Skip to main content
Erschienen in: European Journal of Clinical Microbiology & Infectious Diseases 12/2016

Open Access 06.10.2016 | Original Article

Mitogen-activated protein kinases (MAPKs) are modulated during Francisella tularensis infection, but inhibition of extracellular-signal-regulated kinases (ERKs) is of limited therapeutic benefit

verfasst von: R. J. Saint, R. V. D’Elia, C. Bryant, G. C. Clark, H. S. Atkins

Erschienen in: European Journal of Clinical Microbiology & Infectious Diseases | Ausgabe 12/2016

Abstract

Francisella tularensis is a Gram-negative intracellular bacterium that causes the disease tularemia. The disease can be fatal if left untreated and there is currently no licenced vaccine available; the identification of new therapeutic targets is therefore required. Toll-like receptors represent an interesting target for therapeutic modulation due to their essential role in generating immune responses. In this study, we analysed the in vitro expression of the key mitogen-activated protein kinases (MAPKs) p38, JNK and ERK in murine alveolar macrophages during infection with F. tularensis. The phosphorylation profile of ERK highlighted its potential as a target for therapeutic modulation and subsequently the effect of ERK manipulation was measured in a lethal intranasal F. tularensis in vivo model of infection. The selective ERK1/2 inhibitor PD0325901 was administered orally to mice either pre- or post-challenge with F. tularensis strain LVS. Both treatment regimens selectively reduced ERK expression, but only the pre-exposure treatment produced decreased bacterial burden in the spleen and liver, which correlated with a significant reduction in the pro-inflammatory cytokines IFN-γ, MCP-1, IL-6, and TNF-α. However, no overall improvements in survival were observed for treated animals in this study. ERK may represent a useful therapeutic target where selective dampening of the immune response (to control the damaging pathology seen during infection) is combined with antibiotic treatment required to eradicate bacterial infection. This combination treatment strategy has been shown to be effective in other models of tularemia.

Introduction

Toll-like receptors (TLRs) are sentinels of the innate immune system, functioning to detect invading microorganisms by binding to pathogen-associated molecular patterns (PAMPs) such as lipids, proteins and nucleic acids [13]. The binding event triggers an intracellular signalling cascade through dimerisation of the receptor and the subsequent recruitment of signalling proteins, leading to the production of immune mediators such as chemokines and cytokines. Eleven TLRs have been identified in humans located on multiple cell types including neutrophils, macrophages, and epithelial cells [4, 5]. TLR activity is vital for pathogen clearance, but dysregulation can occur which can have severe consequences for the host. Indeed, it is now well documented that an overactive immune response can be more damaging to the host than the progression of infection by the pathogen [6]. This fine balance is illustrated when a host is infected via the inhalational route with the Gram-negative facultative intracellular bacterium Francisella tularensis, where there is an initial suppression of pro-inflammatory cytokines followed by a “cytokine storm” prior to death [7].
The primary detection of F. tularensis was originally thought to be mediated by TLR4 [8, 9]. However, more recent studies demonstrate that it is likely that TLR2, and not TLR4, confers the ability to detect F. tularensis [1012]. TLR4 was ruled out as a major receptor in the detection of F. tularensis as TLR4-defective mice were no more susceptible to an inhalational infection with F. tularensis than wild-type mice and the survival rates were similar in both mouse strains [13]. In contrast, several studies have reported a requirement for TLR2 in the detection of F. tularensis following in vivo infection by multiple challenge routes or in different types of cultured cell [10, 1419]. More recently, the importance of TLR2-mediated priming of the inflammasome to intracellular survival has been highlighted [20]. Furthermore, a TLR2 deficiency enhanced murine susceptibility to pulmonary infection by the F. tularensis live vaccine strain (LVS) and resulted in decreased production of pro-inflammatory cytokines including TNF-α and IL-6 [10].
A number of studies have previously demonstrated the potential therapeutic benefits of specifically targeting TLRs for the treatment of infectious diseases including those caused by Francisella sp. For example, bacterial DNA containing un-methylated CpG motifs have been used to stimulate TLR9 and induce protection against subsequent challenge with the intracellular pathogens F. tularensis strain LVS and Listeria monocytogenes [21]. Furthermore, stimulation of TLR3 using the synthetic dsRNA analogue poly(I:C) has been evaluated for the treatment of F. tularensis strain LVS or strain SchuS4 infection in Balb/c mice [22]. Treatment as a monotherapy 1 h prior to or 1 h after intranasal infection led to significant improvements in the survival of mice, correlating with an increased influx of neutrophils to the lungs and early up-regulation of cytokines IL-6, MCP-1, and TNF-α [22]. However, it is noteworthy that a combination therapy approach with the antibiotic levofloxacin was required for full recovery.
In comparison to TLR receptors, less research has focused on understanding the activation and phosphorylation states of the downstream immune signalling cascade proteins during F. tularensis infection and/or the potential for selectively targeting these pathways with therapeutics. Previously, it has been demonstrated that the up-regulation of p38, IkB and c-Jun occurs following infection of murine macrophages with F. tularensis strain LVS [15]. In addition, it has been demonstrated that the uptake of Francisella novicida by macrophages is dependent upon ERK activation, and modulation of the MAPK pathways is important for intracellular survival of F. tularensis LVS [23, 24]. Furthermore, within an in-vivo setting it was found that the inhibition of GSK3B (a serine/threonine protein kinase) with lithium chloride significantly reduces the production of pro-inflammatory cytokines such as IFN-γ, IL-12p40, and TNF-α in F. tularensis LVS-infected mice, and increases survival from 60 % in untreated mice to 90 % following treatment [25]. It is hypothesised that targeting specific proteins within the TLR signalling cascades may prevent the wide-ranging effects of direct TLR stimulation. In this study we sought to determine the in vitro expression profile of key mitogen-activated protein kinases (MAPKs) p38, JNK, and ERK following F. tularensis infection. Additionally, we determined the effect of inhibiting ERK expression in vitro and in vivo during a F. tularensis infection.

Materials and methods

Preparation of bacteria

Francisella tularensis live vaccine strain (LVS) was grown overnight on blood cysteine glucose agar (BCGA) plates incubated at 37 °C, and used to inoculate 10 ml PBS to an OD600 of 0.15 (∼1 × 109 CFU/ml). The resulting bacterial suspension was diluted to the required inoculum. To confirm the CFU/ml of the inoculum, serial dilutions were cultured on BCGA plates for subsequent bacterial enumeration.

Infection of MH-S cell line with F. tularensis LVS

A continuous cell line of murine alveolar macrophages (MH-S cells, ECACC UK) were seeded into 6-well plates (Corning, US) at ∼4 × 105 cells/ml and incubated overnight at 37 °C to allow the cells to adhere. Supernatants were then removed and replaced with 1.5 ml of F. tularensis LVS inoculum to generate an MOI of 100. Plates were incubated at 37 °C for 2 h to allow for internalisation of the bacteria, and then the supernatants were removed and replaced with 1.5 ml fresh media. The plates were incubated at 37 °C and, at a range of timepoints, supernatants were removed and stored at −20 °C for cytokine analysis. The cells were washed once with chilled PBS and then lysed by adding 350 μl PhosphoSafe™ extraction reagent (Merck, US) before incubating on ice for 5 min. Full cell recovery was ensured by scraping wells with a rubber policeman (Corning, US): 100 μl of the lysate was used for bacterial enumeration, and a further 250 μl was centrifuged at 13,000 rpm for 5 min to remove cell debris before being stored at −20°C for analysis by western blotting.

In-vivo infection and treatment with ERK inhibitor PD0325901

A total of 65 female 6- to 8-week-old Balb/c mice (Charles River Laboratories, UK) were separated into four groups: group I (20 mice), group II (20 mice), group III (15 mice) and group IV (10 mice). All mice, except those in group IV, were treated daily with either PBS or 0.05 μM PD0325901 (InvivoGen, France) by oral-gavage in a volume of 100 μl. Groups I and II were treated from 24 h pre-infection, while group III was treated from 48 h post-infection. The mice were infected via the intranasal route with approximately 2.5 × 104 CFU of F. tularensis strain LVS. Scoring of the mice occurred twice daily throughout the study. At 48 h and 96 h post-challenge, five mice from each of groups I, II, III, and IV were culled and the lungs, liver, and spleen were removed and placed into aliquots of 2 ml PBS. Ten mice from each of groups I, II, III, and IV were monitored for changes in survival. Organs were homogenised though a 0.2 μm cell sieve (BD Biosciences) and screened for the levels of cytokines, activation of signalling proteins and bacterial enumeration on agar. All animal studies were carried out in accordance with the UK Scientific Procedures Act (1986). Animal handling and infection procedures were carried out by trained staff at Dstl.

Western blots

Proteins were separated on a 4-20 % tris-glycine polyacrylamide gel and then transferred onto Invitrolon™ polyvinylidene fluoride (PVDF) membranes (Invitrogen) using a Novex® Semi-Dry Blotter (Invitrogen) according to manufacturer’s instructions. The PVDF membranes were then incubated in blocking buffer (5 % [w/v] skimmed milk powder and 0.1 % Tween® 20 (Sigma-Aldrich) dissolved in PBS) for 2 h at room temperature. To detect ERK activation, the membranes were washed in wash buffer (0.1 % Tween® 20 in PBS) before being incubated overnight at 4 °C with the primary antibody [p38 - #9218; P-p38 - #9211; SAPK/JNK - #9252; P-SAPK/JNK - #9251; ERK-1 - #4372; ERK-2 - #9108; P-ERK-1/2 - #9101 (NEB)] at a dilution of 1:1,000 in blocking buffer. The membranes were then rinsed in wash buffer and incubated for 1 h at room temperature with the secondary antibody [goat anti-rabbit IgG conjugated to horseradish peroxidise (Bio-Rad)] at a dilution of 1:3,000 in blocking buffer. The membranes were rinsed in wash buffer and developed using enhanced chemiluminescence (ECL)-Plus detection reagents (GE Healthcare) according to manufacturer’s instructions. The membranes were placed in a film cassette and exposed to ECL hyperfilm (GE Healthcare) which was allowed to air-dry before analysis by densitometry.

Cytometric bead array

The quantities of the cytokines IFN-γ, MCP-1, IL-6, IL-12p70, IL-10, and TNF-α present within the lungs were measured using BD™ cytometric bead array (CBA) technology (BD Biosciences) on a BD™ FACScan flow cytometer (BD Biosciences) according to manufacturer’s instructions. A standard curve was generated and the concentration of each cytokine was interpolated using GraphPad Prism® 5.01 software (GraphPad). The levels of IL-12p70 and IL-10 were too low to be detected, and so only the results of IFN-γ, MCP-1, IL-6, and TNF-α are shown.

Statistical analysis

All transformations of data and statistical tests were performed using GraphPad Prism® 5.01 software. Data from cytometric bead arrays were analysed using PRISM, by fitting a quadratic regression to the standard curves and reading the samples as unknowns.

Results

ERK, p38, and JNK represent key signalling molecules in the MAPK cascade [26]. Activation of these proteins, by phosphorylation, has diverse biological effects including cell proliferation, apoptosis, and inflammation [27]. In this study, temporal changes in the expression of total ERK, p38, and JNK and the phosphorylated forms of the proteins produced by in vitro cultured alveolar macrophage (MH-S) cell line in response to infection with F. tularensis LVS was measured via western blot.
The in vitro expression of p38 remained constant during the infection (Fig. 1a). However, there was a significant change in the p38 activation profile. First, a rapid increase in activation occurred immediately after the uptake of LVS; this was a transient response, and the quantities of activated protein present returned to naïve levels by 0.5 h (Fig. 1b). After the rapid and transient activation of p38 there was no re-activation measured throughout the rest of the infection assay. In comparison, limited in vitro activation of JNK proteins was observed during the experiment, with the exception of the 24-h time point, where there was a decrease in the presence of activated JNK-2/3 compared to that found at both 0.75 h and 6 h (Fig. 2d). The expression of total JNK-2/3 remained relatively constant (Fig. 2b), although there was a slight decrease in the expression of total JNK-1 from 6 to 24 h post-uptake (Fig. 2a).
The in vitro expression of total ERK-1 and ERK-2 did not alter significantly at any time point during the infection (Fig. 3a, b). The activation profiles of the two proteins did, however, display some interesting features (Fig. 3c, d). There appeared to be two phases of protein activation, particularly for ERK-2 (Fig. 3d). In the first hour after uptake a transient activation was observed. However, from 1.5 h there was a sustained activation of both ERK proteins which was maintained through to 6 h.
The interesting phosphorylation profile of ERK1/2 following F. tularensis LVS infection in vitro led us to consider modulation of the ERK proteins as an approach for treating infection. An inhibitor of ERK PD0325901was identified and assessed in vivo for its potential as a novel therapy. PD0325901 is a small molecule inhibitor with specific activity against phospho-ERK1/2 and increased potency compared to its predecessors [28, 29]. First, we demonstrated that PD0325901 was able to inhibit ERK expression during an in vitro F. tularensis macrophage infection assay and that it had a biological effect by significantly reducing the secretion of TNF-α (data not shown).
Subsequently, the potential for ERK inhibition (using PD0325901) was explored in vivo using a F. tularensis LVS intranasal model of infection. Balb/C mice infected with F. tularensis LVS showed a substantial increase in the level of ERK activation between 48 h and 96 h, indicating the involvement of ERK phosphorylation during infection in vivo, similarly to in vitro. Daily treatment of infected mice with PD0325901 significantly reduced both ERK-1 and ERK-2 activation in the lungs compared to PBS-treated mice at 96 h (Fig. 4a, b). This was found for both the PD0325901 treatment groups, irrespective of whether administration began at 24 h pre-infection or at 48 h post-infection. However, unexpectedly, at 48 h PD0325901-treated mice appeared to show an increase in ERK activation when compared to PBS-treated mice.
To examine the effect of ERK inhibition on bacterial loads in the organs of infected mice, bacterial enumeration was carried out on samples taken from the lungs, liver, and spleens at 96 h post-infection (Fig. 5a). There was no significant difference in the numbers of bacteria present in the lungs of either of the PD0325901-treated groups compared to PBS-treated controls. However, there was a significant reduction in bacterial burden in the liver and spleen of mice treated with PD0325901 prior to infection when compared to PBS-treated control mice. These differences in the spleen and liver were not detected in mice treated with PD0325901 at 48 h post-infection.
The effect of ERK inhibition on the inflammatory response was examined by measuring the secretion of downstream inflammatory cytokines in the lungs, liver, and spleen of naïve, PBS-treated or PD0325901-treated mice at 96 h after infection (Fig. 5b-e). Measurable levels of IL-6 (Fig. 5b) MCP-1 (Fig. 5c), IFN-γ (Fig. 5d) and TNF-α (Fig. 5e) were detected, but levels of IL-12p40 or IL-10 were below the limit of detection in all samples. At 96 h, no reduction in secretion of any of the cytokines was found in the lungs, despite the observed inhibition of ERK activation in the lungs of mice (Fig. 4). Mice treated from 48 h post-infection with PD0325901 displayed a significant increase in IFN-γ and MCP-1 in livers when compared to PBS-treated mice. The greatest effect of ERK inhibition was observed in spleen samples, where there was a significant decrease in the secreted levels of IFN-γ, MCP-1, IL-6, and TNF-α in mice treated with PD0325901 prior to infection (at day −1) compared to PBS-treated controls. This difference in the spleen was not seen when PD0325901 was administered at 48 h post-infection. Despite the PD0325901-associated inhibition of ERK activation within the lungs and the reduced bacterial burdens in the spleens and livers of infected mice, there was no significant difference in survival of PD0325901-treated groups compared to the PBS-treated groups (Fig. 6).

Discussion

Antibiotics remain the most successful therapeutic regimen for F. tularensis infection, although efficacy is dependent on the correct administration of antibiotic in a timely manner [30]. Aiding antibiotic efficiency through immune-modulation is a strategy that is being explored for infectious diseases.
In these studies, we have demonstrated that total expression of the key MAPKs p38, JNK, and ERK were not significantly altered in an in vitro macrophage model of F. tularensis LVS infection. However, clear temporal changes in the phosphorylated states of the MAPKs were identified. Notably, the in vitro phosphorylation profile of ERK occurred in two distinct peaks of activation within infected macrophages. In addition, we identified that ERK was also significantly upregulated in vivo following intranasal infection of mice with F. tularensis LVS. By administering an inhibitor of ERK phosphorylation commencing both pre- and post-challenge, we aimed to evaluate this as a therapeutic strategy for treating infections caused by this organism. Following oral administration of the inhibitor to mice, a reduction of ERK phosphorylation was successfully achieved in vivo, with the greatest reductions observed when the compound was given prior to infection of mice. Treatment with the inhibitor resulted in significant decreases in bacterial burden in the spleen and liver but not at primary foci of infection of the lung. The reduction in bacterial burden correlated with a significant decrease in the pro-inflammatory cytokines IFN-γ, MCP-1, IL-6, and TNF-α, although survival of infected mice was not improved through administration of the inhibitor.
It has previously been reported that the activation of ERK signalling occurs in response to F. tularensis infection in vitro and in vivo [9, 19, 31]. Additionally, in vitro studies have demonstrated that MAPK activation, including ERK activation, plays an important role in F. tularensis LVS infection-induced apoptosis [32, 33]. To our knowledge, however, this is the first assessment of manipulation of ERK as a possible strategy for controlling F. tularensis infection. We found that ERK signalling in the lungs of infected mice could be successfully dampened by PD0325901 administration via oral gavage. This did not result in a reduction in bacterial numbers in the lungs (the site of bacterial infection), but there were significantly reduced bacterial burdens in both the liver and spleen of pre-treated mice. This may indicate that the proliferation of the bacteria from the primary site of infection has been reduced.
Sustained ERK activity promotes either intrinsic or extrinsic apoptotic pathways by induction of mitochondrial Cytochrome C release or caspase-8 activation, permanent cell cycle arrest, or autophagic vacuolisation [34]. Indeed, it has been suggested that the rapid proliferation of Francisella coincides with the initial activation of the apoptotic signalling pathway [35]. Therefore the relationship between ERK activation and the host mechanism of apoptosis could explain the observed differences in bacterial burden and cytokine response [33, 36]. In untreated F. tularensis infection (and in this study, in PBS-treated mice), the replication of bacteria within the lungs may induce apoptosis of infected cells such as alveolar macrophages via ERK activation, releasing the bacteria for proliferation throughout the host. In contrast, by limiting the activation of ERK through the administration of the inhibitor PD0325901, it is conceivable that the degree of apoptosis could be reduced, preventing cellular release and dissemination of the bacteria. In this context, the lungs act as a trap, with the result that bacterial burdens of this organ are unaffected whereas secondary sites, such as the liver and spleen, demonstrate reduced bacterial burdens as observed here. Further research involving the in vivo administration for the inhibitor and analysis of the presence/absence of apoptotic markers on cellular surfaces is required in order to fully evaluate this hypothesis, which was beyond the scope of this study.
In this study, ERK inhibition did not only alter bacterial burden but also had a significant effect on the pro-inflammatory secretion profile; specifically it was found that IFN-γ, MCP-1, IL-6, and TNF-α were all reduced in vivo following pre-treatment with PD0325901. It is well documented that an inflammatory response is essential for pathogen clearance, although it is also clear that pro-inflammatory responses contribute to the tissue damage and pathology seen during infection [6]. Indeed, the overactive immune response seen late during Francisella infection has been hypothesised to be one of the main contributors of detrimental outcome [7, 37]. Therefore, in this context, the modulation of the immune response achieved through inhibition of ERK with PD0325901 could be beneficial to survival of the host following infection. However, in this study we demonstrated that although dampening of ERK signalling was observed, along with an associated reduction in cytokine secretion and a decreased systemic proliferation of F. tularensis in treated mice, ultimately there was no measurable improvement in survival when compared to PBS-treated mice. This indicates that ERK inhibition alone is not sufficient to improve the outcome of infection with this pathogen. However, it is possible that a combination therapy, for example administering an ERK inhibitor with an antibiotic, could demonstrate a more beneficial outcome of infection. The ERK inhibitor could be used in this context to indirectly reduce systemic spread, whilst a bactericidal antibiotic directly kills the bacteria that remain within the lung. An analogous therapeutic strategy has already been shown to be beneficial in our laboratory for the treatment of the fully virulent SCHUS4 strain of F. tularensis, whereby inhibition of HMGB1, combined with levofloxacin use, significantly increased survival of infected mice compared to controls [7]. In addition, related approaches involving the administration of an antibiotic and an immunodulating therapy have been reported for other bacterial pathogens as well as in the treatment of sepsis [38]. It is important to note that the data generated in this study is derived from a murine model of tularemia as the availability of clinical samples from human cases of the disease is very limited, and it is not possible to conduct clinical trials in humans. However, it is well documented that there are similarities between human tularemia and disease presentation within mouse models, which are well accepted for research on F. tularensis [39, 40]. Therefore it is reasonable to extrapolate that targeting highly conserved MAPKs in humans would likely have a similar therapeutic benefit.
In conclusion, we have shown that PD0325901 can successfully alter ERK activation during an in vivo F. tularensis infection. It caused a reduction in bacterial burden in the liver and spleen if given prior to infection, and also significantly altered the host cytokine profile. The observed difference between mice given PD0325901 prior to or after infection is probably due to the time the compound requires to take effect in vivo and/or as a consequence of the bioavailability of the compound. Ultimately, therapeutic manipulation of ERK did not result in increased survival; however, it may be plausible that PD0325901 and/or other ERK inhibitors would be suitable for assessment within models of infection in combination with an anti-microbial therapy.

Acknowledgments

The authors would like to thank the animal technicians that assisted with these studies, and the UK Ministry of Defence for funding this work.

Compliance with ethical standards

Funding

This work was funded by the UK Ministry of Defence.

Conflict of Interest

To the authors knowledge there is no conflict of interest arising from the publication of this manuscript.

Ethical approval

All procedures performed in studies involving animals were in accordance with the ethical standards of the institution or practice at which the studies were conducted.
Not applicable.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Alexopoulou L, Holt AC, Medzhitov R, Flavell RA (2001) Recognition of double-stranded RNA and activation of NF-kappa B by Toll-like receptor 3. Nature 413:732–738CrossRefPubMed Alexopoulou L, Holt AC, Medzhitov R, Flavell RA (2001) Recognition of double-stranded RNA and activation of NF-kappa B by Toll-like receptor 3. Nature 413:732–738CrossRefPubMed
2.
Zurück zum Zitat Jin MS, Kim SE, Heo JY, Lee ME, Kim HM et al (2007) Crystal structure of the TLR1-TLR2 heterodimer induced by binding of a tri-acylated lipopeptide. Cell 130:1071–1082CrossRefPubMed Jin MS, Kim SE, Heo JY, Lee ME, Kim HM et al (2007) Crystal structure of the TLR1-TLR2 heterodimer induced by binding of a tri-acylated lipopeptide. Cell 130:1071–1082CrossRefPubMed
3.
Zurück zum Zitat Hirschfeld M, Weis JJ, Toshchakov V, Salkowski CA, Cody MJ et al (2001) Signaling by Toll-like receptor 2 and 4 agonists results in differential gene expression in murine macrophages. Infect Immun 69:1477–1482CrossRefPubMedPubMedCentral Hirschfeld M, Weis JJ, Toshchakov V, Salkowski CA, Cody MJ et al (2001) Signaling by Toll-like receptor 2 and 4 agonists results in differential gene expression in murine macrophages. Infect Immun 69:1477–1482CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Droemann D, Goldmann T, Branscheid D, Clark R, Dalhoff K et al (2003) Toll-like receptor 2 is expressed by alveolar epithelial cells type II and macrophages in the human lung. Histochem Cell Biol 119:103–108PubMed Droemann D, Goldmann T, Branscheid D, Clark R, Dalhoff K et al (2003) Toll-like receptor 2 is expressed by alveolar epithelial cells type II and macrophages in the human lung. Histochem Cell Biol 119:103–108PubMed
5.
Zurück zum Zitat Sandor F, Buc M (2005) Toll-like receptors. II. Distribution and pathways involved in TLR signalling. Folia Biol 51:188–197 Sandor F, Buc M (2005) Toll-like receptors. II. Distribution and pathways involved in TLR signalling. Folia Biol 51:188–197
6.
Zurück zum Zitat D’Elia RV, Harrison K, Oyston PC, Lukaszewski RA, Clark GC (2013) Targeting the “cytokine storm” for therapeutic benefit. Clin Vaccine Immunol 20:319–327CrossRefPubMedPubMedCentral D’Elia RV, Harrison K, Oyston PC, Lukaszewski RA, Clark GC (2013) Targeting the “cytokine storm” for therapeutic benefit. Clin Vaccine Immunol 20:319–327CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Lembo A, Pelletier M, Iyer R, Timko M, Dudda JC et al (2008) Administration of a synthetic TLR4 agonist protects mice from pneumonic tularemia. J Immunol 180:7574–7581CrossRefPubMedPubMedCentral Lembo A, Pelletier M, Iyer R, Timko M, Dudda JC et al (2008) Administration of a synthetic TLR4 agonist protects mice from pneumonic tularemia. J Immunol 180:7574–7581CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Cole LE, Mann BJ, Shirey KA, Richard K, Yang Y et al (2011) Role of TLR signaling in Francisella tularensis-LPS-induced, antibody-mediated protection against Francisella tularensis challenge. J Leukoc Biol 90:787–797CrossRefPubMedPubMedCentral Cole LE, Mann BJ, Shirey KA, Richard K, Yang Y et al (2011) Role of TLR signaling in Francisella tularensis-LPS-induced, antibody-mediated protection against Francisella tularensis challenge. J Leukoc Biol 90:787–797CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Malik M, Bakshi CS, Sahay B, Shah A, Lotz SA et al (2006) Toll-like receptor 2 is required for control of pulmonary infection with Francisella tularensis. Infect Immun 74:3657–3662CrossRefPubMedPubMedCentral Malik M, Bakshi CS, Sahay B, Shah A, Lotz SA et al (2006) Toll-like receptor 2 is required for control of pulmonary infection with Francisella tularensis. Infect Immun 74:3657–3662CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Macela A, Stulik J, Hernychova L, Kroca M, Krocova Z et al (1996) The immune response against Francisella tularensis live vaccine strain in Lps(n) and Lps(d) mice. FEMS Immunol Med Microbiol 13:235–238CrossRefPubMed Macela A, Stulik J, Hernychova L, Kroca M, Krocova Z et al (1996) The immune response against Francisella tularensis live vaccine strain in Lps(n) and Lps(d) mice. FEMS Immunol Med Microbiol 13:235–238CrossRefPubMed
12.
Zurück zum Zitat Chen WX, KuoLee R, Shen H, Busa M, Conlan JW (2004) Toll-like receptor 4 (TLR4) does not confer a resistance advantage on mice against low-dose aerosol infection with virulent type A Francisella tularensis. Microb Pathog 37:185–191CrossRefPubMed Chen WX, KuoLee R, Shen H, Busa M, Conlan JW (2004) Toll-like receptor 4 (TLR4) does not confer a resistance advantage on mice against low-dose aerosol infection with virulent type A Francisella tularensis. Microb Pathog 37:185–191CrossRefPubMed
13.
Zurück zum Zitat Chen WX, KuoLee R, Shen H, Busa M, Conlan JW (2005) Toll-like receptor 4 (TLR4) plays a relatively minor role in murine defense against primary intradermal infection with Francisella tularensis LVS. Immunol Lett 97:151–154CrossRefPubMed Chen WX, KuoLee R, Shen H, Busa M, Conlan JW (2005) Toll-like receptor 4 (TLR4) plays a relatively minor role in murine defense against primary intradermal infection with Francisella tularensis LVS. Immunol Lett 97:151–154CrossRefPubMed
14.
Zurück zum Zitat Cole LE, Santiago A, Barry E, Kang TJ, Shirey KA et al (2008) Macrophage proinflammatory response to Francisella tularensis live vaccine strain requires coordination of multiple signaling pathways. J Immunol 180:6885–6891CrossRefPubMedPubMedCentral Cole LE, Santiago A, Barry E, Kang TJ, Shirey KA et al (2008) Macrophage proinflammatory response to Francisella tularensis live vaccine strain requires coordination of multiple signaling pathways. J Immunol 180:6885–6891CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Telepnev M, Golovliov I, Sjostedt A (2005) Francisella tularensis LVS initially activates but subsequently down-regulates intracellular signaling and cytokine secretion in mouse monocytic and human peripheral blood mononuclear cells. Microb Pathog 38:239–247CrossRefPubMed Telepnev M, Golovliov I, Sjostedt A (2005) Francisella tularensis LVS initially activates but subsequently down-regulates intracellular signaling and cytokine secretion in mouse monocytic and human peripheral blood mononuclear cells. Microb Pathog 38:239–247CrossRefPubMed
16.
Zurück zum Zitat Hong KJ, Wickstrum JR, Yeh HW, Parmely MJ (2007) Toll-like receptor 2 controls the gamma interferon response to Francisella tularensis by mouse liver lymphocytes. Infect Immun 75:5338–5345CrossRefPubMedPubMedCentral Hong KJ, Wickstrum JR, Yeh HW, Parmely MJ (2007) Toll-like receptor 2 controls the gamma interferon response to Francisella tularensis by mouse liver lymphocytes. Infect Immun 75:5338–5345CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Cole LE, Elkins KL, Michalek SM, Qureshi N, Eaton LJ et al (2006) Immunologic consequences of Francisella tularensis live vaccine strain infection: role of the innate immune response in infection and immunity. J Immunol 176:6888–6899CrossRefPubMed Cole LE, Elkins KL, Michalek SM, Qureshi N, Eaton LJ et al (2006) Immunologic consequences of Francisella tularensis live vaccine strain infection: role of the innate immune response in infection and immunity. J Immunol 176:6888–6899CrossRefPubMed
18.
Zurück zum Zitat Cole LE, Shirey KA, Barry E, Santiago A, Rallabhandi P et al (2007) Toll-like receptor 2-mediated signaling requirements for Francisella tularensis live vaccine strain infection of murine macrophages. Infect Immun 75:4127–4137CrossRefPubMedPubMedCentral Cole LE, Shirey KA, Barry E, Santiago A, Rallabhandi P et al (2007) Toll-like receptor 2-mediated signaling requirements for Francisella tularensis live vaccine strain infection of murine macrophages. Infect Immun 75:4127–4137CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Thakran S, Li HF, Lavine CL, Miller MA, Bina JE et al (2008) Identification of Francisella tularensis lipoproteins that stimulate the toll-like receptor (TLR)2/TLR1 heterodimer. J Biol Chem 283:3751–3760CrossRefPubMed Thakran S, Li HF, Lavine CL, Miller MA, Bina JE et al (2008) Identification of Francisella tularensis lipoproteins that stimulate the toll-like receptor (TLR)2/TLR1 heterodimer. J Biol Chem 283:3751–3760CrossRefPubMed
20.
Zurück zum Zitat Ghonime MG, Mitra S, Eldomany RA, Wewers MD, Gavrilin MA (2015) Inflammasome priming is similar for francisella species that differentially induce inflammasome activation. PLoS One 10:e0127278CrossRefPubMedPubMedCentral Ghonime MG, Mitra S, Eldomany RA, Wewers MD, Gavrilin MA (2015) Inflammasome priming is similar for francisella species that differentially induce inflammasome activation. PLoS One 10:e0127278CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Elkins KL, Rhinehart-Jones TR, Stibitz S, Conover JS, Klinman DM (1999) Bacterial DNA containing CpG motifs stimulates lymphocyte-dependent protection of mice against lethal infection with intracellular bacteria. J Immunol 162:2291–2298PubMed Elkins KL, Rhinehart-Jones TR, Stibitz S, Conover JS, Klinman DM (1999) Bacterial DNA containing CpG motifs stimulates lymphocyte-dependent protection of mice against lethal infection with intracellular bacteria. J Immunol 162:2291–2298PubMed
22.
Zurück zum Zitat Pyles RB, Jezek GE, Eaves-Pyles TD (2010) Toll-like receptor 3 agonist protection against experimental Francisella tularensis respiratory tract infection. Infect Immun 78:1700–1710CrossRefPubMedPubMedCentral Pyles RB, Jezek GE, Eaves-Pyles TD (2010) Toll-like receptor 3 agonist protection against experimental Francisella tularensis respiratory tract infection. Infect Immun 78:1700–1710CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Parsa KVL, Butchar JP, Rajaram MVS, Cremer TJ, Tridandapani S (2008) The tyrosine kinase Syk promotes phagocytosis of Francisella through the activation of Erk. Mol Immunol 45:3012–3021CrossRefPubMedPubMedCentral Parsa KVL, Butchar JP, Rajaram MVS, Cremer TJ, Tridandapani S (2008) The tyrosine kinase Syk promotes phagocytosis of Francisella through the activation of Erk. Mol Immunol 45:3012–3021CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Rabadi SM, Sanchez BC, Varanat M, Ma Z, Catlett SV et al (2016) Antioxidant defenses of Francisella tularensis modulate macrophage function and production of proinflammatory cytokines. J Biol Chem 291:5009–5021CrossRefPubMed Rabadi SM, Sanchez BC, Varanat M, Ma Z, Catlett SV et al (2016) Antioxidant defenses of Francisella tularensis modulate macrophage function and production of proinflammatory cytokines. J Biol Chem 291:5009–5021CrossRefPubMed
25.
Zurück zum Zitat Zhang P, Katz J, Michalek SM (2009) Glycogen synthase kinase-3 beta (GSK3 beta) inhibition suppresses the inflammatory response to Francisella infection and protects against tularemia in mice. Mol Immunol 46:677–687CrossRefPubMed Zhang P, Katz J, Michalek SM (2009) Glycogen synthase kinase-3 beta (GSK3 beta) inhibition suppresses the inflammatory response to Francisella infection and protects against tularemia in mice. Mol Immunol 46:677–687CrossRefPubMed
26.
Zurück zum Zitat Roux PP, Blenis J (2004) ERK and p38 MAPK-activated protein kinases: a family of protein kinases with diverse biological functions. Microbiol Mol Biol Rev 68:320–344CrossRefPubMedPubMedCentral Roux PP, Blenis J (2004) ERK and p38 MAPK-activated protein kinases: a family of protein kinases with diverse biological functions. Microbiol Mol Biol Rev 68:320–344CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Zhang W, Liu HT (2002) MAPK signal pathways in the regulation of cell proliferation in mammalian cells. Cell Res 12:9–18CrossRefPubMed Zhang W, Liu HT (2002) MAPK signal pathways in the regulation of cell proliferation in mammalian cells. Cell Res 12:9–18CrossRefPubMed
28.
Zurück zum Zitat Barrett SD, Bridges AJ, Dudley DT, Saltiel AR, Fergus JH et al (2008) The discovery of the benzhydroxamate MEK inhibitors CI-1040 and PD 0325901. Bioorg Med Chem Lett 18:6501–6504CrossRefPubMed Barrett SD, Bridges AJ, Dudley DT, Saltiel AR, Fergus JH et al (2008) The discovery of the benzhydroxamate MEK inhibitors CI-1040 and PD 0325901. Bioorg Med Chem Lett 18:6501–6504CrossRefPubMed
29.
Zurück zum Zitat Thompson N, Lyons J (2005) Recent progress in targeting the Raf/MEK/ERK pathway with inhibitors in cancer drug discovery. Curr Opin Pharmacol 5:350–356CrossRefPubMed Thompson N, Lyons J (2005) Recent progress in targeting the Raf/MEK/ERK pathway with inhibitors in cancer drug discovery. Curr Opin Pharmacol 5:350–356CrossRefPubMed
30.
Zurück zum Zitat Klimpel GR, Eaves-Pyles T, Moen ST, Taormina J, Peterson JW et al (2008) Levofloxacin rescues mice from lethal intra-nasal infections with virulent Francisella tularensis and induces immunity and production of protective antibody. Vaccine 26:6874–6882CrossRefPubMedPubMedCentral Klimpel GR, Eaves-Pyles T, Moen ST, Taormina J, Peterson JW et al (2008) Levofloxacin rescues mice from lethal intra-nasal infections with virulent Francisella tularensis and induces immunity and production of protective antibody. Vaccine 26:6874–6882CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat Abplanalp AL, Morris IR, Parida BK, Teale JM, Berton MT (2009) TLR-dependent control of Francisella tularensis infection and host inflammatory responses. PLoS ONE 4:e7920CrossRefPubMedPubMedCentral Abplanalp AL, Morris IR, Parida BK, Teale JM, Berton MT (2009) TLR-dependent control of Francisella tularensis infection and host inflammatory responses. PLoS ONE 4:e7920CrossRefPubMedPubMedCentral
32.
Zurück zum Zitat Hrstka R, Stulik J, Vojtesek B (2005) The role of MAPK signal pathways during Francisella tularensis LVS infection-induced apoptosis in murine macrophages. Microbes Infect 7:619–625CrossRefPubMed Hrstka R, Stulik J, Vojtesek B (2005) The role of MAPK signal pathways during Francisella tularensis LVS infection-induced apoptosis in murine macrophages. Microbes Infect 7:619–625CrossRefPubMed
33.
Zurück zum Zitat Du K, Zheng Q, Zhou M, Zhu L, Ai B et al (2011) Chlamydial antiapoptotic activity involves activation of the Raf/MEK/ERK survival pathway. Curr Microbiol 63:341–346CrossRefPubMed Du K, Zheng Q, Zhou M, Zhu L, Ai B et al (2011) Chlamydial antiapoptotic activity involves activation of the Raf/MEK/ERK survival pathway. Curr Microbiol 63:341–346CrossRefPubMed
34.
Zurück zum Zitat Cagnol S, Chambard JC (2010) ERK and cell death: mechanisms of ERK-induced cell death—apoptosis, autophagy and senescence. FEBS J 277:2–21CrossRefPubMed Cagnol S, Chambard JC (2010) ERK and cell death: mechanisms of ERK-induced cell death—apoptosis, autophagy and senescence. FEBS J 277:2–21CrossRefPubMed
35.
Zurück zum Zitat Lai XH, Sjostedt A (2003) Delineation of the molecular mechanisms of Francisella tularensis-induced apoptosis in murine macrophages. Infect Immun 71:4642–4646CrossRefPubMedPubMedCentral Lai XH, Sjostedt A (2003) Delineation of the molecular mechanisms of Francisella tularensis-induced apoptosis in murine macrophages. Infect Immun 71:4642–4646CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Asim M, Chaturvedi R, Hoge S, Lewis ND, Singh K et al (2010) Helicobacter pylori induces ERK-dependent formation of a Phospho-c-Fos.c-Jun activator protein-1 complex that causes apoptosis in macrophages. J Biol Chem 285:20343–20357CrossRefPubMedPubMedCentral Asim M, Chaturvedi R, Hoge S, Lewis ND, Singh K et al (2010) Helicobacter pylori induces ERK-dependent formation of a Phospho-c-Fos.c-Jun activator protein-1 complex that causes apoptosis in macrophages. J Biol Chem 285:20343–20357CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Cowley SC (2009) Proinflammatory cytokines in pneumonic tularemia: too much too late? J Leukoc Biol 86:469–470CrossRefPubMed Cowley SC (2009) Proinflammatory cytokines in pneumonic tularemia: too much too late? J Leukoc Biol 86:469–470CrossRefPubMed
38.
Zurück zum Zitat Leelahavanichkul A, Bocharov AV, Kurlander R, Baranova IN, Vishnyakova TG et al (2012) Class B scavenger receptor types I and II and CD36 targeting improves sepsis survival and acute outcomes in mice. J Immunol 188:2749–2758CrossRefPubMed Leelahavanichkul A, Bocharov AV, Kurlander R, Baranova IN, Vishnyakova TG et al (2012) Class B scavenger receptor types I and II and CD36 targeting improves sepsis survival and acute outcomes in mice. J Immunol 188:2749–2758CrossRefPubMed
39.
Zurück zum Zitat Fritz DL, England MJ, Miller L, Waag DM (2014) Mouse models of aerosol-acquired tularemia caused by Francisella tularensis types A and B. Comp Med 64:341–350PubMedPubMedCentral Fritz DL, England MJ, Miller L, Waag DM (2014) Mouse models of aerosol-acquired tularemia caused by Francisella tularensis types A and B. Comp Med 64:341–350PubMedPubMedCentral
40.
Zurück zum Zitat Elkins KL, RhinehartJones TR, Culkin SJ, Yee D, Winegar RK (1996) Minimal requirements for murine resistance to infection with Francisella tularesis LVS. Infect Immun 64:3288–3293PubMedPubMedCentral Elkins KL, RhinehartJones TR, Culkin SJ, Yee D, Winegar RK (1996) Minimal requirements for murine resistance to infection with Francisella tularesis LVS. Infect Immun 64:3288–3293PubMedPubMedCentral
Metadaten
Titel
Mitogen-activated protein kinases (MAPKs) are modulated during Francisella tularensis infection, but inhibition of extracellular-signal-regulated kinases (ERKs) is of limited therapeutic benefit
verfasst von
R. J. Saint
R. V. D’Elia
C. Bryant
G. C. Clark
H. S. Atkins
Publikationsdatum
06.10.2016
Verlag
Springer Berlin Heidelberg
Erschienen in
European Journal of Clinical Microbiology & Infectious Diseases / Ausgabe 12/2016
Print ISSN: 0934-9723
Elektronische ISSN: 1435-4373
DOI
https://doi.org/10.1007/s10096-016-2754-1

Weitere Artikel der Ausgabe 12/2016

European Journal of Clinical Microbiology & Infectious Diseases 12/2016 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Bei Herzinsuffizienz muss „Eisenmangel“ neu definiert werden!

16.05.2024 Herzinsuffizienz Nachrichten

Bei chronischer Herzinsuffizienz macht es einem internationalen Expertenteam zufolge wenig Sinn, die Diagnose „Eisenmangel“ am Serumferritin festzumachen. Das Team schlägt vor, sich lieber an die Transferrinsättigung zu halten.

Herzinfarkt mit 85 – trotzdem noch intensive Lipidsenkung?

16.05.2024 Hypercholesterinämie Nachrichten

Profitieren nach einem akuten Myokardinfarkt auch Betroffene über 80 Jahre noch von einer intensiven Lipidsenkung zur Sekundärprävention? Um diese Frage zu beantworten, wurden jetzt Registerdaten aus Frankreich ausgewertet.

ADHS-Medikation erhöht das kardiovaskuläre Risiko

16.05.2024 Herzinsuffizienz Nachrichten

Erwachsene, die Medikamente gegen das Aufmerksamkeitsdefizit-Hyperaktivitätssyndrom einnehmen, laufen offenbar erhöhte Gefahr, an Herzschwäche zu erkranken oder einen Schlaganfall zu erleiden. Es scheint eine Dosis-Wirkungs-Beziehung zu bestehen.

Erstmanifestation eines Diabetes-Typ-1 bei Kindern: Ein Notfall!

16.05.2024 DDG-Jahrestagung 2024 Kongressbericht

Manifestiert sich ein Typ-1-Diabetes bei Kindern, ist das ein Notfall – ebenso wie eine diabetische Ketoazidose. Die Grundsäulen der Therapie bestehen aus Rehydratation, Insulin und Kaliumgabe. Insulin ist das Medikament der Wahl zur Behandlung der Ketoazidose.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.