Free access
Research Article
29 December 2016

pH Alkalinization by Chloroquine Suppresses Pathogenic Burkholderia Type 6 Secretion System 1 and Multinucleated Giant Cells

ABSTRACT

Burkholderia mallei and B. pseudomallei cause glanders and melioidosis, respectively, in humans and animals. A hallmark of pathogenesis is the formation of granulomas containing multinucleated giant cells (MNGCs) and cell death. These processes depend on type 6 secretion system 1 (T6SS-1), which is required for virulence in animals. We examined the cell biology of MNGC formation and cell death. We found that chloroquine diphosphate (CLQ), an antimalarial drug, inhibits Burkholderia growth, phagosomal escape, and subsequent MNGC formation. This depends on CLQ's ability to neutralize the acid pH because other alkalinizing compounds similarly inhibit escape and MNGC formation. CLQ inhibits bacterial virulence protein expression because T6SS-1 and some effectors of type 3 secretion system 3 (T3SS-3), which is also required for virulence, are expressed at acid pH. We show that acid pH upregulates the expression of Hcp1 of T6SS-1 and TssM, a protein coregulated with T6SS-1. Finally, we demonstrate that CLQ treatment of Burkholderia-infected Madagascar hissing cockroaches (HCs) increases their survival. This study highlights the multiple mechanisms by which CLQ inhibits growth and virulence and suggests that CLQ be further tested and considered, in conjunction with antibiotic use, for the treatment of diseases caused by Burkholderia.

INTRODUCTION

Burkholderia mallei, a Gram-negative facultative intracellular bacterium, causes glanders, a disease that is acquired from exposure to infected solipeds. It is a host-adapted clone of B. pseudomallei and an obligate animal pathogen that has lost its capacity to survive in the environment through genomic decay (1). In contrast, B. pseudomallei is a soil saprophyte endemic in Southeast Asia and northern Australia (2). B. pseudomallei infects a broad range of hosts, from plants to humans, a consequence of its 7.2-Mbp genome shaped by horizontal gene acquisition (3). B. pseudomallei causes melioidosis, a disease that is marked by latency, reminiscent of the diseases caused by other granuloma-forming pathogens, such as Mycobacterium tuberculosis (4). Because of a natural resistance to multiple antibiotics, a lack of effective vaccines, a high risk of fatality, and a potential to be weaponized, the two pathogens are listed as tier 1 select agents (www.selectagents.gov/SelectAgentsandToxinsList.html ).
A related species that was formerly classified as B. pseudomallei, B. thailandensis exhibits a high degree of genomic similarity to B. pseudomallei and occupies the same environmental niche (5). Three cases of B. thailandensis infection in humans have been reported (68). Attenuation is partly due to the presence of an arabinose assimilation operon in B. thailandensis because restoration of the ability to metabolize l-arabinose in B. pseudomallei reduces its virulence in Syrian hamsters (9). Attenuation in B. thailandensis is also due to the lack of the 6-deoxyheptan capsular polysaccharides found on B. mallei and B. pseudomallei (10). The importance of the capsule is highlighted by a study that showed that acapsular B. mallei and B. pseudomallei strains have reduced virulence (11). Despite being less virulent, B. thailandensis produces a lethal infection in mice depending on virulence factors similar to those in B. mallei and B. pseudomallei (12, 13) and is virulent in the insect Drosophila melanogaster (14). Because of this, B. thailandensis can serve as a model organism for the study of B. mallei and B. pseudomallei; it can be safely used under biosafety level 2 conditions (1, 13).
B. mallei, B. pseudomallei, and B. thailandensis share similar intracellular lifestyles (15). Following uptake, they proliferate inside the host cell. Although the pathogens have either two or three type 3 secretory apparatuses, type 3 secretion system 3 (T3SS-3) encodes the proteins necessary for proliferation, phagosomal escape, and evasion of autophagy (1618). After escape into the cytoplasm, Burkholderia has the ability to form actin tails and certain strains of B. pseudomallei and B. thailandensis also form lateral flagella; both actin and lateral flagella are important for motility and intercellular spread (18). The pathogens are also endowed with at least four type 6 secretion systems (T6SSs), of which T6SS-1 in B. mallei, B. pseudomallei, and B. thailandensis is important for multinucleated giant cells (MNGCs), leading to the intercellular spread of the pathogens and host cell death (1820). These phenotypes are recapitulated in vivo under disease conditions, such as granulomas, which contain MNGCs that subsequently lyse (2123).
Two models of how Burkholderia spreads from cell to cell and what MNGC formation's role is in its pathogenesis exist. In the first model, intercellular spread is accomplished by propulsion of motile bacilli by actin tails in one cell to form double-membraned vacuoles in the next cell as a result of engulfment by the neighboring cell, reminiscent of the intercellular spread of Listeria and Shigella. Cell fusion and MNGC formation are thought to be unintentional consequences of these events (24). This model is disfavored because few, if any, double-membraned vacuoles containing Burkholderia have been observed and MNGC formation is not part of the pathogenesis of any other actin tail-forming bacteria. In the more favored model, intercellular spread occurs by cell-cell fusion, with a fusogen being inserted in two adjacent and tightly apposed cell membranes (18, 25). Vgr5 of T6SS-5 (also known as T6SS-1) has been identified to be a fusogen (25).
Cell fusion and the formation of MNGCs to promote cell-to-cell spread are integral components of Burkholderia pathogenesis, and we speculate that interference with these processes might be expected to inhibit infection and disease. The use of cell biological approaches to interfere with early events in the intracellular life cycle of Burkholderia may lead to the identification of cellular targets hijacked by the pathogen and potential treatments for the diseases that they cause. Here, we demonstrate that chloroquine diphosphate (CLQ) interferes with growth, phagosomal escape, MNGC formation, and host cell death. The results strongly suggest that CLQ, which is used to treat malaria, and other drugs that have alkalinizing properties should be further evaluated as anti-infective treatments that may be used in conjunction with other therapeutics for glanders and melioidosis.

RESULTS

Chloroquine inhibits MNGC formation in RAW 264.7 cells.

In the absence of CLQ, MNGC formation was induced after infection with B. mallei, B. thailandensis, or B. pseudomallei (Fig. 1A to F; a complete list of strains that were tested and observed to induce MNGCs is shown in Table 1), as shown by either immunofluorescence (Fig. 1A and E) or Wright-Giemsa staining (Fig. 1C). Without CLQ, multiple nuclei were found to be stacked and clustered at cell centers in large Burkholderia-induced MNGCs. These appeared to be morphologically similar to foreign body-type giant cells (FBGCs), whereby nuclei cluster centrally within the giant cells stacked on top of one another, but distinct from Langhans giant cells, in which nuclei are arranged in a horseshoe pattern (26). The rate at which MNGCs form depended on the multiplicity of infection (MOI) and cell confluence. Larger MNGCs were formed from the fusion of smaller MNGCs (see Movie S1 in the supplemental material). Given optimal conditions, we have observed that some MNGCs contain as many as 1,000 nuclei, likely as a result of fusion between several large MNGCs (data not shown).
FIG 1
FIG 1 Chloroquine inhibits Burkholderia-induced MNGC formation in RAW 264.7 cells. (A, C, E) Representative confocal micrographs of macrophages infected with the indicated Burkholderia spp. for 1 h at an MOI of 5 (B. mallei SR1 [A] or B. pseudomallei 1026b [E]) or an MOI of 1 (B. thailandensis [C]) and either left untreated (control [Ctrl]) or treated with 75 μM CLQ or 10 μg/ml nocodazole (NOC) (E). At the indicated times, cells were fixed, subjected to immunofluorescence staining, and imaged with a confocal microscope. Nuclei were stained with DAPI (red), actin was stained with phalloidin (white), and bacteria were stained with anti-Burkholderia Ab (green). Bars = 50 μm (A and E). Alternatively, fixed cells were stained with Wright-Giemsa and imaged with a light microscope. Bar = 30 μm (C). (B, D, F, G) From the microscopy images, MNGC formation was quantified by manually counting all nuclei and cells in a given field, whether they were mononucleated cells or MNGCs. The number of nuclei was divided by the number of cells from an average of 3 images per coverslip per experiment to calculate the ratio of the number of nuclei/number of cells (Ratio [N/C]). Conditions that resulted mostly in mononucleated cells displayed N/C ratios of 1.0, whereas those that resulted in MNGCs displayed N/C ratios of >1. See cell fusion and the formation of MNGCs in Movie S1 in the supplemental material. The total numbers of nuclei counted for the graphs in panels B, D, F, and G were 12,425, 11,818, 18,465, and 4,720, respectively. The P values in panels B, D, and F, representing the differences between the control and experimental conditions at each time point, were calculated using an asymptotic normal test. The P values in panel G represent the differences between CLQ-treated cells and control or NOC-treated cells (P = 0.0002 between control and NOC-treated cells). Error bars represent standard errors of the means (SEMs). Data are from 2 independent experiments for B. mallei (B), 3 independent experiments for B. thailandensis (D), 3 independent experiments for B. pseudomallei (F), and 3 independent experiments for B. pseudomallei with CLQ and NOC (G). Bm, B. mallei; Bp, B. pseudomallei; Bt, B. thailandensis.
TABLE 1
TABLE 1 Bacterial strains used in this studya
Strain testedPropertiesReference
B. mallei strains
    ATCC 23344Type strain, originally isolated in 1944 from a human case of glanders50
    SR1ATCC 23344, sucrose-resistant derivative20, 50
B. pseudomallei strains
    AIAminoglycoside-sensitive derivative of K96243, a clinical isolate from northeastern Thailand3, 19
    MSHR305Clinical isolate from a fatal encephalomyelitis case; the strain is also capable of flagellar motility27
    1026bClinical isolate from a case of septicemic melioidosis with skin, soft tissue, and spleen involvement51
B. thailandensis strain DW503E264 derivative, aminoglycoside sensitive; the strain is also capable of flagellar motility5, 52
B. mallei strains used in the assay whose results are shown in Fig. 8
    SR1(pBHR2-virAG)Broad-host-range plasmid that constitutively expresses virAG20
    DDA0746 ΔvirGSR1 derivative20
    DDA0742 Δhcp1SR1 derivative20
    DDA0729-1 ΔtssM::GFP(pBHR2)SR1 derivative with a broad-host-range plasmid containing no insert53
    DDA0729-1 ΔtssM::GFP(pBHR2-virAG)SR1 derivative with a broad-host-range plasmid that constitutively expresses virAG53
a
All strains are from the USAMRIID collection.
CLQ, a lysosomotropic drug used to block phagosome maturation, was found to inhibit the MNGC formation induced by the three species (Fig. 1A to F). B. mallei-, B. pseudomallei-, and B. thailandensis-infected cells remain mostly mononucleated in the presence of CLQ (Fig. 1B, D, and F). CLQ inhibited MNGC formation by strains capable of actin tail and lateral flagellum formation, e.g., B. pseudomallei MSHR305 (27), as well as those strains that form actin tails but lack lateral flagella, e.g., B. pseudomallei AI, derived from B. pseudomallei K96423 (NCBI accession number NC_006350.1 ). All MNGCs that were induced by the strains that we tested (Table 1), including B. pseudomallei MSHR305 and B. pseudomallei AI, were inhibited by CLQ. Furthermore, CLQ blocked the MNGC formation induced by B. thailandensis in other cell types, such as baby hamster kidney cells (data not shown). We also tested the microtubule-depolymerizing agent nocodazole (NOC), a drug that inhibits the formation of FBGCs (28), which are morphologically similar to Burkholderia-induced MNGCs. NOC did not inhibit the MNGC formation induced by B. pseudomallei (Fig. 1E and G) or B. thailandensis (data not shown).
MNGC formation was quantified by counting the number of nuclei and cells of both mononucleated and multinucleated cells in a given field of view and was expressed as the ratio of the number of nuclei/number of cells (the N/C ratio). Figures 1B, D, and F show the results of quantification in B. mallei-, B. thailandensis-, and B. pseudomallei-infected cells. The N/C ratio was >1 for untreated infected control cells at later time points typically starting at 12 h postinfection (p.i.), whereas the N/C ratio of CLQ-treated infected cells was ∼1. Hence, CLQ interfered with the formation of MNGCs.

Chloroquine inhibits the cell death that invariably accompanies MNGC formation.

Burkholderia-induced MNGC formation is a terminal process that is invariably followed by host cell death (18, 2931). To investigate if CLQ also inhibits the death of infected cells, we used calcein AM to label live cells and ethidium homodimer 1 (EthD-1) to label dead cells. As CLQ had similar effects on all three Burkholderia spp., as shown in Fig. 1, B. thailandensis was used as a model pathogen for B. mallei and B. pseudomallei. Figure 2A shows that CLQ-treated B. thailandensis-infected cells were mostly labeled with calcein AM, which indicates that they were alive, whereas a majority of untreated infected (control) cells were labeled with EthD-1, which indicates that they were dead. Dead cells also retained calcein AM, but labeling was mostly perinuclear and morphologically distinct from the homogeneous staining seen in live cells. In addition, the release of lactate dehydrogenase (LDH) from cells, indicative of cell membrane permeability and death, was measured in B. thailandensis-infected cells left untreated or treated with CLQ. The release of LDH from infected cells (expressed as percent cytotoxicity) was lower with CLQ treatment than no treatment (Fig. 2B) (P = 0.0179 at 18 h p.i.). Of note, CLQ treatment of uninfected cells did cause low levels of cell lysis, although the difference was not statistically significant (Fig. 2C) (P = 0.35 at 18 h p.i.). Lastly, a terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay based on fluorescence imaging was used to look for fragmented DNA, found in either pyroptotic or apoptotic cell death. Figure 2D shows pyknotic nuclei labeled with dUTP in infected cells that received no treatment, in contrast to the lack of labeled cells after CLQ treatment. Thus, cell death induced by Burkholderia can be prevented by CLQ.
FIG 2
FIG 2 Chloroquine inhibits Burkholderia-induced cell death in RAW 264.7 cells. (A) Representative confocal microscopy images of macrophages infected with B. thailandensis at an MOI of 1 and either left untreated (control [Ctrl]) or treated with 75 μM CLQ. At the indicated times, cells were incubated with calcein AM (green; live cells) and EthD-1 (red; dead cells) for 5 min and immediately imaged. Bar = 50 μm. Similar results were observed in 3 independent experiments. (B) Macrophages were infected with B. thailandensis at an MOI of 5 and either left untreated (control) or treated with 75 μM CLQ. At the indicated times, medium from at least 3 wells per time point and treatment was collected. Medium from each well was assayed in duplicate for LDH release. LDH release is expressed as percent cytotoxicity. The P values were calculated using an unpaired t test. Error bars represent SEMs (n = 3 independent experiments). (C) Uninfected macrophages were left untreated (control) or treated with 75 μM CLQ, and at the indicated times, medium from at least 3 wells per condition was collected. For LDH release, medium from each well was assayed in duplicate. The LDH release was not significantly different between control and CLQ-treated cells, although the values for CLQ-treated cells tended to be higher than those for the control cells. Error bars represent SEMs (n = 3 independent experiments). (D) Representative confocal microscopy images of macrophages infected with B. thailandensis at an MOI of 1 and either left untreated (control) or treated with 75 μM CLQ. At the indicated times, cells were fixed and subjected to the TUNEL assay to label pyroptotic or apoptotic cells. Bar = 50 μm. Similar results were observed in 3 independent experiments.

Chloroquine inhibits intracellular replication and prevents phagosomal escape.

On the basis of confocal imaging, fewer bacilli were seen inside CLQ-treated cells, in contrast to untreated infected cells. To quantify intracellular growth, B. thailandensis-infected cells were lysed and plated for determination of the number of CFU. Bacterial counts confirmed that fewer bacilli were present with CLQ treatment than no treatment, and the difference became larger over time (Fig. 3A). The disparity in the numbers of CFU was not a consequence of differences in adherence or uptake; no difference in the number of CFU was seen when cells were treated with CLQ prior to or during infection and when cells were not treated (Fig. 3B). Moreover, no growth defect was observed when B. thailandensis was grown in vitro in Luria-Bertani (LB) broth (Lennox) in the presence of increasing concentrations of CLQ (Fig. 3C). Therefore, CLQ specifically inhibits the growth of Burkholderia inside cells.
FIG 3
FIG 3 Chloroquine inhibits intracellular growth and escape from the phagosome. (A) Macrophages were infected with B. thailandensis at an MOI of 1 and left untreated (control [Ctrl]) or treated with 75 μM CLQ. Coverslips from 2 wells per treatment and time were washed in PBS to remove extracellular bacteria and then lysed in fresh plates at the indicated times. Bacterial lysate was serially diluted and plated for determination of the number of CFU in duplicate. P values were calculated by an asymptotic normal test. Error bars represent SEMs (n = 3 independent experiments). (B) Macrophages were infected with B. thailandensis at an MOI of 20 for 1 h and lysed at 2 h p.i. to determine bacterial loads. Lysed cells from 3 coverslips per condition were plated in duplicate for determination of the number of CFU. CLQ (75 μM) was added 1 h prior to infection and was present at the time of B. thailandensis addition and throughout the infection. Error bars represent SEMs (n = 3 independent experiments). Differences between control and CLQ-treated cells were not significant. (C) Growth curve obtained by plotting the optical density at 600 nm (OD600) versus time for B. thailandensis grown in LB broth (Lennox) and left untreated (control) or treated with 7.5, 75, or 750 μM CLQ. Error bars represent SEMs (n = 2 independent experiments). (D and E) B. thailandensis-infected cells (MOI = 20) with or without 75 μM CLQ treatment were fixed at the indicated times and subjected to immunostaining against Burkholderia and LAMP1. Cells were also stained for nuclei and actin and were stained with the latter to delineate the limits of the cell. The number of bacilli in LAMP1+ phagosomes (ϕ) was scored. The total number of bacilli counted was 766 for control cells and 618 for CLQ-treated cells. P values were calculated using an unpaired t test. Error bars represent SEMs (n = 3 independent experiments). (E) Representative confocal micrographs of cells infected with Burkholderia left untreated (control) or treated with CLQ. Cells were fixed at 6 h p.i. and stained against Burkholderia (green), LAMP1 (red), and nuclei (blue). Arrows, B. thailandensis in LAMP1+ phagosomes (ϕ). Bar = 10 µm. (F and G) B. thailandensis-infected cells (MOI = 20) with or without 75 μM CLQ treatment were fixed and stained for Burkholderia, actin, and nuclei as described in the legend to panels D and E. (F) The number of bacilli with actin tails was scored. The experiments used to count LAMP1+ phagosomes (D and E) were independent of the experiments used to count actin tails. The total number of bacilli counted was 1,036 for control cells and 850 for CLQ-treated cells. The P values were calculated using an unpaired t test. Error bars represent SEMs (n = 3 independent experiments). (G) Representative confocal micrographs of cells infected with Burkholderia left untreated (control) or treated with CLQ. Cells were fixed at 6 h p.i. and stained against Burkholderia (green) and actin (red). Bar = 10 µm.
A hallmark of pathogenesis is escape from the phagosome into the cytoplasm, a prerequisite to the actin tail polymerization and MNGC formation that follow (15). To quantify phagosomal escape, the bacilli inside phagosomes delineated by LAMP1 were scored. A progressive reduction in B. thailandensis and LAMP1 colocalization was seen in untreated cells, whereas with CLQ treatment, B. thailandensis was retained in the phagosomes at later time points (Fig. 3D and E). Scoring for actin tails or nucleation at the bacillus poles confirmed this result. Figures 3F and G show that the number of bacilli associated with actin increased dramatically at 6 h p.i. in control cells, which coincides with the time when 90% of bacilli no longer colocalized with LAMP1. Of note is the 2-h lag in the formation of the actin tail after escape from a LAMP1-positive (LAMP1+) compartment. In contrast, no increase in the number of bacilli with actin tails was seen in CLQ-treated cells. Therefore, one way that the ensuing MNGC formation is prevented by CLQ is by delaying or preventing the pathogen from escaping the phagosome. Taken together, the growth inhibition that was observed with CLQ at 9 to 18 h (Fig. 3A) in the absence of MNGCs (Fig. 1B, D, and F) suggests that CLQ inhibits growth within single cells likely due in part to the inhibition of escape to the cytosol, in addition to reducing growth by inhibiting the subsequent formation of MNGCs, which provides new cells and nutrients for infection. Given the inhibitory effect of CLQ on growth and MNGC formation in macrophages, we next examined the effectiveness of CLQ in the treatment of infected animals.

Chloroquine increases the survival of Burkholderia-infected HCs.

The innate immune systems of insects and mammals have a high degree of similarity (32). Insect hemocytes and mammalian neutrophils phagocytose and kill microorganisms (33). Antimicrobial peptides are also produced in recognition of pathogen-associated molecular patterns by Toll receptors in insects and Toll-like receptors in mammals (33). Insects are a tractable surrogate host that may be used to reduce the number of mammals used (33, 34), and in particular, Madagascar hissing cockroaches (HCs) have been used in B. mallei, B. pseudomallei, and B. thailandensis virulence studies (35). MNGCs are formed in the hemolymph of HCs upon Burkholderia infection, and the virulence in HCs correlates with that in rodent models of infection (35). Here, in 4 separate experiments, groups of 10 or 12 HCs were infected with B. thailandensis, left untreated or treated with CLQ, and followed for 7 days. Treatment with CLQ increased the percentage of B. thailandensis-infected HCs that survived compared to the number of infected HCs that received no treatment that survived (Fig. 4) (P = 0.0270). Ninety-seven percent of HCs survived when they were inoculated with phosphate-buffered saline (PBS) or CLQ alone. Thus, CLQ treatment partially protects HCs infected with Burkholderia.
FIG 4
FIG 4 Chloroquine increases the survival of B. thailandensis-infected HCs. HCs were challenged at day 0 with B. thailandensis at doses ranging from 10 to 20 LD50s and treated with CLQ or PBS on the same day, as described in Materials and Methods. Additional uninfected control groups received CLQ or PBS alone. HC survival (in percent) was monitored for 7 day. The P value represents the significance of the difference at 7 days between the B. thailandensis-infected group and the B. thailandensis-infected and CLQ-treated group and was calculated using an asymptotic normal test. Four independent experiments were performed, each with 10 or 12 HCs per group.

Autophagy is not involved in how chloroquine functions to prevent MNGCs.

We next began studies to address the possible mechanisms by which CLQ prevents MNGC formation. A previous study showed that CLQ may block autophagy by interfering with autophagosome clearance, as evidenced by an increase in the observable number of autophagosomes (36). Similarly, we observed an increase in the number of autophagosomes, stained by monodansylcadaverine (MDC), in uninfected cells treated with CLQ (Fig. 5A), consistent with the blockage of autophagy. Burkholderia infection without CLQ treatment did not result in an increase in the number of autophagosomes; no increase in MDC staining was observed (Fig. 5B). Therefore, we wanted to examine whether CLQ's action involved autophagy by utilizing other inhibitors of autophagy to determine whether they also inhibit MNGC formation. B. thailandensis-infected cells that were treated with the autophagy inhibitors (36) 3-methyladenine (3MA) and leupeptin (LEU) did not inhibit MNGC formation (Fig. 5C and D). Furthermore, rapamycin, an inducer of autophagy, also did not inhibit MNGC formation (Fig. 5C and E). Therefore, autophagy is likely not involved in CLQ's ability to inhibit the replication and MNGC formation of Burkholderia-infected cells.
FIG 5
FIG 5 Chloroquine does not function through autophagy to inhibit MNGC formation. (A) Representative confocal images of uninfected cells left untreated (control) or incubated with 75 μM CLQ. At 9 h posttreatment, cells were stained with monodansylcadaverine (MDC; green, autophagosomes) and propidium iodide (PI; red, dead cells) for 5 min prior to imaging. Bar = 20 μm. (B) Representative confocal image of untreated cells infected with B. thailandensis (MOI = 10) at 9 h p.i. Cells were stained with MDC (green) and PI (red). The images in panels A and B were taken under the same laser power and gain. Data are representative of those from 3 independent experiments. (C) Representative confocal images of cells infected with B. thailandensis at an MOI of 1 and treated with 10 mM 3-methyladenine (3MA), 0.3 mM leupeptin (LEU), or 4 μM rapamycin (RAP). The cells were fixed at 12 h p.i. and stained with DAPI (nuclei; red), phalloidin (actin; white), and an anti-Burkholderia Ab (green). Data are representative of those from 2 independent experiments. Bar = 50 μm. (D) Quantification of MNGC formation as the ratio of the number of nuclei to the number of cells (R [N/C]) in B. thailandensis-infected cells (MOI = 1) left untreated (control [Ctrl]) or treated with 10 mM 3MA or 0.3 mM LEU at 18 h p.i. The total number of nuclei counted was 3,865. Error bars represent SEMs (n = 2 independent experiments). The differences between the control and the experimental treatments, based on an asymptotic normal test, were not significant. (E) Quantification of MNGC formation as the R [N/C] in B. thailandensis-infected cells (MOI = 1) left untreated (control) or treated with 4 μM RAP at 18 h p.i. The total number of nuclei counted was 2,110. Error bars represent SEMs (n = 2 independent experiments). The difference between the controls and the experimental treatments, based on an asymptotic normal test, was not significant.

By shifting the pH from acid to neutral, chloroquine inhibits replication, phagosomal escape, and MNGC formation.

CLQ raises the pH of acidic compartments, accumulates in endolysosomes, and suppresses the activation of lysosomal proteases (37). Using LysoTracker red to label acidic organelles and DQRed bovine serum albumin (BSA), a substrate that fluoresces upon cleavage by acid-activated lysosomal proteases, we confirmed that acidic organelles were neutralized with CLQ (Fig. 6A and B). Therefore, we asked if alkalinization of acidic compartments creates a suboptimal growing environment for Burkholderia resulting in less intracellular growth and replication. To address this, B. thailandensis was grown in LB broth (Lennox) adjusted to pH 6.8 (neutral pH), pH 5.5, or pH 5.0 (acid pH). There was significantly more growth at pH 5.0 or at pH 5.5 than at pH 6.8 at 6 and 8 h (Fig. 6C). Thus, Burkholderia prefers acidic conditions for optimal growth, which is consistent with the lower intracellular growth of B. thailandensis seen in CLQ-treated cells in Fig. 3A.
FIG 6
FIG 6 Chloroquine alkalinizes endocytic compartments, and alkalinization inhibits replication, phagosomal escape, and MNGC formation. (A) Representative confocal images of LysoTracker red staining of uninfected cells untreated (control [Ctrl]) or treated with 75 μM CLQ. Data are representative of those from 3 independent experiments. Bar = 10 μm. (B) Representative confocal images of DQRed BSA fluorescence in uninfected cells untreated (control) or treated with 75 μM CLQ. Data are representative of those from 3 independent experiments. Bar = 10 μm. (C) B. thailandensis growth in LB broth (Lennox) adjusted to pH 5.0, 5.5, or 6.8 for 8 h. The statistical significance of differences between growth at pH 6.8 and that at pH 5.0 or 5.5 was calculated using an unpaired t test. Error bars represent SEMs (n = 3 independent experiments). (D and E) Macrophages were infected with B. thailandensis at an MOI of 20 and left untreated (control) or treated with 250 nM bafilomycin A (BAF) (D) or 30 mM ammonium chloride (NH4Cl) (E). At the indicated times, cells were fixed and stained for LAMP1, Burkholderia, and actin. The number of bacilli in LAMP1+ phagosomes (ϕ) was scored. The number of bacilli counted was 609 for control cells (D and E), 438 for BAF-treated cells (D), and 409 for NH4Cl-treated cells (E). The P values were calculated using an unpaired t test. Error bars represent SEMs (n = 3 independent experiments). (F) Representative confocal images of B. thailandensis-infected cells (MOI = 1) (green) untreated (control) or treated with 250 nm BAF or 30 mM NH4Cl at 15 h p.i. Actin was stained with phalloidin (white), and the nucleus was stained with DAPI (red). Data are representative of those from 3 independent experiments. Bar = 50 μm.
To determine if alkalinization also leads to retention in phagosomes, B. thailandensis-infected cells were treated with bafilomycin A (BAF) or ammonium chloride (NH4Cl), each of which also alkalinizes phagosomes, and scored for LAMP1 colocalization. B. thailandensis was retained in phagosomes in cells treated with BAF (Fig. 6D) or NH4Cl (Fig. 6E), whereas it was not in untreated infected (control) cells. MNGC formation was also inhibited with BAF or NH4Cl (Fig. 6F). Notably, the retention of B. thailandensis in phagosomes by BAF directly points to endocytic organelles as the organelles targeted by CLQ because BAF inhibits vacuolar H+-ATPase-dependent acidification.
CLQ belongs to a class of antimalarial drugs which are purported to have weak base properties (37). Do these CLQ analogs also inhibit MNGC formation? To address this, B. thailandensis-infected cells were treated with 75 μM amodiaquine dihydrochloride dihydrate (ADQ) or 150 μM primaquine bisphosphate (PMQ) and examined for MNGC formation. Figure 7A shows that both drugs inhibited MNGC formation in infected cells. ADQ also inhibited MNGC formation in B. mallei-infected cells (Fig. 7B). Thus, by creating an alkaline environment inside cells, CLQ prevents optimal growth, phagosomal escape, and MNGC formation.
FIG 7
FIG 7 Antimalarial drugs, which have weak base properties, also inhibit MNGC formation. (A) Representative confocal images of B. thailandensis-infected cells (MOI = 1) that were left untreated (control [Ctrl]) or treated with 75 μM amodiaquine dihydrochloride dihydrate (ADQ) or 150 μM primaquine bisphosphate (PMQ) for the indicated times. Cells were labeled with phalloidin (actin; white), DAPI (nuclei; red), and anti-Burkholderia Ab (green). Data are representative of those from 2 independent experiments. Bar = 50 μm. (B) Quantification of MNGC formation as the ratio of the number of nuclei/number of cells (Ratio [N/C]) in B. mallei-infected cells (MOI = 5) left untreated (control) or treated with 75 μM ADQ at the indicated times. The total number of nuclei counted was 10,449. The P values were calculated using an asymptotic normal test. Error bars indicate SEMs (n = 2 independent experiments).

Expression of BopC and BopE under acid pH.

T3SS-3 and its effectors are important for Burkholderia replication and phagosomal escape (17, 18). Therefore, the expression of T3SS-3 effectors was examined after growth at pH 5 and 7. Western blot analyses showed similar BopC protein levels at both pHs, whereas higher levels of BopE were seen at pH 5 than pH 7 (Fig. 8A), likely because these proteins are in different operons. We conclude that the expression of some T3SS-3 effectors is regulated in response to pH and the acid-induced effectors are likely required for replication and phagosomal escape.
FIG 8
FIG 8 T3SS and T6SS proteins are regulated by acid pH. (A) Immunoblot analyses of BopC, BopE, and GroEL in the pellet fractions of B. mallei SR1 grown in M9 broth with 4% glycerol adjusted to pH 5 or pH 7. GroEL served as a loading control. Data are representative of those from 2 independent experiments. (B) Representative confocal images of B. mallei ΔtssM::GFP(pBHR2) grown for 3 h in LB broth (Lennox) adjusted to pH 5 or pH 7. B. mallei ΔtssM::GFP(pBHR2-VirAG) grown in broth at pH 7 served as a positive control. All images were taken under the same laser power and gain. Data are representative of those from 2 independent experiments. Bar = 10 μm. (C) Immunoblot analysis of TssM and DnaK expression when B. mallei SR1 was grown in broth adjusted to pH 5 or pH 7 for 24 h. DnaK served as a loading control. Data are representative of those from 2 independent experiments. (D) Immunoblot analysis of Hcp1 and GroEL expression when B. mallei SR1 was grown at pH 5 or pH 7 for 24 h. Hcp1 was not expressed when B. mallei DDA0746 (ΔvirG) or B. mallei DDA0742 (Δhcp1) was grown at pH 5, whereas B. mallei SR1(pBHR2-virAG) expressed Hcp1 when it was grown at pH 7. GroEL served as a loading control. Data are representative of those from 2 independent experiments. (E) Immunoblot analysis of Hcp1 and GroEL expression when B. pseudomallei AI was grown at pH 5 or pH 7 for 7 h. GroEL served as a loading control. Data are representative of those from 2 independent experiments.

Acid pH upregulates TssM and Hcp1 expression through VirAG.

Bacterial genes transcriptionally regulated by the two-component regulatory system VirAG, including the T6SS-1 gene cluster, are important for MNGCs and virulence (19, 20). Therefore, we examined the expression of TssM, a protein regulated by VirAG, at pH 5 and 7. Figure 8B shows that B. mallei ΔtssM::GFP(pBHR2), a mutant in which the tssM gene has been replaced with a promoterless green fluorescent protein (GFP) gene, fluoresced when it was grown at pH 5 but not when it was grown at pH 7. In contrast, B. mallei ΔtssM::GFP(pBHR2-virAG), a mutant carrying a plasmid that expresses VirAG under the control of a constitutive promoter, fluoresced at pH 7 (Fig. 8B), demonstrating that TssM can be expressed at pH 7 when VirAG is upregulated. Further, the increased expression of TssM in B. mallei at pH 5 compared with that at pH 7 was also confirmed by Western blotting (Fig. 8C). Thus, TssM is produced in response to acid pH.
Hcp1 is an essential T6SS-1 protein that forms the channel of the secretion apparatus (19). Western blot analysis showed that Hcp1 is expressed by B. mallei (Fig. 8D) and B. pseudomallei (Fig. 8E) at pH 5 but is absent or is present in reduced amounts at pH 7. As expected, the B. mallei Δhcp1 mutant did not express Hcp1 at pH 5 (Fig. 8D). Interestingly, no Hcp1 expression was seen in the B. mallei ΔvirG mutant at pH 5, demonstrating that the pH-dependent expression of Hcp1 requires a functional VirAG (Fig. 8D). Hcp1 was expressed at pH 7 in B. mallei SR1(pBHR2-VirAG), in which VirAG is constitutively expressed, as anticipated (Fig. 8D). Thus, acidification drives Hcp1 and TssM upregulation, and by preventing acidification, CLQ inhibits this process.

DISCUSSION

In this study, we illustrate how Burkholderia takes advantage of the host to promote its intracellular survival and how CLQ counteracts this process. On a cellular level, by raising the pH of acidic compartments, CLQ restricts replication, prevents/delays phagosomal escape, inhibits MNGC formation, and prevents host cell death, which releases the pathogen into the extracellular milieu. The consequence is a decrease in intracellular replication and an increase in the rate of survival of infected animals treated with CLQ, illustrated here in the cockroach model of infection. CLQ functions by alkalinizing acidic conditions inside host cells, as other alkalinizing agents also prevent phagosomal escape and inhibit MNGC formation. On a molecular level, alkalinization appears to inhibit the expression of the bacterial genes for T6SS-1 as well as T3SS-3. Of importance is the inhibition of Hcp1 expression, an essential component of the T6SS-1 secretory apparatus, in both pathogens, B. mallei and B. pseudomallei. We show that the levels of expression of Hcp1, the T3SS-3 effector BopE, and TssM (a protein coregulated with T6SS-1) are greater at acid pH than neutral pH. Thus, CLQ functions in multiple ways to affect and limit Burkholderia survival inside hosts.
The higher level of expression of BopE at pH 5 than pH 7 that we first show in this study supports the finding that pH regulates the secretion of T3SS-3 proteins (13, 38), necessary for phagosome escape. This is consistent with other reports (38) and our finding that Burkholderia is retained in the phagosome with CLQ treatment. However, not all T3SS-3 gene expression is regulated by pH because the level of expression of the gene for BopC, another T3SS-3 effector, is similar at pH 5 and pH 7. Expression of T6SS-1 effectors is likely to be differentially regulated as well. In a recent report by Wong et al., VirA and, by extension, T6SS-1 are found to be regulated by the level of glutathione in the cytoplasm (39), yet we also found that the expression of the hcp1 gene, which is also regulated by VirAG, is upregulated at pH 5. Although the acid pH signal in the phagosome, which can reach pH 5, precedes glutathione signaling in the cytosol as Burkholderia transits from the phagosome to the cytoplasm, the latter is likely important for extending and amplifying signals to create robust downstream events. Similar to the findings for T3SS-3 with BopC and BopE, the different T6SS-1 effectors likely receive different and multiple signals to fine-tune the regulation of the secretory system in a temporal and local manner, allowing Burkholderia to take full advantage of its intracellular niche.
Our finding that expression of Hcp1, an essential component of the T6SS apparatus, is upregulated at acid pH likely affects the secretion of many T6SS-1-dependent effectors, including the bacterial fusogen VgrG5 (also known as VgrG1) (25). Because Hcp1 expression is also dependent on VirAG, it is also possible that pH might regulate VirAG, thus influencing the expression of the 60-plus VirAG-regulated genes (20), many of which are also T6SS-1 effectors. Even if VirAG is not regulated by pH, it is more than likely that alkalinization by CLQ may change the expression levels of other T6SS-1 proteins individually, similar to the regulation of TssM, a protein coregulated with T6SS-1. Moreover, the fact that acid pH regulates tssM expression in the phagosome is consistent with its upcoming and immediate role in the cytoplasm; TssM is important in dampening down the immune response by interfering with the ubiquitination of the NF-κB and interferon-sensitive response element pathways (40).
Because T3SS-3 and T6SS-1 function sequentially as Burkholderia traffics from the phagosome to the cytoplasm, the inhibition of MNGC formation by CLQ in our study might be construed as secondary to the inhibition of T3SS-3 function and phagosomal escape that we also observed. However, we show that acid pH (pH 5) upregulates the expression of both secretory systems simultaneously and that this likely occurs in the acidifying phagosome because this compartment and the lysosomes are the only compartments in the cell that have the ability to reach pH 5. This does not preclude CLQ possibly functioning in the cytoplasm, where the pH is ∼7.3. With two pKas of 10.3 and 8.4, CLQ is likely to be diprotonated and retained in both the acidifying phagosome and cytoplasm, even though 100 times more protons are available at pH 5 than at pH 7, likely leading to a much higher concentration of CLQ in acidifying phagosomes. In light of VirA being controlled by glutathione levels (39), it is possible that CLQ also functions to oxidize glutathione, which has a pKa of 8.8, leading to an inhibition of MNGC formation. Precisely how CLQ functions in the cytoplasm will be explored in future studies. Nevertheless, acidification is likely to be one of the earliest signals encountered by Burkholderia as it begins its intracellular life cycle. Evolutionarily, other pathogens, such as Mycobacterium, have utilized and subverted acidification to carve their own intracellular niche.
CLQ is a licensed drug used to prevent and treat malarial diseases (37). Plasmodium falciparum, the etiological agent of malaria, is killed by CLQ because alkalinization of the pH prevents enzymes in the red blood cell from degrading hemoglobin, consequently starving the parasite (37). Similar to the CLQ function in preventing malaria, CLQ might also function to inactivate acid-dependent lysosomal hydrolases to limit Burkholderia intracellular growth through a paucity of nutrients. It is likely that the multifactorial effects of CLQ on inhibiting the cellular pathogenesis of Burkholderia and Plasmodium may also occur with other pathogens that traffic through acidic compartments.
A previous report showed that the formation of MNGCs was enhanced when B. pseudomallei-infected macrophages were activated by CpG deoxyoligonucleotide, and CLQ treatment of these cells reduced but did not completely abolish MNGC formation (41). Further, but in conflict with our findings, CLQ treatment of naive (not CpG-activated) macrophages did not reduce MNGC formation (41). Differences in the CLQ concentration and, possibly, the use of different CLQ derivatives may have resulted in a disparity between the two findings; we utilized a higher concentration of chloroquine diphosphate (75 μM) in our study.
Adler and colleagues showed that induction of autophagy through rapamycin and gamma interferon decreases B. pseudomallei survival (16). From their work, a prediction might be made that CLQ, which is an inhibitor that is sometimes used to block autophagy, would promote Burkholderia survival. However, findings from both studies can be reconciled because the authors showed that survival is reduced only when autophagy is induced above the basal level, and they saw no increase in survival with inhibitors of autophagy (16). Moreover, the two studies employed different drugs with distinct modes of action. Whereas wortmannin specifically targets the type III phosphatidylinositol 3-kinase, CLQ's function is more comprehensive because it has also been reported to inhibit DNA repair, regulate cell-cell adhesion, etc., in addition to regulating other pH-dependent processes (42, 43).
The Burkholderia fusogen Vgr5 has already been identified (25). In addition to this bacterial factor, host cellular proteins may also regulate giant cell formation. Burkholderia-induced cell fusion has been shown to depend in part on host cell surface molecules (44). The chemokine profile and the osteoclast marker genes expressed during B. pseudomallei infection are similar to those of cells undergoing osteoclastogenesis, a natural cellular process that creates giant cells to resorb bone (45). Furthermore, fusion to form osteoclasts is impaired in V-ATPase VO subunit d2-deficient mice (46), suggesting that the proton pump and luminal acidification play key roles in cell fusion and the formation of giant cells. Thus, CLQ may also impair host cell functions, in particular, those that affect acidification, to limit MNGC generation during Burkholderia infection. CLQ also inhibits the formation of FBGCs in vitro (28), further suggesting that it may prevent MNGC formation by affecting host as well as bacterial cell functions.
We have shown that CLQ increases the survival of Burkholderia-infected cockroaches, and future directions of research will include testing of CLQ in murine and higher-order animal models. Moreover, other CLQ analogs and antimalarial drugs are also efficacious in preventing MNGC formation and therefore should be investigated. CLQ may also increase the antimicrobial activity of antibiotics that are less active at the acid pH in the phagosome (47), and we are in the process of following this line of research. Nevertheless, this report underscores the need for CLQ to be further examined as a potential treatment for glanders and melioidosis.

MATERIALS AND METHODS

Bacterial strains and culture.

The bacterial strains and plasmids used in this study are described in Table 1. B. pseudomallei and B. thailandensis strains were grown in LB broth (Lennox) (Sigma-Aldrich) for 14 to 16 h at 37°C. For the assays whose results are shown in Fig. 8A, C, and D, M9 minimal broth was used. All media used to grow B. mallei, LB and M9 medium, were supplemented with 4% glycerol. Kanamycin (10 μg/ml) was added to the cultures of strains carrying plasmids. All work with B. pseudomallei and B. mallei was performed in a biosafety level 3 facility.

Cell culture.

RAW 264.7 cells were maintained in Dulbecco's modified Eagle medium with 6 mM l-glutamine and 10% fetal bovine serum at 37°C with 5% CO2. RAW 264.7 cells (4.5 × 105) were seeded on a 12-mm number 1-1/2 coverslip (Electron Microscopy Sciences) for MNGC formation, whereas 2 × 105 cells were seeded for phagosomal escape studies. For Movie S1 in the supplemental material, 1.6 × 106 macrophages were seeded on a 35-mm fluorodish with a 0.17-mm-thick glass bottom.

Bacterial infection.

For MNGC formation, RAW 264.7 cells were infected the day after plating with B. thailandensis at an MOI of 1 or with B. pseudomallei or B. mallei at an MOI of 5 and incubated at 37°C for 1 h. For Movie S1, cells were infected with B. thailandensis at an MOI of 10. The MOIs used were chosen to optimize the rate of MNGC formation within 18 h. For LAMP1 and actin studies, cells were infected with B. thailandensis at an MOI of 20 and incubated at 37°C for 1 h. After uptake, the coverslips were washed vigorously and successively in five 100-ml volumes of PBS to remove extracellular bacilli. The coverslips were then placed in antibiotic-free medium with or without drugs. For the assay whose results are shown in Fig. 2B, cells were infected with B. thailandensis at an MOI of 5 and incubated at 37°C for 45 min. The coverslips were washed and placed in medium with 10 μg/ml gentamicin for 30 min to kill extracellular bacilli. Subsequently, the coverslips were washed again and placed in antibiotic-free medium with or without drugs. The time p.i. begins at the time of infection and ends at the time of cell fixation or assay. For the lactate dehydrogenase (LDH) release assay whose results are shown in Fig. 2B and C, medium was removed from the wells at the times indicated in Fig. 2B and C and assayed according to the manufacturer's protocol (Roche), and the results were read on a SpectraMax M5 plate reader (Molecular Devices). One hundred percent LDH release was obtained from uninfected cells lysed with 0.2% Triton X-100.

Drug treatment.

Drugs were added after removal of extracellular bacilli and for the remainder of the experiment until fixation or assay. The drug concentrations used were 75 μM (unless otherwise noted) for chloroquine diphosphate (CLQ), 250 nM for bafilomycin A (BAF), 30 mM for ammonium chloride (NH4Cl), 10 μg/ml for nocodazole (NOC), 75 μM for amodiaquine dihydrochloride dihydrate (ADQ), and 150 μM for primaquine bisphosphate (PMQ). All drugs were from Sigma-Aldrich.

Bacterial growth studies.

RAW 264.7 cells (8 × 105) were seeded on an 18-mm coverslip, infected the next day with B. thailandensis (at an MOI of 1 for the assay whose results are shown in Fig. 3A) for 45 min, and treated with 10 μg/ml gentamicin for 30 min to kill extracellular bacilli. The coverslips were washed as stated above and then incubated in antibiotic-free medium with or without 75 μM CLQ. For the assay whose results are shown in Fig. 3B, an MOI of 20 was used and CLQ was also added prior to infection for 1 h and during infection. To discount the possibility of extracellular growth of bacilli that escaped from cells, the coverslips were washed in PBS prior to cell lysis with 0.2% Triton X-100. Dilutions were plated on LB (Lennox) agar plates for determination of the number of CFU. For broth cultures for which the results are shown in Fig. 3C and to estimate the MOI, readings of the optical density at 600 nm were taken using a DU530 UV/visible spectrophotometer (Beckman).

Immunofluorescence staining, confocal microscopy, and live cell imaging.

Cells were fixed with 4% paraformaldehyde for 1 h and blocked in 0.25% saponin, 0.2% BSA fraction V, and goat or donkey serum (1:100; Sigma-Aldrich) for 2 h. Cells were incubated with rabbit anti-Burkholderia antibody (Ab; 1:1,000; kindly provided by David Waag, USAMRIID) and rat anti-LAMP1 Ab (ID4B; 1:200; Developmental Studies Hybridoma Bank) overnight at 4°C. After they were washed, the cells were incubated with secondary Ab conjugated to a fluorophore (1:600; Jackson ImmunoResearch) for 5 h. Cells were stained with 5 μg/ml DAPI (4′,6-diamidino-2-phenylindole) and phalloidin (1:80; Life Technologies) for 2 h prior to mounting with Fluoromount G mounting medium (Electron Microscopy Sciences). Different sets of coverslips were used to score for LAMP1 and actin. Cell preparation and staining for the following assays were performed according to the manufacturers' protocols: the (i) Click-iT TUNEL Alexa Fluor, (ii) Image-iT live lysosomal and nuclear labeling (LysoTracker red DND-99), and (iii) live/dead viability/cytotoxicity (calcein AM and EthD-1) assays (all from Life Technologies) and the autophagy/cytotoxicity dual staining (MDC and propidium iodide [PI]) assay (Cayman). For the assay whose results are shown in Fig. 6B, cells were pulsed with 10 μg/ml DQRed BSA (Life Technologies) for 1 h and chased for 3 h prior to live cell imaging.
Confocal microscopy and live cell imaging were performed on a Zeiss 700 confocal system using either a 40× (numerical aperture [NA], 1.3) or a 100× (NA, 1.4) oil objective lens and Zen 2012 (black edition) software. The pinhole was set to 1 Airy unit. For long-term live cell imaging (Movie S1), a stage-top incubator was used to maintain humidity, temperature, and CO2 levels.

Wright-Giemsa staining and light microscopy.

Permeabilized cells were subjected to Wright-Giemsa stain solution for 10 min, washed 3 times with PBS, and air dried. Light microscopy was performed on a Nikon E800 microscope equipped with a 60× (NA, 1.4) oil objective (Nikon) and a Microfire charge-coupled-device camera (Optronics), and the images were analyzed using Picture Frame software (Optronics).

Hcp1 and BopE expression, purification, and antibody production.

For expression of recombinant B. mallei Hcp1 with a C-terminal 6×His tag (rHcp1-6×His), the hcp1 open reading frame (BMAA0742) was PCR amplified from B. mallei ATCC 23344 genomic DNA using primer pair Bmhcp1-HisF1 (5′-CCCAACGGTCTCACATGCTGGCCGGAATATATCTCAAGG-3′) and Bmhcp1-HisR1 (5′-CCCAACGGTCTCAAGCTTCAATGATGATGATGATGATGCGCCGCCGCGCCATTCGTCCAGTTTGCGGC-3′); BsaI sites are underlined. For expression of recombinant B. pseudomallei BopE with a C-terminal 6×His tag (rBopE-6×His), a DNA fragment encoding amino acids 70 to 261 of BopE was PCR amplified from B. pseudomallei strain K96243 using primer pair BpbopE-HisF1 (5′-CCCAACGGTCTCACATGCGCTACGTCGGCAGCTATCG-3′) and BpbopE-HisR1 (5′-CCCAACGGTCTCAAGCTTCAATGATGATGATGATGATGCGCCGCCGCGCCGTCCGCCGCGTTCGTC-3′); BsaI sites are underlined. The resulting PCR products were digested with BsaI and cloned into pBAD/HisA digested with NcoI/HindIII, generating plasmids pBmhcp1-His and pBpbopE-His, respectively. rHcp1-6×His and rBopE-6×His were overexpressed in Escherichia coli TOP10 cells and purified essentially as previously described (48). Purified rHcp1-6×His and rBopE-6×His were used to raise Hcp1- and BopE-specific polyclonal antisera, respectively, in rabbits at Cocalico Biologicals, Inc., using a standard protocol. All procedures involving animals were performed in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Research Council (49). All protocols were approved by the Cocalico Biologicals, Inc., Animal Care and Use Committee.

Immunoblotting.

For the assay whose results are shown in Fig. 8A, C, and E, B. mallei or B. pseudomallei cultures were grown for 24 h at 37°C in M9 broth with 4% glycerol. Bacterial pellets were suspended in PBS and boiled for 30 min. A micro-bicinchoninic acid protein kit (Thermo Scientific) was used to quantify the proteins. Lysates (10 μg/lane) were run on NuPAGE 10% bis-Tris gel and transferred onto 0.45-μm-pore-size nitrocellulose membranes (Life Technologies). The membranes were blocked with Tris-buffered saline with 0.1% Tween 20 (TBST) and 5% nonfat milk for 2 h. Incubation with rabbit anti-BopC Ab (kindly given by Sunee Korbsrisate, Mahidol University, Bangkok, Thailand), rat anti-BopE Ab (Cocalico), mouse anti-TssM Ab (53), rabbit anti-Hcp1 Ab (Cocalico), mouse anti-DnaK Ab (Stressgen), or rabbit anti-GroEL Ab (Stressgen) occurred at 4°C overnight. All primary Abs were diluted between 1:3,000 and 1:5,000. Following washes with TBST, the membrane was subjected to horseradish peroxidase-anti-rabbit IgG, anti-mouse IgG, or anti-rat IgG Ab (1:10,000; all from Southern Biotech) for 1 h. The blots were visualized using SuperSignal West Pico chemiluminescent substrate (Thermo Scientific) and a Gel Logic Pro 6000 imaging system (Carestream). The blots were stripped using Restore Western blot stripping buffer (Thermo Scientific).

HC maintenance, infection, and treatment.

Madagascar hissing cockroaches (HCs; Gromphadorhina laevigata; Carolina Biological Supply) were housed in the dark at room temperature in mouse cages and fed Kibbles 'n Bits dog food (Big Heart Pet Brands), an occasional apple wedge, and water. The 50 lethal dose (LD50), determined in duplicate experiments (n = 2), was established using 8 HCs per group receiving increasing doses of B. thailandensis. The LD50 was calculated through Probit analysis using SAS software (version 9.3; SAS Institute, Cary, NC) and determined to be 11 CFU.
HCs (∼6 g) were acclimated at 37°C for 2 to 3 weeks. B. thailandensis (10 to 20 LD50s) was suspended in PBS alone or with 12 mg/ml CLQ in a 25-μl volume immediately prior to injection. The animals were infected as previously described (35), and deaths were recorded for 7 days. The dose of CLQ was calculated to provide 300 μg CLQ in 25 μl (50 mg/kg), and the HCs received one dose only. The use of HCs for research is not regulated by any department of the U.S. federal government or any animal use committee.

Statistical analysis.

Unless otherwise noted, statistical significance was calculated by an unpaired t test using the online QuickCalcs program at GraphPad. The ratios of the number of nuclei/number of cells in Fig. 1, 5, and 7 and the CFU counts in Fig. 3A and B were calculated using an asymptotic normal test based on a negative binomial regression model. Models of CFU counts included mixed effects to adjust for repeated measurements on biological replicates. The significance of the difference in 7-day survival between the B. thailandensis- and B. thailandensis-infected and CLQ-treated groups shown in Fig. 4 was also calculated using an asymptomatic normal test based on a generalized linear model having a binomial distribution, identity link, and fixed effect adjustment for animal cohort. All analyses were carried out with SAS software (version 9.4; SAS Institute, Cary, NC). In all figures, only those differences with P values of <0.05 are indicated.

ACKNOWLEDGMENTS

We thank Nicholas R. Carter, Nora D. Doyle, Kiley J. Duncan, Shane D. Falcinelli, and Steven A. Tobery for assistance, David P. Fetterer and Steven J. Kern for statistical analysis, and Donald J. Chabot and Rajendrakumar Lathigra for critical discussions.
The opinions, interpretations, conclusions, and recommendations are those of the authors and are not necessarily endorsed by the U.S. Army. The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, nor does mention of trade names, commercial products, or organizations imply endorsement by the U.S. Government.

Supplemental Material

File (zii999091934s2.pdf)
File (zii999091934sm1.avi)
ASM does not own the copyrights to Supplemental Material that may be linked to, or accessed through, an article. The authors have granted ASM a non-exclusive, world-wide license to publish the Supplemental Material files. Please contact the corresponding author directly for reuse.

REFERENCES

1.
Galyov EE, Brett PJ, and DeShazer D. 2010. Molecular insights into Burkholderia pseudomallei and Burkholderia mallei pathogenesis. Annu Rev Microbiol64:495–517.
2.
Cheng AC and Currie BJ. 2005. Melioidosis: epidemiology, pathophysiology, and management. Clin Microbiol Rev18:383–416.
3.
Holden MT, Titball RW, Peacock SJ, Cerdeno-Tarraga AM, Atkins T, Crossman LC, Pitt T, Churcher C, Mungall K, Bentley SD, Sebaihia M, Thomson NR, Bason N, Beacham IR, Brooks K, Brown KA, Brown NF, Challis GL, Cherevach I, Chillingworth T, Cronin A, Crossett B, Davis P, DeShazer D, Feltwell T, Fraser A, Hance Z, Hauser H, Holroyd S, Jagels K, Keith KE, Maddison M, Moule S, Price C, Quail MA, Rabbinowitsch E, Rutherford K, Sanders M, Simmonds M, Songsivilai S, Stevens K, Tumapa S, Vesaratchavest M, Whitehead S, Yeats C, Barrell BG, Oyston PC, and Parkhill J. 2004. Genomic plasticity of the causative agent of melioidosis, Burkholderia pseudomallei. Proc Natl Acad Sci U S A101:14240–14245.
4.
Gan YH. 2005. Interaction between Burkholderia pseudomallei and the host immune response: sleeping with the enemy?J Infect Dis192:1845–1850.
5.
Brett PJ, DeShazer D, and Woods DE. 1998. Burkholderia thailandensis sp. nov., a Burkholderia pseudomallei-like species. Int J Syst Bacteriol48(Pt 1):317–320.
6.
Glass MB, Gee JE, Steigerwalt AG, Cavuoti D, Barton T, Hardy RD, Godoy D, Spratt BG, Clark TA, and Wilkins PP. 2006. Pneumonia and septicemia caused by Burkholderia thailandensis in the United States. J Clin Microbiol44:4601–4604.
7.
Lertpatanasuwan N, Sermsri K, Petkaseam A, Trakulsomboon S, Thamlikitkul V, and Suputtamongkol Y. 1999. Arabinose-positive Burkholderia pseudomallei infection in humans: case report. Clin Infect Dis28:927–928.
8.
Dharakul T, Tassaneetrithep B, Trakulsomboon S, and Songsivilai S. 1999. Phylogenetic analysis of Ara+ and AraBurkholderia pseudomallei isolates and development of a multiplex PCR procedure for rapid discrimination between the two biotypes. J Clin Microbiol37:1906–1912.
9.
Moore RA, Reckseidler-Zenteno S, Kim H, Nierman W, Yu Y, Tuanyok A, Warawa J, DeShazer D, and Woods DE. 2004. Contribution of gene loss to the pathogenic evolution of Burkholderia pseudomallei and Burkholderia mallei. Infect Immun72:4172–4187.
10.
Heiss C, Burtnick MN, Wang Z, Azadi P, and Brett PJ. 2012. Structural analysis of capsular polysaccharides expressed by Burkholderia mallei and Burkholderia pseudomallei. Carbohydr Res349:90–94.
11.
Atkins T, Prior R, Mack K, Russell P, Nelson M, Prior J, Ellis J, Oyston PC, Dougan G, and Titball RW. 2002. Characterisation of an acapsular mutant of Burkholderia pseudomallei identified by signature tagged mutagenesis. J Med Microbiol51:539–547.
12.
West TE, Frevert CW, Liggitt HD, and Skerrett SJ. 2008. Inhalation of Burkholderia thailandensis results in lethal necrotizing pneumonia in mice: a surrogate model for pneumonic melioidosis. Trans R Soc Trop Med Hyg102(Suppl 1):S119–S126.
13.
Haraga A, West TE, Brittnacher MJ, Skerrett SJ, and Miller SI. 2008. Burkholderia thailandensis as a model system for the study of the virulence-associated type III secretion system of Burkholderia pseudomallei. Infect Immun76:5402–5411.
14.
Pilatova M and Dionne MS. 2012. Burkholderia thailandensis is virulent in Drosophila melanogaster. PLoS One7:e49745.
15.
Allwood EM, Devenish RJ, Prescott M, Adler B, and Boyce JD. 2011. Strategies for intracellular survival of Burkholderia pseudomallei. Front Microbiol2:170.
16.
Cullinane M, Gong L, Li X, Lazar-Adler N, Tra T, Wolvetang E, Prescott M, Boyce JD, Devenish RJ, and Adler B. 2008. Stimulation of autophagy suppresses the intracellular survival of Burkholderia pseudomallei in mammalian cell lines. Autophagy4:744–753.
17.
Srinon V, Muangman S, Imyaem N, Muangsombut V, Lazar Adler NR, Galyov EE, and Korbsrisate S. 2013. Comparative assessment of the intracellular survival of the Burkholderia pseudomallei bopC mutant. J Microbiol51:522–526.
18.
French CT, Toesca IJ, Wu TH, Teslaa T, Beaty SM, Wong W, Liu M, Schroder I, Chiou PY, Teitell MA, and Miller JF. 2011. Dissection of the Burkholderia intracellular life cycle using a photothermal nanoblade. Proc Natl Acad Sci U S A108:12095–12100.
19.
Burtnick MN, Brett PJ, Harding SV, Ngugi SA, Ribot WJ, Chantratita N, Scorpio A, Milne TS, Dean RE, Fritz DL, Peacock SJ, Prior JL, Atkins TP, and DeShazer D. 2011. The cluster 1 type VI secretion system is a major virulence determinant in Burkholderia pseudomallei. Infect Immun79:1512–1525.
20.
Schell MA, Ulrich RL, Ribot WJ, Brueggemann EE, Hines HB, Chen D, Lipscomb L, Kim HS, Mrazek J, Nierman WC, and DeShazer D. 2007. Type VI secretion is a major virulence determinant in Burkholderia mallei. Mol Microbiol64:1466–1485.
21.
Duval CW and White PG. 1907. The histological lesions of experimental glanders. J Exp Med9:352–380.
22.
Wong KT, Puthucheary SD, and Vadivelu J. 1995. The histopathology of human melioidosis. Histopathology26:51–55.
23.
Coleman W and Ewing J. 1903. A case of septicemic glanders in the human subject. J Med Res9:223–240.
24.
Wiersinga WJ, van der Poll T, White NJ, Day NP, and Peacock SJ. 2006. Melioidosis: insights into the pathogenicity of Burkholderia pseudomallei. Nat Rev Microbiol4:272–282.
25.
Toesca IJ, French CT, and Miller JF. 2014. The type VI secretion system spike protein VgrG5 mediates membrane fusion during intercellular spread by pseudomallei group Burkholderia species. Infect Immun82:1436–1444.
26.
Kunisch R, Kamal E, and Lewin A. 2012. The role of the mycobacterial DNA-binding protein 1 (MDP1) from Mycobacterium bovis BCG in host cell interaction. BMC Microbiol12:165.
27.
Stone JK, Johnson SL, Bruce DC, Detter JC, Mayo M, Currie BJ, Gelhaus HC, Keim P, and Tuanyok A. 2013. Complete genome sequence of the encephalomyelitic Burkholderia pseudomallei strain MSHR305. Genome Announc1(4):e00656-13.
28.
McNally AK and Anderson JM. 2005. Multinucleated giant cell formation exhibits features of phagocytosis with participation of the endoplasmic reticulum. Exp Mol Pathol79:126–135.
29.
Brett PJ, Burtnick MN, Su H, Nair V, and Gherardini FC. 2008. iNOS activity is critical for the clearance of Burkholderia mallei from infected RAW 264.7 murine macrophages. Cell Microbiol10:487–498.
30.
Sun GW, Lu J, Pervaiz S, Cao WP, and Gan YH. 2005. Caspase-1 dependent macrophage death induced by Burkholderia pseudomallei. Cell Microbiol7:1447–1458.
31.
Bast A, Krause K, Schmidt IH, Pudla M, Brakopp S, Hopf V, Breitbach K, and Steinmetz I. 2014. Caspase-1-dependent and -independent cell death pathways in Burkholderia pseudomallei infection of macrophages. PLoS Pathog10:e1003986.
32.
Sifri CD and Ausubel FM. 2005. Use of simple non-vertebrate hosts to model mammalian pathogenesis, p 543–563. InCossart P, Boquet P, Normark S, and Rappuoli R (ed), Cellular microbiology, 2nd ed. ASM Press, Washington, DC.
33.
Lavine MD and Strand MR. 2002. Insect hemocytes and their role in immunity. Insect Biochem Mol Biol32:1295–1309.
34.
Kavanagh K and Reeves EP. 2004. Exploiting the potential of insects for in vivo pathogenicity testing of microbial pathogens. FEMS Microbiol Rev28:101–112.
35.
Fisher NA, Ribot WJ, Applefeld W, and DeShazer D. 2012. The Madagascar hissing cockroach as a novel surrogate host for Burkholderia pseudomallei, B. mallei and B. thailandensisBMC Microbiol12:117.
36.
Vinod V, Padmakrishnan CJ, Vijayan B, and Gopala S. 2014. ‘How can I halt thee?’ The puzzles involved in autophagic inhibition. Pharmacol Res82:1–8.
37.
Li Q and Weina PJ. 2011. Antimalarial drugs, p 622. InAge of the artemisinins. Nova Science Publishers, Inc., New York, NY.
38.
Jitprasutwit S, Thaewpia W, Muangsombut V, Lulitanond A, Leelayuwat C, Lertmemongkolchai G, and Korbsrisate S. 2010. Effect of acidic pH on the invasion efficiency and the type III secretion system of Burkholderia thailandensis. J Microbiol48:526–532.
39.
Wong J, Chen Y, and Gan YH. 2015. Host cytosolic glutathione sensing by a membrane histidine kinase activates the type VI secretion system in an intracellular bacterium. Cell Host Microbe18:38–48.
40.
Tan KS, Chen Y, Lim YC, Tan GY, Liu Y, Lim YT, Macary P, and Gan YH. 2010. Suppression of host innate immune response by Burkholderia pseudomallei through the virulence factor TssM. J Immunol184:5160–5171.
41.
Utaisincharoen P, Kespichayawattana W, Anuntagool N, Chaisuriya P, Pichyangkul S, Krieg AM, and Sirisinha S. 2003. CpG ODN enhances uptake of bacteria by mouse macrophages. Clin Exp Immunol132:70–75.
42.
Cho JY. 2008. Chloroquine, an anti-malarial agent, acts as a novel regulator of beta 1-integrin-mediated cell-cell adhesion. Biol Pharm Bull31:726–730.
43.
Trotta RF, Brown ML, Terrell JC, and Geyer JA. 2004. Defective DNA repair as a potential mechanism for the rapid development of drug resistance in Plasmodium falciparum. Biochemistry43:4885–4891.
44.
Suparak S, Muangsombut V, Riyapa D, Stevens JM, Stevens MP, Lertmemongkolchai G, and Korbsrisate S. 2011. Burkholderia pseudomallei-induced cell fusion in U937 macrophages can be inhibited by monoclonal antibodies against host cell surface molecules. Microb Infect13:1006–1011.
45.
Boddey JA, Day CJ, Flegg CP, Ulrich RL, Stephens SR, Beacham IR, Morrison NA, and Peak IR. 2007. The bacterial gene lfpA influences the potent induction of calcitonin receptor and osteoclast-related genes in Burkholderia pseudomallei-induced TRAP-positive multinucleated giant cells. Cell Microbiol9:514–531.
46.
Lee SH, Rho J, Jeong D, Sul JY, Kim T, Kim N, Kang JS, Miyamoto T, Suda T, Lee SK, Pignolo RJ, Koczon-Jaremko B, Lorenzo J, and Choi Y. 2006. v-ATPase V0 subunit d2-deficient mice exhibit impaired osteoclast fusion and increased bone formation. Nat Med12:1403–1409.
47.
Maurin M and Raoult D. 1994. Phagolysosomal alkalinization and intracellular killing of Staphylococcus aureus by amikacin. J Infect Dis169:330–336.
48.
Burtnick MN and Brett PJ. 2013. Burkholderia mallei and Burkholderia pseudomallei cluster 1 type VI secretion system gene expression is negatively regulated by iron and zinc. PLoS One8:e76767?xpp zrp [.
49.
National Research Council. 2011. Guide for the care and use of laboratory animals, 8th ed. National Academies Press, Washington, DC.
50.
Nierman WC, DeShazer D, Kim HS, Tettelin H, Nelson KE, Feldblyum T, Ulrich RL, Ronning CM, Brinkac LM, Daugherty SC, Davidsen TD, Deboy RT, Dimitrov G, Dodson RJ, Durkin AS, Gwinn ML, Haft DH, Khouri H, Kolonay JF, Madupu R, Mohammoud Y, Nelson WC, Radune D, Romero CM, Sarria S, Selengut J, Shamblin C, Sullivan SA, White O, Yu Y, Zafar N, Zhou L, and Fraser CM. 2004. Structural flexibility in the Burkholderia mallei genome. Proc Natl Acad Sci U S A101:14246–14251.
51.
Hayden HS, Lim R, Brittnacher MJ, Sims EH, Ramage ER, Fong C, Wu Z, Crist E, Chang J, Zhou Y, Radey M, Rohmer L, Haugen E, Gillett W, Wuthiekanun V, Peacock SJ, Kaul R, Miller SI, Manoil C, and Jacobs MA. 2012. Evolution of Burkholderia pseudomallei in recurrent melioidosis. PLoS One7:e36507.
52.
Burtnick M, Bolton A, Brett P, Watanabe D, and Woods D. 2001. Identification of the acid phosphatase (acpA) gene homologues in pathogenic and non-pathogenic Burkholderia spp. facilitates TnphoA mutagenesis. Microbiology147:111–120.
53.
Shanks J, Burtnick MN, Brett PJ, Waag DM, Spurgers KB, Ribot WJ, Schell MA, Panchal RG, Gherardini FC, Wilkinson KD, and DeShazer D. 2009. Burkholderia mallei tssM encodes a putative deubiquitinase that is secreted and expressed inside infected RAW 264.7 murine macrophages. Infect Immun77:1636–1648.

Information & Contributors

Information

Published In

cover image Infection and Immunity
Infection and Immunity
Volume 85Number 1January 2017
eLocator: 10.1128/iai.00586-16
Editor: Shelley M. Payne, University of Texas at Austin

History

Received: 11 July 2016
Returned for modification: 24 August 2016
Accepted: 23 October 2016
Published online: 29 December 2016

Permissions

Request permissions for this article.

Keywords

  1. Burkholderia
  2. Madagascar hissing cockroaches
  3. type 3 secretion system
  4. type 6 secretion system
  5. acidification
  6. actin tails
  7. autophagy
  8. chloroquine
  9. multinucleated giant cells
  10. phagosomal escape

Contributors

Authors

Jennifer Chua
Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, USA
Jeffrey L. Senft
Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, USA
Stephen J. Lockett
Optical Microscopy and Analysis Laboratory, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
Paul J. Brett
Department of Microbiology and Immunology, University of South Alabama, Mobile, Alabama, USA
Mary N. Burtnick
Department of Microbiology and Immunology, University of South Alabama, Mobile, Alabama, USA
David DeShazer
Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, USA
Arthur M. Friedlander
Bacteriology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, USA
Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA

Editor

Shelley M. Payne
Editor
University of Texas at Austin

Notes

Address correspondence to Jennifer Chua, [email protected], or Arthur M. Friedlander, [email protected].

Metrics & Citations

Metrics

Note: There is a 3- to 4-day delay in article usage, so article usage will not appear immediately after publication.

Citation counts come from the Crossref Cited by service.

Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Simply select your manager software from the list below and click Download.

View Options

Figures and Media

Figures

Media

Tables

Share

Share

Share the article link

Share with email

Email a colleague

Share on social media

American Society for Microbiology ("ASM") is committed to maintaining your confidence and trust with respect to the information we collect from you on websites owned and operated by ASM ("ASM Web Sites") and other sources. This Privacy Policy sets forth the information we collect about you, how we use this information and the choices you have about how we use such information.
FIND OUT MORE about the privacy policy