Skip to main content
  • Research article
  • Open access
  • Published:

Global transcriptional response to mammalian temperature provides new insight into Francisella tularensis pathogenesis

Abstract

Background

After infecting a mammalian host, the facultative intracellular bacterium, Francisella tularensis, encounters an elevated environmental temperature. We hypothesized that this temperature change may regulate genes essential for infection.

Results

Microarray analysis of F. tularensis LVS shifted from 26°C (environmental) to 37°C (mammalian) showed ~11% of this bacterium's genes were differentially-regulated. Importantly, 40% of the protein-coding genes that were induced at 37°C have been previously implicated in virulence or intracellular growth of Francisella in other studies, associating the bacterial response to this temperature shift with pathogenesis. Forty-four percent of the genes induced at 37°C encode proteins of unknown function, suggesting novel Francisella virulence traits are regulated by mammalian temperature. To explore this possibility, we generated two mutants of loci induced at 37°C [FTL_1581 and FTL_1664 (deoB)]. The FTL_1581 mutant was attenuated in a chicken embryo infection model, which was likely attributable to a defect in survival within macrophages. FTL_1581 encodes a novel hypothetical protein that we suggest naming temperature-induced, virulence-associated locus A, tivA. Interestingly, the deoB mutant showed diminished entry into mammalian cells compared to wild-type LVS, including primary human macrophages and dendritic cells, the macrophage-like RAW 264.7 line, and non-phagocytic HEK-293 cells. This is the first study identifying a Francisella gene that contributes to uptake into both phagocytic and non-phagocytic host cells.

Conclusion

Our results provide new insight into mechanisms of Francisella virulence regulation and pathogenesis. F. tularensis LVS undergoes considerable gene expression changes in response to mammalian body temperature. This temperature shift is important for the regulation of genes that are critical for the pathogenesis of Francisella. Importantly, the compilation of temperature-regulated genes also defines a rich collection of novel candidate virulence determinants, including tivA (FTL_1581). An analysis of tivA and deoB (FTL_1664) revealed that these genes contribute to intracellular survival and entry into mammalian cells, respectively.

Background

Francisella tularensis is a Gram-negative bacterium that is pathogenic to humans [1]. This organism causes mortality in up to 60% of infected individuals if untreated [2]. Based on the potential to weaponize this organism, the Center for Disease Control and Prevention has classified F. tularensis as a Category A biodefense agent [3]. It is therefore vital to understand how this organism responds to environmental and host signals, and how these cues alter expression of virulence determinants. During the course of a natural Francisella infection, this bacterium may transition from an amoeba [4] or an arthropod host [5] to colonize human cells. Accompanying this transition, it is likely that chemical and physical signals alert Francisella that it has entered a mammalian host.

The manner by which F. tularensis integrates environmental stimuli to regulate gene expression is fundamental to the success of this organism as an intracellular pathogen. The most well-studied virulence factors of F. tularensis are encoded in the Francisella Pathogenicity Island (FPI) [6]. The amount of one of the virulence proteins encoded in this cluster, IglC, increases in response to growth in macrophages and hydrogen peroxide [7]. In addition, iron limitation has been shown to induce transcription and protein levels of IglACD and PdpB [8, 9] as well as stimulate siderophore production by F. tularensis [10]. Previously, we have shown that differing culture conditions greatly influence host-pathogen interactions and the ability of F. tularensis live vaccine strain (LVS) to activate macrophages [11].

Prior to the current study, there has only been a single published report characterizing the global transcriptional Francisella response to an environmental cue, specifically iron limitation [8]. Important insights into the regulation of virulence factors like iglC were defined in this analysis. However, numerous genes associated with virulence by other studies are not affected by different iron concentrations [8, 12–15]. Because F. tularensis may transition between hosts, mammalian body temperature is another signal that is likely to be critical for this pathogen.

Pathogenic bacteria that encounter a shift in temperature during their life cycle sometimes respond with enhanced virulence factor expression [16–19]. However, there are discrepancies among the specific groups of genes that are affected by temperature and the mechanism of regulation between organisms. For example, Shigella increases production of its Type III secretion system in response to mammalian temperature [20]. The homologous secretion apparatus in pathogenic Salmonella, however, is not regulated by temperature [21]. Regarding the mechanism of regulation, genes involved in the heat-shock response are often induced at mammalian temperatures relative to those of the environment. This regulation is usually due to the presence of a conserved inverted repeat regulatory structure in the promoter region [22], or control by a σ32-type heat shock sigma factor [23]. In contrast, the bacterium responsible for whooping cough, Bordetella pertussis, uses a two-component system comprised of BvgS and BvgA to alter transcription of genes in response to temperature. Following induction of bvgAS at 37°C, phosphorylation by BvgS allows BvgA-binding to promoter regions of virulence genes, such as the adhesin, fimX [24]. Given the uncertainty of target temperature-regulated genes and differences in mechanism among bacteria, it is necessary to define the temperature regulon in individual species. Studying gene expression changes induced by a shift to mammalian temperature could provide valuable insight into Francisella virulence.

A few previous studies have investigated the Francisella response to temperature at the molecular level. High temperature (42–44°C) synonymous with heat stress has been shown to have dramatic effects on F. tularensis gene and protein expression [25–27] including increased production of the heat shock proteins GroEL, GroES, DnaK, and ClpB [27]. Heat stress has also been shown to enhance the virulence of a mutant form of F. tularensis LVS [28]. Interestingly, F. novicida has been shown to alter its outer membrane at 25°C versus 37°C by differentially modifying the lipid A component of the lipopolysaccharide [29]. In addition, it has been observed that virulent F. tularensis clinical isolates increase the mannose modification of their lipopolysaccharide when cultivated at temperatures less than 25°C [30]. The results from these previous studies suggest that a global transcript analysis following a shift to mammalian body temperature should reveal gene regulation that is critical to Francisella pathogenesis.

Here we show that a shift from environmental to mammalian body temperature significantly alters the transcriptome of F. tularensis LVS. Many genes that we identified as significantly induced at 37°C have been previously implicated in Francisella virulence, supporting the notion that this temperature shift is important for the regulation of pathogenesis. We provide evidence that the product of a gene encoding the hypothetical protein most profoundly induced by mammalian temperature contributes to virulence and intracellular growth. Moreover, data presented here indicate that a locus up-regulated at 37°C was required for optimal uptake by eukaryotic cells. This is the first F. tularensis gene identified to be involved in the entry of this organism into both phagocytic and non-phagocytic host cells. The results from this study support a model where detection of mammalian body temperature by F. tularensis is important for regulation of physiology necessary for successful infection.

Results

F. tularensisLVS global temperature regulation

To study Francisella gene expression changes associated with exposure to mammalian temperature, we conducted a microarray analysis of LVS as a model F. tularensis strain (Table 1). Gene expression of LVS cultured at 26°C (non-mammalian environment) was compared to bacteria shifted to 37°C (mammalian host body temperature). Labeled cDNA target was produced from RNA isolated from F. tularensis LVS and was subsequently hybridized to a custom Agilent Francisella microarray. Global gene expression data were analyzed with a J5 statistical test, which was selected to limit the number of false positives [31]. This analysis identified 95 genes with significantly increased expression and 125 genes with decreased expression in response to a shift to mammalian body temperature (see additional files 1 and 2, respectively). Collectively, this represents approximately ~11% of the genes in the entire LVS genome. Genes with a significant change in expression were examined with the Gene Pattern program by hierarchical clustering (Fig. 1). The clustered data exhibited a distinct pattern of transcript induction and repression in response to mammalian body temperature (Fig. 1). Together, these data indicate that this temperature shift has a broad impact on F. tularensis transcription.

Table 1 Strains, plasmids, and primers used in this study.
Figure 1
figure 1

Global Francisella gene expression changes induced by mammalian body temperature. Genes that were identified as statistically significant by J5 scores across three independent microarray experiments were subjected to hierarchical clustering. Fluorescence intensities were standardized by the minimum mean ratio array normalization followed by log2 transformation. Values were clustered using the Pearson correlation in GenePattern by independently inputting individual data from each experiment. Oligonucleotides testing individual LVS ORFs are in duplicate on each array and the results are displayed as two columns per experiment in GenePattern. The 95 induced genes and 125 repressed genes clustered together in this GenePattern output, validating the J5 statistical analysis from GEDA. (B).

The microarray data were confirmed using quantitative real time PCR (Q-PCR), in which eight differentially-expressed genes were analyzed, representing transcripts that were both significantly up- and down-regulated (Fig. 2A). Induced genes chosen for validation were the carbamoyl-phosphate synthase large chain, carB (FTL_0029), a hypothetical lipoprotein (FTL_1581), a dimethyladenosine transferase annotated to function in kasugamycin resistance, ksgA (FTL_1595), and phosphopentomutase, deoB (FTL_1664). The down-regulated genes tested by Q-PCR included two hypothetical proteins (FTL_1315 and FTL_1846), a cold shock protein, cspC (FTL_1361), and a metal ion transporter, tlyC (FTL_1697). A similar pattern of expression was observed in both Q-PCR and in the microarray experiments (Fig. 2A). A correlation plot (Fig. 2B) showed a strong, positive association between both data sets (R2 = 0.94). This indicated the microarray platform and the subsequent statistical analysis were robust compared to the sensitive, though low-throughput Q-PCR approach.

Figure 2
figure 2

Validation of the microarray results and comparison with Q-PCR. (A) RNA from the microarray experiments depicted in panel A were tested for up- or down-regulation of specific genes using Q-PCR. Q-PCR data are represented with solid bars, whereas values from microarray experiments are depicted as striped bars. Both data sets are presented as mean ± SEM from three individual experiments. (B) Correlation analysis of the microarray and Q-PCR transcript measurements for nine select F. tularensis LVS ORFs. The microarray log2 values were plotted against the Q-PCR log2 data. The correlation coefficient (R2) between the two analyses is 0.94.

To analyze the differentially-regulated genes more thoroughly, we categorized their presumed protein products based on their Clusters of Orthologous Groups (COG) category. As expected, many of the induced and repressed genes were involved in central biological functions such as metabolism, transcription, translation, DNA replication, and RNA genes (Fig. 3). Heat shock proteins belong to the category labeled "posttranslational modification/protein turnover/chaperones", and as anticipated, these genes were induced at the higher temperature, providing additional support to our microarray analysis (Fig. 3). The heat shock protein result was confirmed by analyzing Hsp70 protein quantity by Western blotting, which showed significantly more Hsp70 in bacteria shifted to 37°C versus F. tularensis LVS cultured at 26°C (data not shown). COG categories found only among the up-regulated transcripts included genes functioning in secretion and cell division, suggesting that host temperature may trigger these cellular processes and reflect, in part, enhanced growth rate (Fig. 3). In addition, the COG for bacterial defense mechanisms, which includes type I site-specific restriction-modification systems, was uniquely down-regulated. This response may have evolved to allow this bacterium to better contend with environmental stresses, such as invading bacteriophages. We observed that a large percentage of both up- and down-regulated genes (44% and 64% respectively) did not belong to a known COG category (Fig. 3). Proteins with an unknown COG category may have a novel biological role in F. tularensis associated with their temperature regulation.

Figure 3
figure 3

COG analysis of genes up- or down-regulated by mammalian temperature. COG categories were identified for each significantly up- or down-regulated gene based on the genomic annotation (accession, NC_07880). Specific COG categories were consolidated into general categories as follows: J, K, L, and RNA genes were combined into "Transcription/translation/DNA replication/RNA"; P, C, G, E, F, H, I, and Q were merged into "Transport and metabolism"; categories N and U were combined into "Cellular functions/trafficking/secretion"; and categories R, S, and uncategorized genes were classified as "Unknown". All other COG categories were reported as they were originally assigned.

F. tularensisLVS temperature regulation of genes necessary for infection

We hypothesized that the shift from environmental to mammalian temperature may be used to regulate genes important for infection. Surprisingly, none of the genes encoded in the FPI were significantly up-regulated at 37°C in the microarray analysis. We also confirmed, by immunoblotting, that IglC protein levels were equivalent in bacteria grown at the two different temperatures (data not shown). Upon further scrutiny, we did notice that one gene in the FPI, pdpC (FTL_0116; FTL_1162), was near the statistical threshold for induction at 37°C. We therefore analyzed pdpC transcript levels by Q-PCR as before, which confirmed that this gene was induced at mammalian body temperature (Fig. 2A).

We next compared our list of genes induced at 37°C with those previously shown to be necessary for Francisella infection, or postulated to be involved in the pathogenesis of this organism (Table 2). Forty percent of the protein-coding genes significantly up-regulated at 37°C have been reported or predicted to be important for intracellular growth and/or virulence of F. tularensis (Table 2). The list depicted in Table 2 included an assortment of metabolic genes, chaperones, genes encoding hypothetical proteins, and others. The data from Table 2 and the pdpC expression results in Fig. 2A further supported our hypothesis that genes important for Francisella infection are regulated by mammalian body temperature.

Table 2 List of genes induced at 37°C shown or implicated to be associated with intracellular growth and/or virulence of F. tularensis.

FTL_1581 is associated with F. tularensisLVS virulence

FTL_1581, annotated as a hypothetical lipoprotein, was induced by mammalian temperature more profoundly than any other hypothetical protein gene in the F. tularensis LVS genome (based on both fold change and J5 score; Additional file 1). We selected this gene for further study to test our hypothesis that temperature regulates genes necessary for infection. A BLAST search [32] against the National Center for Biotechnology Information database of non-redundant sequences did not identify proteins from organisms other than Francisella that had >30% identity to FTL_1581 (data not shown). This suggested that FTL_1581 may have a function unique to F. tularensis. The primary structure of FTL_1581 was further examined by a PROSITE analysis [33] which indicated this protein likely contained a lipoprotein signal sequence (Fig. 4A) consistent with its annotation. This analysis also revealed that the signal sequence overlapped with a motif similar to that of the Enterobacterial TraT complement resistance protein (Fig. 4A). Although the initial BLAST search did not retrieve homologous proteins with high degrees of identity, it did show that FTL_1581 contained a domain with 20–24% identity and 40–44% similarity with the vacuolating cytotoxin (VacA; jhp0819) [34, 35] and paralogs (jhp0556, jhp0856) of Helicobacter pylori J99 (Fig. 4A). Because FTL_1581 contained domains similar to other proteins that had a role in pathogenesis, we hypothesized that this protein may contribute to the virulence of F. tularensis.

Figure 4
figure 4

FTL_1581 contributes to F. tularensis LVS virulence. (A) A schematic of FTL_1581 protein is depicted showing domains predicted by PROSITE. (B) Competition studies in the chicken embryo infection model using a mixture of LVS and 1581d (1:1 based on OD600; viable bacteria in the inoculum were quantified by diluting and plating the input). Competition ratios (1581d: LVS) were normalized to the input to account for differences in the inoculum. These ratios were analyzed for statistical significance by Chi square; P < 0.001 at days 1, 2, and 3 post-infection. Data are mean competition ratio ± SEM of three embryos per time point within one experiment and are representative of duplicate experiments. (C) Macrophages were infected with LVS, 1581d, or 1581d/pFTL_1581 and were lysed at the indicated times. Data are mean ± SEM of triplicate wells within one experiment and are representative of four experiments performed using cells from separate donors. Following log transformation, differences in CFU were determined by a Student's t-Test in which P = 0.003 at 24 h.

We disrupted FTL_1581 in LVS, producing strain 1581d, to determine if this gene was associated with F. tularensis virulence. To assess the virulence of 1581d, we utilized a competition assay based on the chicken embryo infection model [36–38]. The chicken embryo produces a robust innate immune response comprised of complement, phagocytic cells, and cytokine production [39, 40]. Here, chicken embryos were infected with a ~1:1 mixture of LVS and 1581d. At days 1, 2, and 3 post-infection, wild-type LVS exhibited superior survival compared to 1581d (P < 0.001 at each time point) (Fig. 4B). This result was not due to a growth defect since 1581d grew identically to wild-type LVS when cultivated in bacterial growth medium (data not shown). In a separate experiment in which chicken embryos were infected only with 1581d, isolates from homogenates were all resistant to hygromycin, indicating that this mutant did not revert to wild type during infection (data not shown). Since 1581d was attenuated compared to LVS, the results suggest the function of the FTL_1581 gene product contributes to F. tularensis virulence.

We wanted to determine if the virulence attenuation of 1581d in the chicken embryo infection model was due to a reduced ability to inactivate complement, as FTL_1581 contained a putative complement resistance domain (Fig. 4A). Therefore, we subjected LVS and 1581d to serum sensitivity assays. There was equivalent survival of wild-type LVS and 1581d when cultured in media containing 20% serum or 20% heat-inactivated serum for up to 20 h (data not shown). To ensure that the serum complement was functional, E. coli DH5α was used as a control. Here, the CFU from the serum-treated E. coli exhibited a reduction of 3 logs relative to the input or heat-inactivated serum groups after 30 min (data not shown). This suggested that the disruption in 1581d does not affect complement resistance.

We determined if FTL_1581 contributed to growth in a macrophage environment. Primary human monocyte-derived macrophages were infected in vitro with either LVS or 1581d. At various time-points, macrophages were lysed, and the lysates were diluted and plated to enumerate viable CFU. We observed attenuated growth of 1581d in macrophages at 24 h post-infection (P = 0.003) (Fig. 4C). When FTL_1581 was complemented in trans in 1581d, wild-type level of growth was restored (Fig. 4C). This complementation confirmed that the reduced intracellular fitness of 1581d was due to the inactivation of FTL_1581, and not due to polar effects or alternate mutations. This experiment (Fig. 4C) also suggested that the attenuation of 1581d in the chicken embryo model (Fig. 4B) was likely due to a defect in intracellular survival. Together, the data presented here implicate FTL_1581, a Francisella ORF induced by mammalian body temperature that has no obvious homologs, with virulence and intracellular survival. Therefore, we propose that FTL_1581 be named temperature-induced, virulence-associated locus A, or tivA.

Involvement of FTL_1664 (deoB) in uptake of F. tularensisLVS

Many of the genes critical for Francisella infection involved metabolism (Table 2), a major COG category induced at 37°C (Fig. 3). This suggested that physiology vital for the success of F. tularensis as a pathogen was regulated by the temperature shift. Therefore, we were interested in determining the contribution of temperature-regulated metabolic genes toward Francisella pathogenesis. Previously, a microarray-based negative selection screen of F. novicida transposon mutants identified the phosphopentomutase, deoB, as a gene contributing to growth and/or survival in mice [14]. In other bacteria, proteins encoded by deoB homologs normally catalyze the reversible reaction between ribose-1-phosphate and ribose-5-phosphate or between deoxyribose-1-phosphate and deoxyribose-5-phosphate [41, 42]. An F. tularensis LVS chromosomal disruption mutant of deoB, FTL_1664, was constructed (strain 1664d). This mutant had a cellular and colony morphology similar to its wild-type parent strain (data not shown). Also, 1664d grew similarly to wild-type LVS in bacterial culture medium (data not shown).

We employed the chicken embryo infection model [36–38] to confirm that deoB contributes to LVS pathogenesis, as it did in F. novicida [14]. Here, chicken embryos that had been infected with F. tularensis LVS 1664d exhibited significantly enhanced survival compared to those infected with wild-type LVS over a 5 day period (P = 0.0254) (data not shown) corroborating the deoB data from F. novicida [14].

Accessing the host cytoplasm to replicate intracellularly is a hallmark of Francisella pathogenesis. Therefore, we next tested if the virulence attenuation of 1664d in the chicken embryo infection model was due to a defect in entering host cells. At two hours post-infection in vitro, human, monocyte-derived macrophages were treated with gentamicin to kill extracellular bacteria, followed by extensive washing. We consistently observed that substantially fewer 1664d cells were phagocytosed relative to wild-type LVS (Fig. 5A) (P = 0.00005). Importantly, trans complementation of 1664d (1664d/pFTL_1664) rescued the uptake defect (Fig. 5A). This suggested deoB is involved in a bacterial mechanism that enhances uptake of F. tularensis. This finding was extended by conducting similar uptake assays in primary human dendritic cells (Fig. 5A). In both phagocytic cell types, the uptake defect of 1664d was reproduced and complemented (Fig. 5A).

Figure 5
figure 5

deoB is important for entry of F. tularensis LVS. (A) Primary human macrophages (MΦs) or dendritic cells (DCs) were infected with either LVS, 1664d, or 1664d/pFTL_1664 in microtitre plates. After a two-hour incubation, wells were treated with gentamicin to kill extracellular bacteria, followed by vigorous washing. Subsequently, phagocytic cells were lysed and serial dilutions of lysates were plated for CFU enumeration. Data are mean ± SEM of triplicate wells within one experiment and are representative of four (primary human macrophages) or two experiments (dendritic cells) performed using cells from separate donors. Following log transformation, differences in CFU between LVS and 1664d were determined by a Student's t-Test in which P = 0.00005 and 0.003 for MΦs and DCs, respectively. (B) Bacteria were stained with the fluorescent green stain, Syto-9 prior to infection. After a two-hour incubation with RAW 264.7 cells, extracellular bacteria were washed away and cells were analyzed under brightfield (BF) and fluorescence (Fluor.) microscopy. The exposure time was extended to enhance sensitivity of detecting the low numbers of 1664d within the RAW 264.7 cells, leading to some background fluorescence. Fluorescence images were initially captured in grayscale and pseudocolored using Adobe Photoshop. Data displayed are representative of duplicate experiments. Scale bar = 50 μm. (C) Human embryonic kidney 293 (HEK-293) cells were infected similarly to the phagocytic cells in panel A. Data are mean ± SEM of triplicate wells within one experiment and are representative of three experiments. Following log transformation, differences in CFU between LVS and 1664d were determined by a Student's t-Test in which P = 0.0004.

We then qualitatively assessed uptake into phagocytes by microscopy to confirm the quantitative results obtained with CFU measurements. LVS, 1664d, and 1664d/pFTL_1664 were treated with the green fluorescent stain, Syto-9, and then incubated with RAW 264.7 cells, a murine macrophage-like cell line. After a two hour incubation and washes, cells were observed by fluorescence microscopy. Here, RAW 264.7 cells infected with LVS or 1664d/pFTL_1664 exhibited bright green fluorescence (Fig. 5B). In contrast, the RAW 264.7 cells infected with 1664d produced considerably less fluorescence despite comparable inocula to the cultures (Fig. 5B). The results obtained by microscopy suggested the discrepancy in CFU between wildtype and 1664d was not due to killing in the early phagosome. Rather, the levels of fluorescence in Fig. 5B were consistent with the CFU uptake data presented in Fig. 5A and confirmed that the temperature-regulated deoB is important for optimal uptake by mammalian phagocytes.

To determine if deoB contributed to entry into non-phagocytic cells, we exposed the human embryonic kidney cell line HEK-293 to either LVS, 1664d, or 1664d/pFTL_1664. At two hours post-infection, these cells were treated with gentamicin to kill extracellular bacteria followed by extensive washing. Subsequently, HEK-293 cells were lysed and the lysates were diluted and plated to enumerate CFU. Here we observed that 1664d showed reduced entry into the non-phagocytic HEK-293 cells (Fig. 5C). The uptake defect was again rescued by trans complementation (Fig. 5C). These data suggest that deoB contributes significantly to entry into both phagocytic (Fig. 5A and 5B) and non-phagocytic cells (Fig. 5C).

Discussion

In this paper, we provide evidence that F. tularensis LVS undergoes significant gene expression changes in response to mammalian body temperature (Fig. 1 and 2, Table 2, and Additional files 1 and 2). We hypothesized that genes important for infection of mammals may be induced during transition to this higher temperature. Although some genes, such as ribosomal RNA and tRNA genes, may reflect an increased growth rate at 37°C, our overall hypothesis was supported by the finding that 40% of the protein coding genes induced at 37°C have been shown or implicated to be important for successful Francisella infection (Table 2). In addition, we showed that two specific genes induced at 37°C contribute to the fitness of F. tularensis LVS during infection. While infection of a mammalian host exerts a complex set of signals on F. tularensis in addition to temperature, it is likely that many of the unexplored genes induced at mammalian temperature have a role in pathogenesis of F. tularensis. Differential responses to temperature among virulent and less virulent F. tularensis strains will also provide a focused list of relevant candidate virulence determinants to investigate further. The products of temperature-regulated genes with central roles in physiology and virulence could be targets for novel therapeutics or mutation to generate defined live attenuated vaccines.

One gene induced at 37°C, tivA (FTL_1581), encodes a protein with little overall identity to other proteins. It does, however, contain regions of meager similarity to the domains of proteins involved in the pathogenesis of other bacteria (Fig. 4A). We showed that this gene was necessary for full F. tularensis LVS virulence in the chicken embryo model, most likely because it contributed to optimal replication in primary human macrophages (Fig. 4B and 4C). In a previous high-throughput microarray-based, negative-selection screen for F. novicida genes important for a murine infection, the tivA allele of F. novicida (FTN_0573) was not identified [14]. There are two possible explanations for this discrepancy. There may be intrinsic differences between F. tularensis LVS and F. novicida pathogenesis [43] and their reliance on tivA. A more probable explanation is that F. tularensis LVS and the more pathogenic F. tularensis Schu S4 contain a single copy of this gene. In contrast, F. novicida has two homologs of tivA in its genome, suggesting the possibility of functional redundancy. These alternate forms (FTN_1103, 52% identity; FTN_1101, 49% identity) may have compensated for a mutation of tivA (FTN_0573) during the negative-selection screen [14], thereby masking any effects on pathogenesis. This possibility underscores the value of analyzing several different strains when probing F. tularensis pathogenesis.

Strain 1664d, a disruption mutant of deoB, exhibited significant reduction in uptake into mammalian cells (Fig. 5) indicating that this gene's product is involved in a mechanism that enhances entry. It would be advantageous for Francisella to couple expression of a gene important for uptake (deoB) with mammalian temperature, a cue encountered early in infection. Augmenting entry would improve access to the intracellular environment in which F. tularensis proliferates [44]. A previous study showed that pyrB (FTT1665) from F. tularensis Schu S4 was important for invasion in the human HepG2 hepatocellular carcinoma line [12]. However, a Schu S4 mutant of pyrB was not defective in uptake by J774.1 macrophage-like cells [12], suggesting that the Francisella mechanisms of optimal uptake by phagocytes and invasion into hepatocytes are separate phenomena. In another prior study, mutants of MglA, a key regulator of the FPI [45], and mutants of six genes controlled by this protein were not defective for cell entry [46]. This suggests that Francisella has evolved separate regulatory mechanisms for enhancing uptake and for intracellular survival. However, based on our data, genes critical for both phenomena are affected by mammalian temperature indicating that this cue is an important signal for multiple regulatory networks in Francisella. The uptake defect we have shown with the deoB mutant in both phagocytic and non-phagocytic cells has not been described previously.

In our system, DeoB may have a direct or indirect role in the uptake of Francisella. DeoB may influence the LPS structure of Francisella, secondarily improving interactions with host cell receptors. Importantly, complement receptors and mannose receptors are crucial for optimal Francisella phagocytosis [47–50]. This model is consistent with the fact that deoB is induced at 37°C and the finding that Francisella LPS structure is different at lower temperature versus mammalian temperature [29, 30]. Alternatively, DeoB may directly promote Francisella entry into host cells as this protein may have an additional function aside from being a phosphopentomutase. Another possibility is that mutation of metabolic genes in pathogenic bacteria can yield pleiotropic effects, resulting in defects in virulence mechanisms, including invasion [12]. Further investigation is necessary to determine the precise mechanism of Francisella DeoB in host cell entry.

The data presented here suggest that some genes important for Francisella during infection are induced by mammalian temperature. Although we showed by Q-PCR that pdpC transcripts were induced at 37°C, none of the other loci in the FPI [6] were up-regulated at mammalian body temperature. This is consistent with other findings suggesting that IglC protein levels were not induced by a shift from 37°C to 42°C [7]. Because many of the loci in the FPI are essential for intra-amoeba growth [45], which would occur at lower, ambient temperatures, it seems logical that these genes are not regulated by temperature. Therefore, the virulence associated-genes induced by mammalian temperature that we have identified are on a separate regulon than most of the genes of the FPI. This suggests Francisella possesses an intricate regulatory circuit to maximize its success in diverse environments.

Conclusion

F. tularensis LVS undergoes significant gene expression changes in response to mammalian temperature. This temperature shift is important for the regulation of pathogenesis. Our study characterizes a previously underappreciated environmental cue that regulates the expression of F. tularensis genes associated with virulence in other studies. Importantly, the collection of temperature-regulated genes also defines a rich set of novel candidate virulence determinants, including tivA (FTL_1581). Detailed investigation of tivA and deoB (FTL_1664) revealed unknown or unrecognized roles of these genes in intracellular survival and entry into mammalian cells, respectively.

Methods

Bacterial strains and growth conditions

Bacterial strains used in this study can be found in Table 1. All broth cultures were grown with agitation (250 rpm). For general cultivation of Escherichia coli, bacteria were grown at 37°C on LB agar plates, or in LB broth. F. tularensis LVS, a model organism for tularaemia, was used in this study. For F. tularensis LVS strains, frozen stock cultures were streaked onto chocolate II agar plates and incubated at 37°C, 5% CO2 for 2–4 days. These bacteria were subsequently used to inoculate broth cultures. For experiments assessing the effect of temperature on transcript levels, Chamberlain's chemically defined broth medium (CDM) [51] was inoculated with LVS and incubated at 37°C overnight. This start-up culture was used to inoculate fresh CDM (2 ml start-up culture into 25 ml fresh broth) and incubated at 26°C for 24 h. Following this incubation, the optical density (OD600) of this culture was recorded and RNA was extracted. For the temperature shift, 5 ml of the same culture was diluted into 25 ml fresh CDM and incubated at 37°C until attaining an OD600 comparable to the 26°C culture (typically ~6 h). When the 37°C culture reached the desired OD600, RNA was harvested. This strategy of growing bacteria to comparable OD600 prior to RNA extraction was similar to a previous study assessing global temperature regulation in Group A Streptococcus [19] and mitigates the effects of growth phase on the subsequent analysis.

For macrophage and chicken embryo infections, LVS strains were grown in TSBc (trypticase soy broth [Becton, Dickinson and Company] supplemented with 0.1% L-cysteine hydrochloride monohydrate) at 37°C while shaking at 250 rpm. When required, antibiotics were added to the media at the following concentrations: ampicillin at 150 μg/ml for E. coli; kanamycin at 35 μg/ml for E. coli and 10 μg/ml for F. tularensis LVS; chloramphenicol at 5 μg/ml for F. tularensis LVS; polymixin at 100 μg/ml; and hygromycin at 200 μg/ml.

The rationale for using CDM for the transcriptional analysis, while TSBc was used for all other experiments is two-fold. Using CDM for general cultivation of LVS is not practical because CDM is an aqueous mixture of 22 nutrients, some of which do not have a long shelf-life. Secondly, CDM was selected for transcriptional analyses to benefit future studies to analyze synergistic effects of temperature and media components.

RNA extraction

Immediately upon removal from the shaking incubator, six ml of broth culture were mixed with 18 ml TriReagent LS (Molecular Research Center). Chloroform (4.8 ml) was added to this material, and the aqueous phase was subsequently separated by centrifugation in a Phase Lock Heavy tube (Eppendorf). RNA was precipitated from this aqueous layer with isopropanol, followed by centrifugation. Pelleted material was washed in 80% ethanol and resuspended in nuclease-free water. This RNA-containing mixture was treated with DNase (Turbo DNA-free, Ambion), and then precipitated using ammonium acetate and ethanol. RNA quantity was measured spectrophotometrically, and quality was assessed using an Agilent Bioanalyzer.

Microarray analysis

Custom Agilent Francisella microarrays designed using the eArray framework were used in this study. All predicted open reading frames, including pseudogenes, were included for F. tularensis subsp. holarctica LVS, F. tularensis subsp. tularensis (SchuS4), F. tularensis subsp. holarctica OSU18, F. novicida U112 and the Francisella plasmids, pOM1, and pFNL10. Individual genes from LVS, SchuS4, and OSU18 were spotted in duplicate on this array, whereas genes from the other strains and plasmids were printed as single copies. cDNA was synthesized from RNA by reactions using random hexamers (Invitrogen) and MMLV (Agilent). The cDNA target was labelled with Alexa Fluor 555 conjugated dUTP according to manufacturer's protocol (AF555-aha-dUTP; Invitrogen). Labelling reactions were incubated at 40°C for 3 hours. After cDNA synthesis, RNA was hydrolyzed with with NaOH and EDTA, and subsequently, the pH was neutralized with an equivalent volume of HCl. The labelled cDNA was precipitated using isopropanol and ammonium acetate and washed with 80% ethanol. 0.5 μg of labelled cDNA was hybridized to custom Agilent 8 × 15 K microarrays according to the protocol of the manufacturer and incubated at 60°C for 18 hours in a rotary oven. Following hybridization, arrays were washed with Agilent wash buffers before being scanned on an Agilent microarray scanner. At least 3 separate RNA extractions were used for each condition tested by microarray (26°C or 37°C-grown bacteria).

Gene Expression Data Analyzer (GEDA; http://bioinformatics.upmc.edu/GE2/GEDA.html) was utilized for analysis of the microarray data [31]. This online software package was used for minimum mean ratio array normalization followed by log2 transformation. Data were grouped into two categories, cultivated at 26°C and shifted to 37°C, and analyzed for significance with the J5 statistical test [31]. The J5 metric was designed for data sets composed of limited replicates, thereby reducing the chance of generating false positives [31]. Genes were considered to be significantly differentially regulated if they had a J5 score greater than 2. Additional files 1 and 2 consist of lists showing both the J5 value and fold change for genes that were significantly up- or down-regulated upon a shift to mammalian temperature. Normalized, log2 transformed intensity values for genes that were differentially expressed were input into GenePattern [52] and data are presented after hierarchical clustering (Pearson correlation).

Categories assigned for clusters of orthologous groups were identified for each significantly up- or down-regulated gene based on the annotation assigned from the F. tularensis holarctica genome (accession, NC_07880). Specific clusters of orthologous groups categories were consolidated into general categories as follows: J, K, L, and RNA genes were combined into "Transcription/translation/DNA replication/RNA"; P, C, G, E, F, H, I, and Q were merged into "Transport and metabolism"; categories N and U were combined into "Cellular functions/trafficking/secretion"; and categories R, S, and uncategorized genes were classified as "Unknown". All other clusters of orthologous groups categories were reported as they were originally assigned.

Quantitative real time PCR

One μg of total RNA was used as a template for cDNA synthesis catalyzed by Superscript III (Invitrogen). Diluted cDNA was used as a template for real time reactions containing primer sets (designed by Primer 3 [53] for the desired genes) and SYBR Green Supermix (BioRad). These reactions were carried out on a BioRad IQ5 real time machine. The bacterial 50S ribosomal protein L24 (FTL_0247) was used as an internal reference, as it was observed to have no change in expression following a shift to 37°C according to the microarray analysis (data not shown). The quantitative real time PCR data are presented as log2 transformed fold change values (37°C versus 26°C).

Construction of plasmids

Plasmids and primers used in this study can be found in Table 1. The disruption plasmids, pMQ131hyg1581d and pMQ131hyg1664d were constructed using the following procedures. First, the PvuI/SpeI fragment of pMP615 containing hyg under the control of the groEL promoter was subcloned into pMQ131 (R. Shanks and G. O'Toole, unpublished data) that had been digested with PvuI/XbaI which produced pMQ131hyg. The internal 560 base pairs of FTL_1581 were amplified by PCR using the primers 1581_560F and 1581_560R and this amplicon was initially TA cloned into pGEM-T (Promega). The KpnI/BamHI fragment of this construct containing the internal portion of FTL_1581 was subcloned into pMQ131hyg that had been digested with these same enzymes, which produced pMQ131hyg1581d. Similarly, the internal 900 base pairs of FTL_1664 were amplified by PCR using the primers 1664_900F and 1664_900R and this amplicon was initially TA-cloned into pGEM-T (Promega). Subsequently, this fragment internal to FLT_1664 was subcloned (KpnI/BamHI) into pMQ131hyg, producing the plasmid pMQ131hyg1664d.

The FTL_1581 complementing plasmid (pFTL_1581) was constructed using the following procedures. The primers 1581_clone_up and 1581_clone_down were used to amplify 600 base pairs upstream, the entire FTL_1581 open reading frame, and 100 base pairs downstream of this gene by PCR. This amplicon encompassed the predicted native FTL_1581 promoter, this gene's coding region, as well as the predicted transcriptional stop (data not shown). Initially, this amplicon was TA-cloned into pGEM-T (Promega). The SacI/SacII fragment of this construct containing FTL_1581 was subsequently subcloned into pMQ2 (R. Shanks and G. O'Toole, unpublished data) that had been digested with these same enzymes. This yielded pFTL_1581.

The FTL_1664 (deoB) complementing plasmid (pFTL_1664) was constructed using the following procedures. Initially, the KanR gene from pFNLTP8 was deleted using an inverse PCR strategy [54]. The primers F8AgeI and F8XhoI were used to amplify pFNLTP8. Following PCR, template DNA was digested with DpnI leaving only amplified product. This amplicon was self-ligated, producing pF8AX. The cat194 gene was amplified by PCR using the primers XhoICAT and AgeICAT with pMQ2 as a template, and this amplicon was TA-cloned into pGEM-T (Promega). The Age I/XhoI fragment of this construct that encoded the cat194 gene was subcloned into pF8AX using these same enzymes producing pF8CAT. 1664_clone_up and 1664_clone_down were used to amplify 600 base pairs upstream, the entire FTL_1664 open reading frame, and 100 base pairs downstream of this gene by PCR. This amplicon encompassed the predicted native FTL_1664 promoter, this gene's coding region, as well as the predicted transcriptional stop (data not shown). Initially, this amplicon was TA-cloned into pGEM-T(Promega). The BamHI/KpnI fragment of this construct containing FTL_1664 was subsequently subcloned into pF8CAT that had been digested with these same enzymes. This yielded pFTL_1664.

Construction of the FTL_1664 and FTL_1581 mutants

F. 0tularensis LVS disruption mutants of FTL_1664 and FTL_1581 were constructed as previously reported [55]. A homologous recombination between the LVS chromosomal copy of FTL_1581 and the internal FTL_1581 fragment in pMQ131hyg1581d would disrupt the coding sequence of 38 amino acids at the C-terminus. Also, homologous recombination between chromosomal FTL_1664 and pMQ131hyg1664d would disrupt coding sequence for the C-terminal 56 amino acids of the encoded protein. Disruption constructs (either pMQ131hyg1664d or pMQ131hyg1581d) were mobilized into F. tularensis LVS by triparental mating. Mating mixes were composed of LVS (109 CFU), E. coli/pRK2013 (107 CFU), and either E. coli/pMQ131hyg1664d or E. coli/pMQ131hyg1581d (107 CFU). Similarly to the previously described Francisella conjugation methodology [56], the mating mixture here was plated on LB agar plates, and incubated at room temperature for 18 hours. Cells were resuspended in phosphate buffered saline (PBS) and spread onto chocolate II agar plates containing polymyxin (100 μg/ml) and hygromycin. Hygromycin-resistant colonies were screened by PCR using primers specific for either FTL_1581 or FTL_1664 and the vector-portion of pMQ131hyg1581d or pMQ131hyg1664d. The F. tularensis LVS FTL_1664 and FTL_1581 disruption mutants (FTL_1664::pMQ131hyg1664d; FTL_1581::pMQ131hyg1581d) are referred to as strains 1664d and 1581d respectively. Mobilization of complementation constructs into the disruption mutants was accomplished by electroporation as previously described [57].

Complement resistance assay

Bacteria (1.5 × 109 CFU) were incubated in solutions of PBS with 20% serum (human serum [Gemini Bioscience]) or 20% heat-inactivated serum at 37°C while shaking at 250 rpm. Serum was incubated at 56°C for 30 minutes to inactivate complement. At certain time-points, cell suspensions were diluted and plated to enumerate CFU.

Chicken embryo infections

Chicken embryos were infected with F. tularensis LVS, 1664d, and 1581d as previously described [37, 38]. Whiteleghorn chicken eggs (fertilized, specific pathogen free) were purchased from Charles River Laboratories, North Franklin, CT, USA. Eggs were incubated at 37°C with gentle rocking and humidity for one week prior to infection. Following the initial 7 day incubation, those without a live embryo were discarded. For infections, the surface of the egg shell was sterilized with 70% ethanol. The egg shell membrane was exposed and removed after introducing a small hole in the shell. Bacteria suspended in PBS (100 μl) were then injected beneath the chorioallantoic membrane. The hole in the egg shell was covered with transparent tape and subsequently eggs were incubated as previously described. On a daily basis, eggs were candled to assess viability for 6 days. As previously reported [37], embryos that expired within 24 h of the infection were presumed to have experienced lethal trauma during inoculation, and were removed from the experiment. Survival differences in chicken embryo infections were analyzed with the log rank test in GraphPad Prism 5.

Competition studies in the chicken embryo infection model were conducted similar to the single infections described above. Here chicken embryos were infected with a mixture of LVS and 1581d (1:1 based on OD600, ~105 total bacteria). Actual input (6.5 × 105 total bacteria) was determined by diluting the inoculum followed by plating to enumerate viable bacteria. Three infected, viable eggs were sacrificed at 1, 2, and 3 days (9 total eggs) post infection. Egg contents were homogenized with an OMNI tissue homogenizer, and serial dilutions of the homogenates were plated on chocolate II agar plates with or without antibiotic. The total number of 1581d (plates with antibiotic) was subtracted from the number of total bacteria (plates without antibiotic) to determine the amount of viable wild-type LVS. Competition ratios (1581d: LVS) were normalized to the input to account for small differences in the inocula, as has been done previously [58, 59]. Competition ratios were analyzed for statistical significance by Chi square.

Mammalian cell infections

Human monocytes were differentiated into macrophages by in vitro culture as has been described previously [11, 57, 60]. Buffy coats from blood donations (Central Blood Bank, Pittsburgh, Pennsylvania) served as the source of monocytes. Monocytes were purified using Ficoll gradients (Amersham Biosciences) to isolate PBMCs, Optiprep gradients (Axis-Shield) to enrich for monocytes, and panning on plastic to further purify monocytes (final purity >95% based on microscopy). Cells were cultured in 60 mm culture dishes for 7 days at 37°C with 5% CO2 in 7 ml of DMEM (Invitrogen) supplemented with 20% FCS (Invitrogen), 10% human serum (Gemini Biosciences), 25 mM HEPES (Gibco), and 1% Glutamax (Invitrogen) [11, 57, 60]. Macrophages were detached from the culture dish on day 7 using a lidocaine/EDTA solution (5 mM EDTA and 4 mg/ml lidocaine in PBS pH 7.2). For monocyte-derived dendritic cells, human monocytes were seeded at a density of 1 × 106 cells/ml in 24 well plates (Costar) in complete RPMI [10% FCS, 25 mM HEPES, 1% non-essential amino acids, 1% sodium pyruvate, 1% Glutamax and 0.1% 2-mercaptoethanol (all from Gibco)] supplemented with 1000 U/ml GM-CSF and 1000 U/ml IL-4 (both from eBioscience) and incubated at 37°C with 5% CO2. On day 3 of culture, 10% of the media was replaced with fresh complete RPMI supplemented with GM-CSF and IL-4 and non-adherent cells were harvested at day 5. For in vitro infections, all primary cells were washed and resuspended in DMEM supplemented with 1% human serum, 25 mM HEPES, and 1% Glutamax and then plated onto Primaria-coated 96-well culture dishes (Becton, Dickinson and Company) at a density of 5.0 × 104 cells per well.

Murine macrophage-like RAW 264.7 cells (ATCC number TIB-71) were routinely cultured in DMEM supplemented with 10% fetal calf serum, 25 mM HEPES, and 1% Glutamax with 100 U/ml penicillin-streptomycin. Two days prior to infection, the medium for these cells was replaced with DMEM supplemented with 10% human serum, 25 mM HEPES, and 1% Glutamax, and infections were carried out using this same medium.

HEK-293 cells (ATCC number CRL-1573), a non-phagocytic cell line [61], were cultivated in DMEM supplemented with 10% fetal calf serum, 25 mM HEPES, and 1% Glutamax with 100 U/ml penicillin-streptomycin. One day prior to infection, HEK-293 cells were seeded in 96-well plates at a concentration of 5 × 104 cells per well in this same medium devoid of antibiotic and infections were carried out in this same medium.

For infection experiments, bacterial cultures were adjusted to an OD600 of 0.3 (approximately 1.5 × 109 CFU/ml) and diluted to attain a multiplicity of infection (MOI) of 500, which typically yields infection rate >80% after a 2 hour co-incubation [11, 62]. Alternatively, macrophages were suspended in DMEM supplemented with 10% human serum, 25 mM HEPES, and 1% Glutamax and transferred to culture dishes before infection. Prior to administration of the bacteria onto the macrophages, the F. tularensis strains were incubated at 37°C with 5% CO2 for 20 min in this same medium. Macrophages were infected with bacteria that had been diluted to an MOI of 40. This alternative infection protocol used a lower MOI because it took advantage of the complement-mediated uptake of F. tularensis into macrophages [47]. Comparable results were obtained regardless of which infection protocol was used. Actual MOIs were measured by plating serial dilutions of inocula on chocolate II agar plates.

For intracellular CFU enumeration, mammalian cells were exposed to the initial bacterial load for 2 hours at 37°C with 5% CO2 and then incubated with gentamicin (20 μg/ml) for 20 min to kill extracellular bacteria. The cells were washed with warm Hank's balanced salt solution and then incubated at 37°C with 5% CO2 for another 22–48 h with fresh culture media. At the indicated time-points post-infection, mammalian cells were lysed with 0.02% sodium dodecyl sulfate. The 2 hour time point was used to determine entry similarly to a prior study [46]. Serial dilutions of the lysates were plated on chocolate II agar plates for enumeration of viable bacteria. CFU counts were converted to log10 values and analyzed by the Student's t-Test to determine statistical differences. For uptake analysis, CFU were normalized to actual MOI to account for minor variations in the density of inocula [46].

For fluorescence microscopy, bacteria were stained with 10 μM Syto 9 (Invitrogen) for 15 min and washed three times with PBS prior to infection. Here, stained cells were diluted in DMEM supplemented with 10% human serum, 25 mM HEPES, and 1% Glutamax, and were incubated at 37°C with 5% CO2 for 20 min. These bacteria were used to inoculate RAW 264.7 in this same medium at an MOI of 10.

References

  1. Ellis J, Oyston PC, Green M, Titball RW: Tularemia. Clin Microbiol Rev. 2002, 15 (4): 631-646. 10.1128/CMR.15.4.631-646.2002.

    PubMed Central  PubMed  Google Scholar 

  2. Sjostedt A: Tularemia: history, epidemiology, pathogen physiology, and clinical manifestations. Ann N Y Acad Sci. 2007, 1105: 1-29. 10.1196/annals.1409.009.

    PubMed  Google Scholar 

  3. Dennis DT, Inglesby TV, Henderson DA, Bartlett JG, Ascher MS, Eitzen E, Fine AD, Friedlander AM, Hauer J, Layton M, et al: Tularemia as a biological weapon: medical and public health management. Jama. 2001, 285 (21): 2763-2773. 10.1001/jama.285.21.2763.

    CAS  PubMed  Google Scholar 

  4. Berdal BP, Mehl R, Meidell NK, Lorentzen-Styr AM, Scheel O: Field investigations of tularemia in Norway. FEMS Immunol Med Microbiol. 1996, 13 (3): 191-195. 10.1111/j.1574-695X.1996.tb00235.x.

    CAS  PubMed  Google Scholar 

  5. Oyston PC, Quarry JE: Tularemia vaccine: past, present and future. Antonie Van Leeuwenhoek. 2005, 87 (4): 277-281. 10.1007/s10482-004-6251-7.

    PubMed  Google Scholar 

  6. Nano FE, Schmerk C: The Francisella pathogenicity island. Ann N Y Acad Sci. 2007, 1105: 122-137. 10.1196/annals.1409.000.

    CAS  PubMed  Google Scholar 

  7. Golovliov I, Ericsson M, Sandstrom G, Tarnvik A, Sjostedt A: Identification of proteins of Francisella tularensis induced during growth in macrophages and cloning of the gene encoding a prominently induced 23-kilodalton protein. Infect Immun. 1997, 65 (6): 2183-2189.

    PubMed Central  CAS  PubMed  Google Scholar 

  8. Deng K, Blick RJ, Liu W, Hansen EJ: Identification of Francisella tularensis genes affected by iron limitation. Infect Immun. 2006, 74 (7): 4224-4236. 10.1128/IAI.01975-05.

    PubMed Central  CAS  PubMed  Google Scholar 

  9. Lenco J, Hubalek M, Larsson P, Fucikova A, Brychta M, Macela A, Stulik J: Proteomics analysis of the Francisella tularensis LVS response to iron restriction: induction of the F. tularensis pathogenicity island proteins IglABC. FEMS Microbiol Lett. 2007, 269 (1): 11-21. 10.1111/j.1574-6968.2006.00595.x.

    CAS  PubMed  Google Scholar 

  10. Sullivan JT, Jeffery EF, Shannon JD, Ramakrishnan G: Characterization of the siderophore of Francisella tularensis and role of fslA in siderophore production. J Bacteriol. 2006, 188 (11): 3785-3795. 10.1128/JB.00027-06.

    PubMed Central  CAS  PubMed  Google Scholar 

  11. Carlson PE, Carroll JA, O'Dee DM, Nau GJ: Modulation of virulence factors in Francisella tularensis determines human macrophage responses. Microb Pathog. 2007, 42 (5–6): 204-214. 10.1016/j.micpath.2007.02.001.

    PubMed Central  CAS  PubMed  Google Scholar 

  12. Qin A, Mann BJ: Identification of transposon insertion mutants of Francisella tularensis tularensis strain Schu S4 deficient in intracellular replication in the hepatic cell line HepG2. BMC Microbiol. 2006, 6: 69-10.1186/1471-2180-6-69.

    PubMed Central  PubMed  Google Scholar 

  13. Su J, Yang J, Zhao D, Kawula TH, Banas JA, Zhang JR: Genome-wide identification of Francisella tularensis virulence determinants. Infect Immun. 2007, 75 (6): 3089-3101. 10.1128/IAI.01865-06.

    PubMed Central  CAS  PubMed  Google Scholar 

  14. Weiss DS, Brotcke A, Henry T, Margolis JJ, Chan K, Monack DM: In vivo negative selection screen identifies genes required for Francisella virulence. Proc Natl Acad Sci USA. 2007, 104 (14): 6037-6042. 10.1073/pnas.0609675104.

    PubMed Central  CAS  PubMed  Google Scholar 

  15. Twine S, Bystrom M, Chen W, Forsman M, Golovliov I, Johansson A, Kelly J, Lindgren H, Svensson K, Zingmark C, et al: A mutant of Francisella tularensis strain SCHU S4 lacking the ability to express a 58-kilodalton protein is attenuated for virulence and is an effective live vaccine. Infect Immun. 2005, 73 (12): 8345-8352. 10.1128/IAI.73.12.8345-8352.2005.

    PubMed Central  CAS  PubMed  Google Scholar 

  16. Carroll JA, El-Hage N, Miller JC, Babb K, Stevenson B: Borrelia burgdorferi RevA antigen is a surface-exposed outer membrane protein whose expression is regulated in response to environmental temperature and pH. Infect Immun. 2001, 69 (9): 5286-5293. 10.1128/IAI.69.9.5286-5293.2001.

    PubMed Central  CAS  PubMed  Google Scholar 

  17. Miller JF, Mekalanos JJ, Falkow S: Coordinate regulation and sensory transduction in the control of bacterial virulence. Science. 1989, 243 (4893): 916-922. 10.1126/science.2537530.

    CAS  PubMed  Google Scholar 

  18. Ojaimi C, Brooks C, Casjens S, Rosa P, Elias A, Barbour A, Jasinskas A, Benach J, Katona L, Radolf J, et al: Profiling of temperature-induced changes in Borrelia burgdorferi gene expression by using whole genome arrays. Infect Immun. 2003, 71 (4): 1689-1705. 10.1128/IAI.71.4.1689-1705.2003.

    PubMed Central  CAS  PubMed  Google Scholar 

  19. Smoot LM, Smoot JC, Graham MR, Somerville GA, Sturdevant DE, Migliaccio CA, Sylva GL, Musser JM: Global differential gene expression in response to growth temperature alteration in group A Streptococcus. Proc Natl Acad Sci USA. 2001, 98 (18): 10416-10421. 10.1073/pnas.191267598.

    PubMed Central  CAS  PubMed  Google Scholar 

  20. Tobe T, Yoshikawa M, Sasakawa C: Thermoregulation of virB transcription in Shigella flexneri by sensing of changes in local DNA superhelicity. J Bacteriol. 1995, 177 (4): 1094-1097.

    PubMed Central  CAS  PubMed  Google Scholar 

  21. Hueck CJ: Type III protein secretion systems in bacterial pathogens of animals and plants. Microbiol Mol Biol Rev. 1998, 62 (2): 379-433.

    PubMed Central  CAS  PubMed  Google Scholar 

  22. Zuber U, Schumann W: CIRCE, a novel heat shock element involved in regulation of heat shock operon dnaK of Bacillus subtilis. J Bacteriol. 1994, 176 (5): 1359-1363.

    PubMed Central  CAS  PubMed  Google Scholar 

  23. Straus DB, Walter WA, Gross CA: The heat shock response of E. coli is regulated by changes in the concentration of sigma 32. Nature. 1987, 329 (6137): 348-351. 10.1038/329348a0.

    CAS  PubMed  Google Scholar 

  24. Konkel ME, Tilly K: Temperature-regulated expression of bacterial virulence genes. Microbes Infect. 2000, 2 (2): 157-166. 10.1016/S1286-4579(00)00272-0.

    CAS  PubMed  Google Scholar 

  25. Ericsson M, Golovliov I, Sandstrom G, Tarnvik A, Sjostedt A: Characterization of the nucleotide sequence of the groE operon encoding heat shock proteins chaperone-60 and -10 of Francisella tularensis and determination of the T-cell response to the proteins in individuals vaccinated with F. tularensis. Infect Immun. 1997, 65 (5): 1824-1829.

    PubMed Central  CAS  PubMed  Google Scholar 

  26. Ericsson M, Tarnvik A, Kuoppa K, Sandstrom G, Sjostedt A: Increased synthesis of DnaK, GroEL, and GroES homologs by Francisella tularensis LVS in response to heat and hydrogen peroxide. Infect Immun. 1994, 62 (1): 178-183.

    PubMed Central  CAS  PubMed  Google Scholar 

  27. Meibom KL, Dubail I, Dupuis M, Barel M, Lenco J, Stulik J, Golovliov I, Sjostedt A, Charbit A: The heat-shock protein ClpB of Francisella tularensis is involved in stress tolerance and is required for multiplication in target organs of infected mice. Mol Microbiol. 2008, 67 (6): 1384-1401. 10.1111/j.1365-2958.2008.06139.x.

    CAS  PubMed  Google Scholar 

  28. Bhatnagar NB, Elkins KL, Fortier AH: Heat stress alters the virulence of a rifampin-resistant mutant of Francisella tularensis LVS. Infect Immun. 1995, 63 (1): 154-159.

    PubMed Central  CAS  PubMed  Google Scholar 

  29. Shaffer SA, Harvey MD, Goodlett DR, Ernst RK: Structural heterogeneity and environmentally regulated remodeling of Francisella tularensis subspecies novicida lipid A characterized by tandem mass spectrometry. J Am Soc Mass Spectrom. 2007, 18 (6): 1080-1092. 10.1016/j.jasms.2007.03.008.

    PubMed Central  CAS  PubMed  Google Scholar 

  30. Mohapatra NP, Balagopal A, Soni S, Schlesinger LS, Gunn JS: AcpA is a Francisella acid phosphatase that affects intramacrophage survival and virulence. Infect Immun. 2007, 75 (1): 390-396. 10.1128/IAI.01226-06.

    PubMed Central  CAS  PubMed  Google Scholar 

  31. Patel S, Lyons-Weiler J: caGEDA: a web application for the integrated analysis of global gene expression patterns in cancer. Appl Bioinformatics. 2004, 3 (1): 49-62.

    CAS  PubMed  Google Scholar 

  32. Altschul SF, Madden TL, Schaffer AA, Zhang J, Zhang Z, Miller W, Lipman DJ: Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Res. 1997, 25 (17): 3389-3402. 10.1093/nar/25.17.3389.

    PubMed Central  CAS  PubMed  Google Scholar 

  33. Falquet L, Pagni M, Bucher P, Hulo N, Sigrist CJ, Hofmann K, Bairoch A: The PROSITE database, its status in 2002. Nucleic Acids Res. 2002, 30 (1): 235-238. 10.1093/nar/30.1.235.

    PubMed Central  CAS  PubMed  Google Scholar 

  34. Blaser MJ: Role of vacA and the cagA locus of Helicobacter pylori in human disease. Aliment Pharmacol Ther. 1996, 10 (Suppl 1): 73-77.

    CAS  PubMed  Google Scholar 

  35. Xiang Z, Censini S, Bayeli PF, Telford JL, Figura N, Rappuoli R, Covacci A: Analysis of expression of CagA and VacA virulence factors in 43 strains of Helicobacter pylori reveals that clinical isolates can be divided into two major types and that CagA is not necessary for expression of the vacuolating cytotoxin. Infect Immun. 1995, 63 (1): 94-98.

    PubMed Central  CAS  PubMed  Google Scholar 

  36. Townsend MK, Carr NJ, Iyer JG, Horne SM, Gibbs PS, Pruss BM: Pleiotropic phenotypes of a Yersinia enterocolitica flhD mutant include reduced lethality in a chicken embryo model. BMC Microbiol. 2008, 8: 12-10.1186/1471-2180-8-12.

    PubMed Central  PubMed  Google Scholar 

  37. Nix EB, Cheung KK, Wang D, Zhang N, Burke RD, Nano FE: Virulence of Francisella spp. in chicken embryos. Infect Immun. 2006, 74 (8): 4809-4816. 10.1128/IAI.00034-06.

    PubMed Central  CAS  PubMed  Google Scholar 

  38. de Bruin OM, Ludu JS, Nano FE: The Francisella pathogenicity island protein IglA localizes to the bacterial cytoplasm and is needed for intracellular growth. BMC Microbiol. 2007, 7: 1-10.1186/1471-2180-7-1.

    PubMed Central  PubMed  Google Scholar 

  39. Herren CD, Mitra A, Palaniyandi SK, Coleman A, Elankumaran S, Mukhopadhyay S: The BarA-UvrY two-component system regulates virulence in avian pathogenic Escherichia coli O78:K80:H9. Infect Immun. 2006, 74 (8): 4900-4909. 10.1128/IAI.00412-06.

    PubMed Central  CAS  PubMed  Google Scholar 

  40. Siatskas C, Boyd R: Regulation of chicken haemopoiesis by cytokines. Dev Comp Immunol. 2000, 24 (1): 37-59. 10.1016/S0145-305X(99)00051-8.

    CAS  PubMed  Google Scholar 

  41. Tozzi MG, Camici M, Mascia L, Sgarrella F, Ipata PL: Pentose phosphates in nucleoside interconversion and catabolism. Febs J. 2006, 273 (6): 1089-1101. 10.1111/j.1742-4658.2006.05155.x.

    CAS  PubMed  Google Scholar 

  42. Hammer-Jespersen K, Munch-Petersen A: Phosphodeoxyribomutase from Escherichia coli. Purification and some properties. Eur J Biochem. 1970, 17 (3): 397-407. 10.1111/j.1432-1033.1970.tb01179.x.

    CAS  PubMed  Google Scholar 

  43. Titball RW, Petrosino JF: Francisella tularensis genomics and proteomics. Ann N Y Acad Sci. 2007, 1105: 98-121. 10.1196/annals.1409.015.

    CAS  PubMed  Google Scholar 

  44. Sjostedt A: Intracellular survival mechanisms of Francisella tularensis, a stealth pathogen. Microbes Infect. 2006, 8 (2): 561-567. 10.1016/j.micinf.2005.08.001.

    PubMed  Google Scholar 

  45. Lauriano CM, Barker JR, Yoon SS, Nano FE, Arulanandam BP, Hassett DJ, Klose KE: MglA regulates transcription of virulence factors necessary for Francisella tularensis intraamoebae and intramacrophage survival. Proc Natl Acad Sci USA. 2004, 101 (12): 4246-4249. 10.1073/pnas.0307690101.

    PubMed Central  CAS  PubMed  Google Scholar 

  46. Brotcke A, Weiss DS, Kim CC, Chain P, Malfatti S, Garcia E, Monack DM: Identification of MglA-regulated genes reveals novel virulence factors in Francisella tularensis. Infect Immun. 2006, 74 (12): 6642-6655. 10.1128/IAI.01250-06.

    PubMed Central  CAS  PubMed  Google Scholar 

  47. Clemens DL, Lee BY, Horwitz MA: Francisella tularensis enters macrophages via a novel process involving pseudopod loops. Infect Immun. 2005, 73 (9): 5892-5902. 10.1128/IAI.73.9.5892-5902.2005.

    PubMed Central  CAS  PubMed  Google Scholar 

  48. Ben Nasr A, Haithcoat J, Masterson JE, Gunn JS, Eaves-Pyles T, Klimpel GR: Critical role for serum opsonins and complement receptors CR3 (CD11b/CD18) and CR4 (CD11c/CD18) in phagocytosis of Francisella tularensis by human dendritic cells (DC): uptake of Francisella leads to activation of immature DC and intracellular survival of the bacteria. J Leukoc Biol. 2006, 80 (4): 774-786. 10.1189/jlb.1205755.

    CAS  PubMed  Google Scholar 

  49. Schulert GS, Allen LA: Differential infection of mononuclear phagocytes by Francisella tularensis: role of the macrophage mannose receptor. J Leukoc Biol. 2006, 80 (3): 563-571. 10.1189/jlb.0306219.

    PubMed Central  CAS  PubMed  Google Scholar 

  50. Balagopal A, MacFarlane AS, Mohapatra N, Soni S, Gunn JS, Schlesinger LS: Characterization of the receptor-ligand pathways important for entry and survival of Francisella tularensis in human macrophages. Infect Immun. 2006, 74 (9): 5114-5125. 10.1128/IAI.00795-06.

    PubMed Central  CAS  PubMed  Google Scholar 

  51. Chamberlain RE: Evaluation of Live Tularemia Vaccine Prepared in a Chemically Defined Medium. Appl Microbiol. 1965, 13: 232-235.

    PubMed Central  CAS  PubMed  Google Scholar 

  52. Golub TR, Slonim DK, Tamayo P, Huard C, Gaasenbeek M, Mesirov JP, Coller H, Loh ML, Downing JR, Caligiuri MA, et al: Molecular classification of cancer: class discovery and class prediction by gene expression monitoring. Science. 1999, 286 (5439): 531-537. 10.1126/science.286.5439.531.

    CAS  PubMed  Google Scholar 

  53. Rozen S, Skaletsky H: Primer3 on the WWW for general users and for biologist programmers. Methods Mol Biol. 2000, 132: 365-386.

    CAS  PubMed  Google Scholar 

  54. Horzempa J, Comer JE, Davis SA, Castric P: Glycosylation substrate specificity of Pseudomonas aeruginosa 1244 pilin. J Biol Chem. 2006, 281 (2): 1128-1136. 10.1074/jbc.M510975200.

    PubMed Central  CAS  PubMed  Google Scholar 

  55. Shanks RM, Caiazza NC, Hinsa SM, Toutain CM, O'Toole GA: Saccharomyces cerevisiae-based molecular tool kit for manipulation of genes from gram-negative bacteria. Appl Environ Microbiol. 2006, 72 (7): 5027-5036. 10.1128/AEM.00682-06.

    PubMed Central  CAS  PubMed  Google Scholar 

  56. Golovliov I, Sjostedt A, Mokrievich A, Pavlov V: A method for allelic replacement in Francisella tularensis. FEMS Microbiol Lett. 2003, 222 (2): 273-280.

    CAS  PubMed  Google Scholar 

  57. Horzempa J, Tarwacki DM, Carlson PE, Robinson CM, Nau GJ: Characterization and application of a glucose-repressible promoter in Francisella tularensis. Appl Environ Microbiol. 2008, 74 (7): 2161-2170. 10.1128/AEM.02360-07.

    PubMed Central  CAS  PubMed  Google Scholar 

  58. Edelstein PH, Hu B, Shinzato T, Edelstein MA, Xu W, Bessman MJ: Legionella pneumophila NudA Is a Nudix hydrolase and virulence factor. Infect Immun. 2005, 73 (10): 6567-6576. 10.1128/IAI.73.10.6567-6576.2005.

    PubMed Central  CAS  PubMed  Google Scholar 

  59. Smedley JG, Jewell E, Roguskie J, Horzempa J, Syboldt A, Stolz DB, Castric P: Influence of pilin glycosylation on Pseudomonas aeruginosa 1244 pilus function. Infect Immun. 2005, 73 (12): 7922-7931. 10.1128/IAI.73.12.7922-7931.2005.

    PubMed Central  CAS  PubMed  Google Scholar 

  60. Robinson CM, Nau GJ: Interleukin-12 and Interleukin-27 Regulate Macrophage Control of Mycobacterium tuberculosis. J Infect Dis. 2008, 198 (3): 359-366. 10.1086/589774.

    PubMed Central  CAS  PubMed  Google Scholar 

  61. Tachado SD, Zhang J, Zhu J, Patel N, Cushion M, Koziel H: Pneumocystis-mediated IL-8 release by macrophages requires coexpression of mannose receptors and TLR2. J Leukoc Biol. 2007, 81 (1): 205-211. 10.1189/jlb.1005580.

    CAS  PubMed  Google Scholar 

  62. Lai XH, Golovliov I, Sjostedt A: Expression of IglC is necessary for intracellular growth and induction of apoptosis in murine macrophages by Francisella tularensis. Microb Pathog. 2004, 37 (5): 225-230.

    CAS  PubMed  Google Scholar 

  63. Maier TM, Havig A, Casey M, Nano FE, Frank DW, Zahrt TC: Construction and characterization of a highly efficient Francisella shuttle plasmid. Appl Environ Microbiol. 2004, 70: 7511-7519. 10.1128/AEM.70.12.7511-7519.2004.

    PubMed Central  CAS  PubMed  Google Scholar 

  64. Ruvkun G, Ausubel FM: A general method for site-directed mutagenesis in prokaryotes. Nature. 1981, 289: 85-88. 10.1038/289085a0.

    CAS  PubMed  Google Scholar 

  65. LoVullo ED, Sherrill LA, Perez LL, Pavelka MS: Genetic tools for highly pathogenic Francisella tularensis subsp. tularensis. Microbiology. 2006, 152 (Pt 11): 3425-3435. 10.1099/mic.0.29121-0.

    CAS  PubMed  Google Scholar 

  66. Tempel R, Lai XH, Crosa L, Kozlowicz B, Heffron F: Attenuated Francisella novicida transposon mutants protect mice against wild-type challenge. Infect Immun. 2006, 74 (9): 5095-5105. 10.1128/IAI.00598-06.

    PubMed Central  CAS  PubMed  Google Scholar 

  67. Gray CG, Cowley SC, Cheung KK, Nano FE: The identification of five genetic loci of Francisella novicida associated with intracellular growth. FEMS Microbiol Lett. 2002, 215 (1): 53-56. 10.1111/j.1574-6968.2002.tb11369.x.

    CAS  PubMed  Google Scholar 

  68. Maier TM, Casey MS, Becker RH, Dorsey CW, Glass EM, Maltsev N, Zahrt TC, Frank DW: Identification of Francisella tularensis Himar1-based transposon mutants defective for replication in macrophages. Infect Immun. 2007, 75 (11): 5376-5389. 10.1128/IAI.00238-07.

    PubMed Central  CAS  PubMed  Google Scholar 

  69. Charity JC, Costante-Hamm MM, Balon EL, Boyd DH, Rubin EJ, Dove SL: Twin RNA Polymerase-Associated Proteins Control Virulence Gene Expression in Francisella tularensis. PLoS Pathog. 2007, 3 (6): e84-10.1371/journal.ppat.0030084.

    PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgements

We thank John W. Robison for technical assistance, and James Carroll, and Cory M. Robinson for insightful discussion. The authors thank Deanna Tarwacki for assistance with dendritic cell cultivation and infections. We also thank Francis E. Nano for helpful correspondence regarding the chicken embryo challenge model. We thank Thomas Zahrt and Martin Pavelka for providing us with plasmids and Karen Elkins for providing F. tularensis LVS. This work was funded by the National Institutes of Health grants AI074402 and AI050018. JH is a recipient of T32 AI060525, "Immunology of Infectious Disease."

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Gerard J Nau.

Additional information

Authors' contributions

JH wrote the manuscript, constructed plasmids and mutants, conducted the experiments, envisioned and designed the study. PEC designed the microarrays and contributed toward the execution of microarray experiments. DMO conducted the real time PCR. Both PEC and DMO assisted with the chicken embryo infections. RMQS constructed plasmids and participated in the drafting of the manuscript. GJN oversaw the work, contributed to the conception and design of this research, and participated in the drafting of this manuscript. All authors read and approved the final version of the manuscript.

Electronic supplementary material

12866_2008_605_MOESM1_ESM.doc

Additional file 1: Table S1. This table shows a list of F. tularensis LVS genes significantly induced at 37°C. (DOC 115 KB)

12866_2008_605_MOESM2_ESM.doc

Additional file 2: Table S2. This table displays a list of F. tularensis LVS genes significantly down-regulated at 37°C. (DOC 162 KB)

Authors’ original submitted files for images

Rights and permissions

Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://creativecommons.org/licenses/by/2.0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Horzempa, J., Carlson, P.E., O'Dee, D.M. et al. Global transcriptional response to mammalian temperature provides new insight into Francisella tularensis pathogenesis. BMC Microbiol 8, 172 (2008). https://doi.org/10.1186/1471-2180-8-172

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1471-2180-8-172

Keywords