a major component of the adaptive response to F. tularensis. In agreement with these results, vaccines designed to promote a cell-mediated adaptive immune response have had promising results. Mice vaccinated with Salmonella typhimurium, which was engineered to express the lipoprotein TUL4, had lower bacterial burdens after LVS challenge than mice vaccinated with WT S. typhimurium, even though the modified S. typhimurium produced a weaker adaptive response than LVS itself.Citation97 Although these mice generated antibody responses against TUL4, protection was abolished after cyclosporin A treatment to inhibit T-cell activation, suggesting that protection was predominantly cell mediated rather than humoral. Similarly, vaccination with immunostimulating complexes that were composed of micelles impregnated with TUL4 and the adjuvant Quil A, designed to promote T-cell responses, resulted in reduced bacterial burdens after LVS challenge.Citation98 Another study found that in mice vaccinated with F. tularensis LVS and rechallenged 90 days later with Schu S4, survival was correlated with activation of T-cells in the spleen.Citation99 As bacterial burden up to day 4 of Schu S4 infection was found to be significantly different between LVS-immunized and nonimmunized mice in the spleen, but not in the lungs, the authors concluded that cell-mediated immunity conferred protection against systemic dissemination of F. tularensis rather than against replication in the lungs. However, another study found that airway administration of IL-17A, but not intraperitoneal administration, delayed time to death following respiratory F. tularensis LVS infection.Citation100 The T helper 1 (Th1)-associated transcription factor T-bet has been reported to play an important role in control of F. tularensis LVS infection. However, although T-bet knockout T-cells from lungs were unable to control intracellular bacterial replication in macrophages, T-bet knockout splenocytes were not deficient in this capacity, suggesting different roles for these two T-cell populations.Citation101

Our understanding of the relative importance of different T-cell populations is still evolving. Cowley et alCitation102 showed that membrane-bound TNF-α expressed by CD8+ and CD4+ T-cells contributed to control of intracellular replication in macrophages but was essential only for protection mediated by CD8+ T-cells. It was further found that IL-23-mediated stimulation of Th17 cells promoted secretion of IL-17A, which contributed to the Th1 and IL-12 responses against F. tularensis LVS.Citation103 This pathway was essential in controlling lung bacterial burden after intratracheal infection. Other investigators have found that airway administration of IL-23 and IL-17A delayed time to death (although these treatments did not increase survival), and neutralization of these cytokines by monoclonal antibody increased mortality.Citation100 Cowley et alCitation104Citation106 identified another population of T-cells that controls intracellular replication of F. tularensis LVS through IFN-γ-dependent and -independent mechanisms, including production of IL-17A. These T-cells express CD3 and αβ T-cell receptors but do not express CD4, CD8, or NK cell markers.

Cell-mediated immunity can operate in synergy with humoral immunity to protect against F. tularensis infection. Although immune serum has been shown to protect against F. tularensis LVS infection, the protective effect of serum transfer is dependent upon both T-cells and IFN-γ.Citation96,Citation107 Mice vaccinated with F. tularensis lipopolysaccharide (LPS) can survive infection even in the absence of CD4+ or CD8+ T-cells, although depletion of CD8+ T-cells results in delayed clearance.Citation108 However, serum transfer from mice immunized with LPS and boosted with F. tularensis LVS failed to protect naïve mice depleted of either CD4+ or CD8+ T-cells.Citation108 A combination vaccine containing tetanus-toxin-conjugated O-polysaccharide (to generate an antibody response) and an LVS mutant lacking the O-polysaccharide (to generate a T-cell response) protected mice against intranasal and intradermal infection with the type A strain Schu S4 and the type B strain FSC 108 better than either vaccine component alone.Citation109 Humoral immunity may also enhance the T-cell response. It has been reported that immunoglobulin (Ig) A knockout mice recruited fewer IFN-γ+ T-cells to the lungs than WT mice 9 days after respiratory F. tularensis LVS infection.Citation77

Humoral immunity

The mouse antibody response to F. tularensis has been fairly well characterized. Mice infected with a sublethal infectious dose generate a robust antibody response characterized by specific IgG2 and IgM.Citation107 Rats generate a similar antibody profile against F. tularensis LVS.Citation110 Although antibodies are induced against a variety of bacterial antigens, a large proportion of the humoral response is directed against LPS in both mice and humans.Citation111Citation113

Much of the early information pertaining to F. tularensis immunity was obtained from studies of the antibody response and from treatment of infected individuals with immune animal serum. Due perhaps to the variable virulence of different strains of F. tularensis, much of these early data were contradictory. In one of the earliest studies, FoshayCitation60 compared recovery times of tularemia patients who received only symptomatic treatment or streptomycin with those of patients who received immune serum or hyperimmune serum generated by inoculation of ungulates with formalin-fixed F. tularensis or highly virulent live cultures, respectively. It was found that serum treatment, especially hyperimmune serum, reduced the time necessary for recovery. Although use of animal serum in humans often led to “serum sickness”, this study was one of the first to indicate that humoral immunity could confer at least limited benefit against F. tularensis. Thorpe and Marcus,Citation114 on the other hand, found that passive serum transfer conferred only minor protection against virulent strains.

Other investigators have obtained evidence that antibodies contribute to survival of F. tularensis infection, especially against challenge with F. tularensis LVS, although they are not necessarily sufficient to ensure survival. Fortier et alCitation96 made the intriguing observation that, while transfer of immune serum to naïve animals could protect against an otherwise lethal challenge with F. tularensis LVS, transfer of T-cell-depleted splenocytes failed to confer protection, suggesting a diminished role for B-cells. This work is partially contradicted by a report that showed higher bacterial burdens in the lungs, livers, and spleens of B-cell-knockout mice compared with WT mice after aerosol infection with F. tularensis LVS.Citation80 However, the authors also found no difference in mean time to death or liver pathology in B-cell-knockout mice versus WT mice after aerosol or intradermal infection.Citation80 Elkins et alCitation115 found that athymic nude mice challenged intradermally with a sublethal dose of F. tularensis LVS were able to survive subsequent intraperitoneal or intravenous infection with an inoculum of approximately 5,000 times the median lethal dose for nude mice. Priming with LVS did not protect against S. typhimurium, indicating that the protection was specific. As these mice were athymic, this protection was likely to be humoral in nature. Rhinehart-Jones et alCitation107 observed protection against F. tularensis LVS when serum from mice that had been infected intradermally with LVS was administered to naïve mice. These mice were also protected by transfer of LVS-specific IgG (but not IgM). Protection was found to be dependent upon host IFN-γ, and nude mice were not protected by serum transfer, implicating a role for IFN-γ-secreting T-cells in antibody-mediated clearance.Citation107 These results were corroborated by Sjöstedt et al,Citation84 who found that IFN-γ and TNF-α were required for protection against secondary intravenous infection with F. tularensis LVS, although the requirement for these cytokines was reduced in immunized mice. When naïve and immunized mice were treated with neutralizing monoclonal antibodies against these cytokines, immunized mice were able to tolerate a 50-fold larger infectious dose than naïve mice. Kirimanjeswara et alCitation116 confirmed that immune serum could protect against F. tularensis LVS in an IFN-γ-dependent manner, and further demonstrated that this protection was dependent upon FcγR expression on phagocytes. Interestingly, Kirimanjeswara et alCitation116 were able to show protection even when serum was administered up to 48 hours after intranasal infection, validating early 20th century attempts to use immune sera therapeutically.Citation60,Citation116 Transfer of immune serum or purified specific IgG has also been shown to protect rats against both F. tularensis LVS and Schu S4,Citation110 although rats have been reported to be more resistant to F. tularensis infection than mice.Citation113 Intriguingly, although targeting of F. tularensis Schu S4 to phagocyte Fc receptors by antibody opsonization enhanced phagocytosis, cellular entry using these receptors delayed bacterial replication and phagosomal escape and enhanced production of ROS.Citation40

Several candidate antigens have been investigated as possible targets for protective humoral immunity. F. tularensis LPS is poorly immunostimulatory in comparison with LPS from other Gram-negative bacteria;Citation117 however, much of the antibody response to F. tularensis is directed against LPS.Citation112,Citation113 Fulop et alCitation108 have reported that mice immunized with LPS were protected against intraperitoneal F. tularensis LVS infection, and that serum from LPS-immunized mice was sufficient to protect naïve mice against LVS challenge. However, LPS immunization was insufficient to protect against challenge with the virulent strain Schu S4.Citation108 Twine et alCitation118 investigated the humoral response of BALB/c and CH3/HeN mice to F. tularensis LVS. Both mouse strains were protected against Schu S4 challenge by immunization with LVS, but C57BL/6 and DBA/2 mice were not protected. The authors found that the former strains of mice generated antibodies against numerous bacterial antigens, such as protein chain elongation factor thermo stable and peroxidase, which did not elicit antibody responses from unprotected strains.

Although IgG and IgM are highly upregulated following F. tularensis infection,Citation107 there is evidence to suggest that IgA is also a critical component of host defense. Baron et alCitation78 showed that WT C57BL/6 mice could be protected against lethal F. tularensis LVS infection by intranasal vaccination with inactivated F. tularensis and IL-12 as an adjuvant. However, IgA knockout mice could not be protected in this fashion. IgA knockout mice generate IgG and IgM responses to F. tularensis LVS that are comparable with responses of WT mice but exhibit reduced IFN-γ responses, higher bacterial burden, and reduced survival following intranasal LVS infection.Citation77 The precise mechanisms responsible for IgA-mediated protection remain to be determined.

Evasion of innate and adaptive immunity

F. tularensis is highly adept at evading recognition and destruction by the host immune system, as evidenced by the considerable delay between infection and the onset of the inflammatory response.Citation66,Citation119 The pathogen employs multiple survival strategies, including intracellular replication, expression of an atypical LPS, and induction of alternative and aberrant activation of the immune response.

As has been described, F. tularensis is capable of replicating within host cells, including macrophages.Citation32,Citation33,Citation64 In the absence of opsonizing conditions (eg, when F. tularensis uses the mannose receptor to facilitate cell entry), F. tularensis rapidly leaves the phagosome and enters the cytoplasm, where replication occurs.Citation40,Citation41 Although some bacteria remain extracellular throughout infection,Citation50,Citation51 cytosolic replication may provide some protection against detection by TLRs and other surface receptors, and from clearance by stimulated phagocytes. However, entry into the cytosol allows activation of other cellular defenses such as the inflammasome.Citation85

Although the F. tularensis LPS has been used successfully to vaccinate mice against intraperitoneal infection with F. tularensis LVS,Citation120,Citation121 it is well known that the LPS of F. tularensis is poorly immunogenic and, in particular, is a poor stimulator of TLR4.Citation117,Citation122 This low level of TLR4 recognition has been ascribed to the unusual structure of the lipid A component.Citation123 Whereas the highly immunogenic lipid A of many Gram-negative species is hexa-acylated with short acyl chains, the lipid A of F. tularensis is tetra-acylated with long acyl chains.Citation117,Citation123,Citation124 F. tularensis LVS has also been reported to alter expression of its carbohydrate capsule, which incorporates polymers of LPS O-antigen, in order to evade recognition by host immunity.Citation125

F. tularensis also employs several active measures to inhibit or redirect the host immune response. In 2003, Telepnev et alCitation126 found that infection of the mouse macrophage J774A.1 cell line with F. tularensis LVS resulted in the cells becoming unresponsive to Escherichia coli LPS. Infected cells failed to degrade the NF-κB inhibitor IκB and secreted less TNF-α and IL-1β. These findings were corroborated in 2005 when Bosio and DowCitation127 reported that LVS-infected bone marrow-derived macrophages and dendritic cells, and dendritic cells isolated from the airways of F. tularensis LVS-infected mice, failed to secrete increased quantities of TNF-α and IL-6 but instead secreted increased quantities of the immunosuppressive cytokine TGF-β. Similar observations were reported in mice infected via aerosol with type A strain Schu S4.Citation128 These immunosuppressive effects have been observed in human cells. Telepnev et alCitation129 have reported that the THP-1 human macrophage cell line and human peripheral blood monocytes infected with F. tularensis LVS displayed a brief period of stimulation, including NF-κB activation and TNF-α secretion, followed by inactivation of the NF-κB response and suppression of cytokine secretion within 5 hours. Interestingly, F. tularensis LVS mutated in the gene iglC, a Francisella pathogenicity island gene that encodes a 23 kDa protein produced during infection of macrophages, was unable to suppress immune activation.Citation19,Citation24,Citation126,Citation129,Citation130

In addition to downregulating inflammatory cytokine secretion, F. tularensis also upregulates anti-inflammatory cytokines and promotes alternative activation of immune cells. Murine macrophages and dendritic cells infected with F. tularensis LVS exhibit alternative activation, characterized by increased expression of arginase-1, IL-4, IL-13, and TGF-β.Citation127,Citation131 F. tularensis LVS also induces secretion of anti-inflammatory PGE2 from bone marrow-derived mouse macrophages, although F. novicida mutants that do not induce PGE2 secretion do not exhibit a defect in intracellular replication.Citation26,Citation132,Citation133 PGE2 elicited by Francisella infection was found to downregulate major histocompatibility complex class II through an intermediary host factor dubbed FTMΦSN (F. tularensis macrophage supernatant), which drives production of IL-10.Citation134,Citation135 Predictably, Schu S4 has also been found to suppress secretion of inflammatory cytokines by infected dendritic cells; moreover, Schu S4 infection of dendritic cells also blunted the inflammatory capacity of nearby uninfected cells.Citation136 Interestingly, human dendritic cells infected with Schu S4 were found not to secrete TGF-β, which was reported to be upregulated in murine cells infected with LVS.Citation136 It has been suggested that alternative activation of macrophages results in part from the abundance of cell debris found in Francisella-infected lungs. F. novicida-infected J774 cells exposed to large quantities of necrotic cell debris produced high levels of arginase-1 and showed reduced capacity for efferocytosis, which would lead to further accumulation of cell debris in vivo.Citation70 The reduced secretion of inflammatory cytokines and increased secretion of anti-inflammatory cytokines ultimately result in a delay in the recruitment of inflammatory cells to the lungs following pulmonary infection.Citation48,Citation66

F. tularensis is able to suppress the immune response in granulocytes as well as macrophages and dendritic cells. A 2006 report indicated that F. tularensis LVS phagocytized by neutrophils inhibits assembly of the NADPH oxidase complex and therefore the respiratory burst, even when the neutrophils receive additional stimulation in the form of phorbol 12-myristate 13-acetate.Citation137 Later studies showed that F. tularensis LVS, as well as virulent type A and type B strains, can inhibit NADPH oxidase function postassembly, and that inhibition is dependent upon fevR, a regulator of the Francisella pathogenicity island.Citation138,Citation139

The signaling mechanisms by which F. tularensis modulates the immune response remain poorly understood. It has been reported that F. novicida inhibits cellular responses to IFN-γ by interfering with STAT-1 signaling.Citation140 Nallaparaju et alCitation141 have found that the Francisella outer membrane protein C reduces IFN-γ signaling in infected macrophages, preventing production of nitric oxide. It has also been shown that lipid products of F. tularensis Schu S4 but not LVS can inhibit IL-12 p40 secretion by inhibiting translocation of NF-κB, IRF1, and IRF8.Citation142

Various factors involved in immunosuppression by, and virulence of, F. tularensis appear to be upregulated as a specific response to the host environment. The Francisella pathogenicity island gene iglC, which is necessary for immunosuppression, is upregulated during replication in macrophages but not during replication in Chamberlain medium.Citation126,Citation130 Similarly, it has been observed that bacteria grown in Mueller–Hinton medium, which contains concentrations of amino acids that exceed those available to F. tularensis during infection, expressed increased levels of SodB and MglB and reduced levels of IglB, IglC, and KatG and longer, more capsule-like O-antigen than bacteria grown in macrophages or in the more physiologically similar Brain Heart Infusion medium.Citation54,Citation125 These differences may explain why F. tularensis LVS grown on Thayer–Martin agar or in Mueller–Hinton medium is initially immunostimulatory before adopting an immunosuppressive phenotype, as the bacteria acclimate to the cellular environment.Citation129,Citation143

Conclusion

F. tularensis has been known to science for over a century and has been the subject of an impressive body of research over that time – research that has intensified and accelerated as a result of recently increased interest in biodefense.Citation1,Citation14 A considerable breadth of information is available on the interaction of this pathogen with mammalian hosts. In particular, it is well established that F. tularensis maintains a low immunological profile during early infection, evading surveillance and replicating within the relatively protective environment of the host–cell cytoplasm before accumulated cell debris and sheer bacterial load make an intense immune response unavoidable. However, despite decades of work, we have yet to develop a safe, effective, and well-characterized vaccine that protects against respiratory infection involving highly virulent type A strains. Key to the development of such a vaccine will be research on the host response to F. tularensis type A strains at mucosal surfaces – a field that is gaining momentum. Ultimately, defining the differences between host responses to virulent and attenuated strains of F. tularensis may shed important new insights into development of effective prophylactic and therapeutic treatments.

Acknowledgments

This work was supported by US National Institutes of Health (Grant PO1 AI056320).

Disclosure

The authors report no conflicts of interest in this work.

References

  • McCoyGA plague-like disease of rodentsPublic Health Bulletin1911435372
  • WherryWBLambBHInfection of man with Bacterium tularenseJ Infect Dis191418971321132915108712
  • McCrumbFJrSnyderMJWoodwardTEStudies on human infection with Pasteurella tularensis: comparison of streptomycin and chloramphenicol in the prophylaxis of clinical diseaseTrans Assoc Am Physicians1957707479 discussion 79–8013496116
  • McCoyGWChapinCWFurther observations on a plague-like disease of rodents with a preliminary note on the causative agent, Bacterium tularenseJ Infect Dis19121016172
  • HesselbrockWFoshayLThe morphology of Bacterium tularenseJ Bacteriol194549320923116560913
  • EllisJOystonPCGreenMTitballRWTularemiaClin Microbiol Rev200215463164612364373
  • TitballRWJohanssonAForsmanMWill the enigma of Francisella tularensis virulence soon be solved?Trends Microbiol200311311812312648943
  • McLendonMKApicellaMAAllenLAFrancisella tularensis: taxonomy, genetics, and immunopathogenesis of a potential agent of biowarfareAnnu Rev Microbiol20066016718516704343
  • GoethertHKTelfordSR3rdDifferential mortality of dog tick vectors due to infection by diverse Francisella tularensis tularensis genotypesVector Borne Zoonotic Dis20111191263126821612530
  • McCrumbFRAerosol infection of man with Pasteurella tularensisBacteriol Rev196125326226716350172
  • CoxCSAerosol survival of Pasteurella tularensis disseminated from the wet and dry statesAppl Microbiol19712134824864994903
  • HarrisSJapanese biological warfare research on humans: a case study of microbiology and ethicsAnn N Y Acad Sci199266621521297279
  • DennisDTInglesbyTVHendersonDATularemia as a biological weapon: medical and public health managementJAMA2001285212763277311386933
  • OystonPCSjöstedtATitballRWTularaemia: bioterrorism defence renews interest in Francisella tularensisNat Rev Microbiol200421296797815550942
  • EigelsbachHTDownsCMProphylactic effectiveness of live and killed tularemia vaccines. I. Production of vaccine and evaluation in the white mouse and guinea pigJ Immunol19618741542513889609
  • TigerttWDSoviet viable Pasteurella tularensis vaccines. A review of selected articlesBacteriol Rev19622635437313985026
  • SalomonssonEKuoppaKForslundALReintroduction of two deleted virulence loci restores full virulence to the live vaccine strain of Francisella tularensisInfect Immun20097783424343119506014
  • NanoFESchmerkCThe Francisella pathogenicity islandAnn N Y Acad Sci2007110512213717395722
  • NanoFEZhangNCowleySCA Francisella tularensis pathogenicity island required for intramacrophage growthJ Bacteriol2004186196430643615375123
  • ChouAYKennettNJNixEBSchmerkCLNanoFEElkinsKLGeneration of protection against Francisella novicida in mice depends on the pathogenicity protein pdpA, but not pdpC or pdpDMicrobes Infect2013151281682723880085
  • de BruinOMDuplantisBNLuduJSThe biochemical properties of the Francisella pathogenicity island (FPI)-encoded proteins IglA, IglB, IglC, pdpB and dotU suggest roles in type VI secretionMicrobiology2011157Pt 123483349121980115
  • BromsJEMeyerLLavanderMLarssonPSjöstedtADotU and vgrG, core components of type VI secretion systems, are essential for Francisella LVS pathogenicityPLoS One201274e3463922514651
  • BarkerJRChongAWehrlyTDThe Francisella tularensis pathogenicity island encodes a secretion system that is required for phagosome escape and virulenceMol Microbiol20097461459147020054881
  • SanticMMolmeretMKloseKEJonesSKwaikYAThe Francisella tularensis pathogenicity island protein IglC and its regulator MglA are essential for modulating phagosome biogenesis and subsequent bacterial escape into the cytoplasmCell Microbiol20057796997915953029
  • DaiSMohapatraNPSchlesingerLSGunnJSRegulation of Francisella tularensis virulenceFront Microbiol2010114421687801
  • WoolardMDBarriganLMFullerJRIdentification of Francisella novicida mutants that fail to induce prostaglandin E(2) synthesis by infected macrophagesFront Microbiol201341623403609
  • BromsJEMeyerLSunKLavanderMSjöstedtAUnique substrates secreted by the type VI secretion system of Francisella tularensis during intramacrophage infectionPLoS One2012711e5047323185631
  • SmulevichGJakopitschCDroghettiEObingerCProbing the structure and bifunctionality of catalase-peroxidase (KatG)J Inorg Biochem2006100456858516516299
  • LindgrenHShenHZingmarkCGolovliovIConlanWSjöstedtAResistance of Francisella tularensis strains against reactive nitrogen and oxygen species with special reference to the role of KatGInfect Immun20077531303130917210667
  • BakshiCSMalikMReganKSuperoxide dismutase B gene (sodB)-deficient mutants of Francisella tularensis demonstrate hypersensitivity to oxidative stress and attenuated virulenceJ Bacteriol2006188176443644816923916
  • MelilloAAMahawarMSellatiTJIdentification of Francisella tularensis live vaccine strain CuZn superoxide dismutase as critical for resistance to extracellularly generated reactive oxygen speciesJ Bacteriol2009191206447645619684141
  • BuddinghGJWomackFCObservations on the infection of chick embryos with Bacterium tularense, brucella, and Pasteurella pestisJ Exp Med194174321322219871129
  • ShepardCCNonacid-fast bacteria and HeLa cells: their uptake and subsequent intracellular growthJ Bacteriol195977670171413664649
  • FortierAHLeibyDANarayananRBGrowth of Francisella tularensis LVS in macrophages: the acidic intracellular compartment provides essential iron required for growthInfect Immun1995634147814837890413
  • ClemensDLLeeBYHorwitzMAFrancisella tularensis enters macrophages via a novel process involving pseudopod loopsInfect Immun20057395892590216113308
  • ClemensDLHorwitzMAUptake and intracellular fate of Francisella tularensis in human macrophagesAnn N Y Acad Sci2007110516018617435118
  • PieriniLMUptake of serum-opsonized Francisella tularensis by macrophages can be mediated by class A scavenger receptorsCell Microbiol2006881361137016882038
  • BalagopalAMacFarlaneASMohapatraNSoniSGunnJSSchlesingerLSCharacterization of the receptor-ligand pathways important for entry and survival of Francisella tularensis in human macrophagesInfect Immun20067495114512516926403
  • SchulertGSAllenLADifferential infection of mononuclear phagocytes by Francisella tularensis: role of the macrophage mannose receptorJ Leukoc Biol200680356357116816147
  • GeierHCelliJPhagocytic receptors dictate phagosomal escape and intracellular proliferation of Francisella tularensisInfect Immun20117962204221421422184
  • ClemensDLLeeBYHorwitzMAVirulent and avirulent strains of Francisella tularensis prevent acidification and maturation of their phagosomes and escape into the cytoplasm in human macrophagesInfect Immun20047263204321715155622
  • SjöstedtAIntracellular survival mechanisms of Francisella tularensis, a stealth pathogenMicrobes Infect20068256156716239121
  • AsareRAbu KwaikYMolecular complexity orchestrates modulation of phagosome biogenesis and escape to the cytosol of macrophages by Francisella tularensisEnviron Microbiol20101292559258620482590
  • ChecrounCWehrlyTDFischerERHayesSFCelliJAutophagy-mediated reentry of Francisella tularensis into the endocytic compartment after cytoplasmic replicationProc Natl Acad Sci U S A200610339145781458316983090
  • SteeleSBruntonJZiehrBTaft-BenzSMoormanNKawulaTFrancisella tularensis harvests nutrients derived via ATG5-independent autophagy to support intracellular growthPLoS Pathog201398e100356223966861
  • LaiXHGolovliovISjöstedtAFrancisella tularensis induces cytopathogenicity and apoptosis in murine macrophages via a mechanism that requires intracellular bacterial multiplicationInfect Immun20016974691469411402018
  • HallJDCravenRRFullerJRPicklesRJKawulaTHFrancisella tularensis replicates within alveolar type II epithelial cells in vitro and in vivo following inhalationInfect Immun20077521034103917088343
  • HallJDWoolardMDGunnBMInfected-host-cell repertoire and cellular response in the lung following inhalation of Francisella tularensis Schu S4, LVS, or U112Infect Immun200876125843585218852251
  • HorzempaJO’DeeDMShanksRMNauGJFrancisella tularensis DeltapyrF mutants show that replication in nonmacrophages is sufficient for pathogenesis in vivoInfect Immun20107862607261920385757
  • ForestalCAMalikMCatlettSVFrancisella tularensis has a significant extracellular phase in infected miceJ Infect Dis2007196113413717538893
  • YuJJRaulieEKMurthyAKGuentzelMNKloseKEArulanandamBPThe presence of infectious extracellular Francisella tularensis subsp. novicida in murine plasma after pulmonary challengeEur J Clin Microbiol Infect Dis200827432332518087734
  • LindgrenHHonnMGolovlevIKadzhaevKConlanWSjöstedtAThe 58-kilodalton major virulence factor of Francisella tularensis is required for efficient utilization of ironInfect Immun200977104429443619651867
  • OlakanmiOGunnJSSuSSoniSHassettDJBritiganBEGallium disrupts iron uptake by intracellular and extracellular Francisella strains and exhibits therapeutic efficacy in a murine pulmonary infection modelAntimicrob Agents Chemother201054124425319917753
  • HazlettKRCaldonSDMcArthurDGAdaptation of Francisella tularensis to the mammalian environment is governed by cues which can be mimicked in vitroInfect Immun200876104479448818644878
  • PanXTamilselvamBHansenEJDaeflerSModulation of iron homeostasis in macrophages by bacterial intracellular pathogensBMC Microbiol2010106420184753
  • LindgrenHHonnMSalomonssonEKuoppaKForsbergASjöstedtAIron content differs between Francisella tularensis subspecies tularensis and subspecies holarctica strains and correlates to their susceptibility to H(2)O(2)-induced killingInfect Immun20117931218122421189323
  • CouncilmanWStrongRPlague-like infections in rodentsTrans Assoc Am Phys192136135143
  • ChiavoliniDAlroyJKingCAIdentification of immunologic and pathologic parameters of death versus survival in respiratory tularemiaInfect Immun200876248649618025095
  • FoshayLHesselbrockWHWittenbergHJRodenbergAHVaccine prophylaxis against tularemia in manAm J Public Health Nations Health194232101131114518015689
  • FoshayLA comparative study of the treatment of tularemia with immune serum, hyperimmune serum and streptomycinAm J Med1946118018820993803
  • ConlanJWNorthRJEarly pathogenesis of infection in the liver with the facultative intracellular bacteria Listeria monocytogenes, Francisella tularensis, and Salmonella typhimurium involves lysis of infected hepatocytes by leukocytesInfect Immun19926012516451711452350
  • ConlanJWKuoLeeRShenHWebbADifferent host defences are required to protect mice from primary systemic vs pulmonary infection with the facultative intracellular bacterial pathogen, Francisella tularensis LVSMicrob Pathog200232312713411855943
  • AnthonyLDBurkeRDNanoFEGrowth of Francisella spp. in rodent macrophagesInfect Immun1991599329132961879943
  • FortierAHPolsinelliTGreenSJNacyCAActivation of macrophages for destruction of Francisella tularensis: identification of cytokines, effector cells, and effector moleculesInfect Immun19926038178251541555
  • Bar-HaimEGatOMarkelGCohenHShaffermanAVelanBInterrelationship between dendritic cell trafficking and Francisella tularensis dissemination following airway infectionPLoS Pathog2008411e100021119023422
  • MaresCAOjedaSSMorrisEGLiQTealeJMInitial delay in the immune response to Francisella tularensis is followed by hypercytokinemia characteristic of severe sepsis and correlating with upregulation and release of damage-associated molecular patternsInfect Immun20087673001301018411294
  • SharmaJLiQMishraBBPenaCTealeJMLethal pulmonary infection with Francisella novicida is associated with severe sepsisJ Leukoc Biol200986349150419401387
  • CowleySCEditorial: proinflammatory cytokines in pneumonic tularemia: too much too late?J Leukoc Biol200986346947019720615
  • SharmaJMaresCALiQMorrisEGTealeJMFeatures of sepsis caused by pulmonary infection with Francisella tularensis type A strainMicrob Pathog2011511–2394721440052
  • MaresCASharmaJLiQDefect in efferocytosis leads to alternative activation of macrophages in Francisella infectionsImmunol Cell Biol201189216717220585334
  • MalikMBakshiCSMcCabeKMatrix metalloproteinase 9 activity enhances host susceptibility to pulmonary infection with type A and B strains of Francisella tularensisJ Immunol200717821013102017202364
  • SchwartzJTBarkerJHKaufmanJFayramDCMcCrackenJMAllenLAFrancisella tularensis inhibits the intrinsic and extrinsic pathways to delay constitutive apoptosis and prolong human neutrophil lifespanJ Immunol201218873351336322357630
  • StenmarkSSunnemarkDBuchtASjöstedtARapid local expression of interleukin-12, tumor necrosis factor alpha, and gamma interferon after cutaneous Francisella tularensis infection in tularemia-immune miceInfect Immun19996741789179710085019
  • ElkinsKLCooperAColombiniSMCowleySCKiefferTLIn vivo clearance of an intracellular bacterium, Francisella tularensis LVS, is dependent on the p40 subunit of interleukin-12 (IL-12) but not on IL-12 p70Infect Immun20027041936194811895957
  • SlightSRLinYMessmerMKhaderSAFrancisella tularensis LVS-induced Interleukin-12 p40 cytokine production mediates dendritic cell migration through IL-12 receptor beta1Cytokine201155337237921669537
  • LopezMCDuckettNSBaronSDMetzgerDWEarly activation of NK cells after lung infection with the intracellular bacterium, Francisella tularensis LVSCell Immunol20042321–2758515922718
  • FuruyaYKirimanjeswaraGSRobertsSMetzgerDWIncreased susceptibility of IgA-deficient mice to pulmonary Francisella tularensis live vaccine strain infectionInfect Immun20138193434344123836815
  • BaronSDSinghRMetzgerDWInactivated Francisella tularensis live vaccine strain protects against respiratory tularemia by intranasal vaccination in an immunoglobulin A-dependent fashionInfect Immun20077552152216217296747
  • ElkinsKLColombiniSMKriegAMDe PascalisRNK cells activated in vivo by bacterial DNA control the intracellular growth of Francisella tularensis LVS. Microbes InfectJan20091114956
  • ChenWKuoLeeRShenHConlanJWSusceptibility of immunodeficient mice to aerosol and systemic infection with virulent strains of Francisella tularensisMicrob Pathog200436631131815120157
  • DuckettNSOlmosSDurrantDMMetzgerDWIntranasal interleukin-12 treatment for protection against respiratory infection with the Francisella tularensis live vaccine strainInfect Immun20057342306231115784575
  • PolsinelliTMeltzerMSFortierAHNitric oxide-independent killing of Francisella tularensis by IFN-gamma-stimulated murine alveolar macrophagesJ Immunol19941533123812458027551
  • LindgrenHStenmanLTarnvikASjöstedtAThe contribution of reactive nitrogen and oxygen species to the killing of Francisella tularensis LVS by murine macrophagesMicrobes Infect20057346747515788155
  • SjöstedtANorthRJConlanJWThe requirement of tumour necrosis factor-alpha and interferon-gamma for the expression of protective immunity to secondary murine tularaemia depends on the size of the challenge inoculumMicrobiology1996142Pt 6136913748704976
  • HenryTBrotckeAWeissDSThompsonLJMonackDMType I interferon signaling is required for activation of the inflammasome during Francisella infectionJ Exp Med2007204598799417452523
  • ColeLESantiagoABarryEMacrophage proinflammatory response to Francisella tularensis live vaccine strain requires coordination of multiple signaling pathwaysJ Immunol2008180106885689118453609
  • AbplanalpALMorrisIRParidaBKTealeJMBertonMTTLR-dependent control of Francisella tularensis infection and host inflammatory responsesPLoS One2009411e792019936231
  • EdwardsJARockx-BrouwerDNairVCelliJRestricted cytosolic growth of Francisella tularensis subsp. tularensis by IFN-gamma activation of macrophagesMicrobiology2010156Pt 232733919926654
  • SjöstedtAConlanJWNorthRJNeutrophils are critical for host defense against primary infection with the facultative intracellular bacterium Francisella tularensis in mice and participate in defense against reinfectionInfect Immun1994627277927838005668
  • KuoLeeRHarrisGConlanJWChenWRole of neutrophils and NADPH phagocyte oxidase in host defense against respiratory infection with virulent Francisella tularensis in miceMicrobes Infect201113544745621277990
  • MetzgerDWSalmonSLKirimanjeswaraGDiffering effects of IL-10 in cutaneous and pulmonary Francisella tularensis LVS infectionInfect Immun20138162022202723529615
  • GentryMTaorminaJPylesRBRole of primary human alveolar epithelial cells in host defense against Francisella tularensis infectionInfect Immun20077583969397817502386
  • KetavarapuJMRodriguezARYuJJMast cells inhibit intramacrophage Francisella tularensis replication via contact and secreted products including IL-4Proc Natl Acad Sci U S A2008105279313931818591675
  • RodriguezARYuJJGuentzelMNMast cell TLR2 signaling is crucial for effective killing of Francisella tularensisJ Immunol2012188115604561122529298
  • EigelsbachHTHunterDHJanssenWADangerfieldHGRabinowitzSGMurine model for study of cell-mediated immunity: protection against death from fully virulent Francisella tularensis infectionInfect Immun197512599910051193736
  • FortierAHSlayterMVZiembaRMeltzerMSNacyCALive vaccine strain of Francisella tularensis: infection and immunity in miceInfect Immun1991599292229281879918
  • SjöstedtASandstromGTarnvikAHumoral and cell-mediated immunity in mice to a 17-kilodalton lipoprotein of Francisella tularensis expressed by Salmonella typhimuriumInfect Immun1992607285528621612751
  • GolovliovIEricssonMAkerblomLSandstromGTarnvikASjöstedtAAdjuvanticity of ISCOMs incorporating a T cell-reactive lipoprotein of the facultative intracellular pathogen Francisella tularensisVaccine19951332612677631511
  • AndersonRVCraneDDBosioCMLong lived protection against pneumonic tularemia is correlated with cellular immunity in peripheral, not pulmonary, organsVaccine201028406562657220688042
  • MarkelGBar-HaimEZahavyEThe involvement of IL-17A in the murine response to sub-lethal inhalational infection with Francisella tularensisPLoS One201056e1117620585449
  • MelilloAAForemanOBosioCMElkinsKLT-bet regulates immunity to F. tularensis LVS infection, particularly in lungsInfect Immun20148241477149024421047
  • CowleySCSedgwickJDElkinsKLDifferential requirements by CD4+ and CD8+ T cells for soluble and membrane TNF in control of Francisella tularensis live vaccine strain intramacrophage growthJ Immunol2007179117709771918025217
  • LinYRitcheaSLogarAInterleukin-17 is required for T helper 1 cell immunity and host resistance to the intracellular pathogen Francisella tularensisImmunity200931579981019853481
  • CowleySCElkinsKLMultiple T cell subsets control Francisella tularensis LVS intracellular growth without stimulation through macrophage interferon gamma receptorsJ Exp Med2003198337938912885873
  • CowleySCHamiltonEFrelingerJASuJFormanJElkinsKLCD4-CD8- T cells control intracellular bacterial infections both in vitro and in vivoJ Exp Med2005202230931916027239
  • CowleySCMeierovicsAIFrelingerJAIwakuraYElkinsKLLung CD4-CD8- double-negative T cells are prominent producers of IL-17A and IFN-gamma during primary respiratory murine infection with Francisella tularensis live vaccine strainJ Immunol2010184105791580120393138
  • Rhinehart-JonesTRFortierAHElkinsKLTransfer of immunity against lethal murine Francisella infection by specific antibody depends on host gamma interferon and T cellsInfect Immun1994628312931378039881
  • FulopMMastroeniPGreenMTitballRWRole of antibody to lipopolysaccharide in protection against low- and high-virulence strains of Francisella tularensisVaccine200119314465447211483272
  • SebastianSPinkhamJTLynchJGCellular and humoral immunity are synergistic in protection against types A and B Francisella tularensisVaccine200927459760519022323
  • Mara-KooshamGHuttJALyonsCRWuTHAntibodies contribute to effective vaccination against respiratory infection by type A Francisella tularensis strainsInfect Immun20117941770177821282410
  • Porsch-OzcurumezMKischelNPriebeHSplettstosserWFinkeEJGrunowRComparison of enzyme-linked immunosorbent assay, Western blotting, microagglutination, indirect immunofluorescence assay, and flow cytometry for serological diagnosis of tularemiaClin Diagn Lab Immunol20041161008101515539498
  • ConlanJOystonPCVaccines against Francisella tularensisAnn N Y Acad Sci2007110532535017395730
  • CowleySCElkinsKLImmunity to FrancisellaFront Microbiol201122621687418
  • ThorpeBDMarcusSPhagocytosis and intracellular fate of Pasteurella tularensis. 3. In vivo studies with passively transferred cells and seraJ Immunol19659457858514299033
  • ElkinsKLRhinehart-JonesTNacyCAWinegarRKFortierAHT-cell-independent resistance to infection and generation of immunity to Francisella tularensisInfect Immun19936138238298432603
  • KirimanjeswaraGSGoldenJMBakshiCSMetzgerDWProphylactic and therapeutic use of antibodies for protection against respiratory infection with Francisella tularensisJ Immunol2007179153253917579074
  • HajjarAMHarveyMDShafferSALack of in vitro and in vivo recognition of Francisella tularensis subspecies lipopolysaccharide by Toll-like receptorsInfect Immun200674126730673816982824
  • TwineSMPetitMDShenHMykytczukNCKellyJFConlanJWImmunoproteomic analysis of the murine antibody response to successful and failed immunization with live anti-Francisella vaccinesBiochem Biophys Res Commun20063463999100816781667
  • AsareRKwaikYAExploitation of host cell biology and evasion of immunity by Francisella tularensisFront Microbiol2010114521687747
  • FulopMMancheeRTitballRRole of lipopolysaccharide and a major outer membrane protein from Francisella tularensis in the induction of immunity against tularemiaVaccine19951313122012258578807
  • ConlanJWShenHWebbAPerryMBMice vaccinated with the O-antigen of Francisella tularensis LVS lipopolysaccharide conjugated to bovine serum albumin develop varying degrees of protective immunity against systemic or aerosol challenge with virulent type A and type B strains of the pathogenVaccine20022029–303465347112297391
  • AncutaPPedronTGirardRSandstromGChabyRInability of the Francisella tularensis lipopolysaccharide to mimic or to antagonize the induction of cell activation by endotoxinsInfect Immun1996646204120468675305
  • OkanNAKasperDLThe atypical lipopolysaccharide of FrancisellaCarbohydr Res2013378798323916469
  • VinogradovEPerryMBConlanJWStructural analysis of Francisella tularensis lipopolysaccharideEur J Biochem2002269246112611812473106
  • ZarrellaTMSinghABitsaktsisCHost-adaptation of Francisella tularensis alters the bacterium’s surface-carbohydrates to hinder effectors of innate and adaptive immunityPLoS One201167e2233521799828
  • TelepnevMGolovliovIGrundstromTTarnvikASjöstedtAFrancisella tularensis inhibits Toll-like receptor-mediated activation of intracellular signalling and secretion of TNF-alpha and IL-1 from murine macrophagesCell Microbiol200351415112542469
  • BosioCMDowSWFrancisella tularensis induces aberrant activation of pulmonary dendritic cellsJ Immunol2005175106792680116272336
  • BosioCMBielefeldt-OhmannHBelisleJTActive suppression of the pulmonary immune response by Francisella tularensis Schu4J Immunol200717874538454717372012
  • TelepnevMGolovliovISjöstedtAFrancisella tularensis LVS initially activates but subsequently down-regulates intracellular signaling and cytokine secretion in mouse monocytic and human peripheral blood mononuclear cellsMicrob Pathog2005385–623924715925273
  • GolovliovIEricssonMSandstromGTarnvikASjöstedtAIdentification of proteins of Francisella tularensis induced during growth in macrophages and cloning of the gene encoding a prominently induced 23-kilodalton proteinInfect Immun1997656218321899169749
  • ShireyKAColeLEKeeganADVogelSNFrancisella tularensis live vaccine strain induces macrophage alternative activation as a survival mechanismJ Immunol200818164159416718768873
  • WoolardMDWilsonJEHensleyLLFrancisella tularensis-infected macrophages release prostaglandin E2 that blocks T cell proliferation and promotes a Th2-like responseJ Immunol200717842065207417277110
  • WoolardMDHensleyLLKawulaTHFrelingerJARespiratory Francisella tularensis live vaccine strain infection induces Th17 cells and prostaglandin E2, which inhibits generation of gamma interferon-positive T cellsInfect Immun20087662651265918391003
  • WilsonJEKatkereBDrakeJRFrancisella tularensis induces ubiquitin-dependent major histocompatibility complex class II degradation in activated macrophagesInfect Immun200977114953496519703975
  • HuntDWilsonJEWeihKAFrancisella tularensis elicits IL-10 via a PGE(2)-inducible factor, to drive macrophage MARCH1 expression and class II down-regulationPLoS One201275e3733022615981
  • ChaseJCCelliJBosioCMDirect and indirect impairment of human dendritic cell function by virulent Francisella tularensis Schu S4Infect Immun200977118019518981246
  • McCaffreyRLAllenLAFrancisella tularensis LVS evades killing by human neutrophils via inhibition of the respiratory burst and phagosome escapeJ Leukoc Biol20068061224123016908516
  • BrotckeAMonackDMIdentification of fevR, a novel regulator of virulence gene expression in Francisella novicidaInfect Immun20087683473348018559431
  • McCaffreyRLSchwartzJTLindemannSRMultiple mechanisms of NADPH oxidase inhibition by type A and type B Francisella tularensisJ Leukoc Biol201088479180520610796
  • RothKMGunnJSLafuseWSatoskarARFrancisella inhibits STAT1-mediated signaling in macrophages and prevents activation of antigen-specific T cellsInt Immunol2009211192819001470
  • NallaparajuKCYuJJRodriguezSAEvasion of IFN-gamma signaling by Francisella novicida is dependent upon Francisella outer membrane protein CPLoS One201163e1820121483828
  • IrelandRWangRAlingerJBSmallPBosioCMFrancisella tularensis Schu S4 and Schu S4 lipids inhibit IL-12p40 in primary human dendritic cells by inhibition of IRF1 and IRF8J Immunol201319131276128623817430
  • LoegeringDJDrakeJRBanasJAFrancisella tularensis LVS grown in macrophages has reduced ability to stimulate the secretion of inflammatory cytokines by macrophages in vitroMicrob Pathog200641621822516996713