Coxiella burnetii-Infected NK Cells Release Infectious Bacteria by Degranulation

Svea Matthiesen, Luca Zaeck, Kati Franzke, Rico Jahnke, Charlie Fricke, Michael Mauermeir, Stefan Finke, Anja Lührmann, Michael R Knittler, Svea Matthiesen, Luca Zaeck, Kati Franzke, Rico Jahnke, Charlie Fricke, Michael Mauermeir, Stefan Finke, Anja Lührmann, Michael R Knittler

Abstract

Natural killer (NK) cells are critically involved in the early immune response against various intracellular pathogens, including Coxiella burnetii and Chlamydia psittaciChlamydia-infected NK cells functionally mature, induce cellular immunity, and protect themselves by killing the bacteria in secreted granules. Here, we report that infected NK cells do not allow intracellular multiday growth of Coxiella, as is usually observed in other host cell types. C. burnetii-infected NK cells display maturation and gamma interferon (IFN-γ) secretion, as well as the release of Coxiella-containing lytic granules. Thus, NK cells possess a potent program to restrain and expel different types of invading bacteria via degranulation. Strikingly, though, in contrast to Chlamydia, expulsed Coxiella organisms largely retain their infectivity and, hence, escape the cell-autonomous self-defense mechanism in NK cells.

Keywords: Chlamydia psittaci; Coxiella burnetii; NK cells; cell-autonomous immunity.

Copyright © 2020 American Society for Microbiology.

Figures

FIG 1
FIG 1
Uptake and functional activation of KY-2 cells during C. burnetii infection. (a) Flow cytometric analysis of infected KY-2 cells (72 hpi; MOI, 10) in the absence or presence of MDC (200 μM). To quantify C. burnetii-positive NK cells (green), the negative cell population (black) was identified and gated via corresponding noninfected controls and then subtracted from the total cell population (left). The corresponding plot (right) shows the amount of positive KY-2 cells (***, P < 0.001 versus control [infected without treatment]; n = 3; mean ± SD). (b) To identify intra- and extracellular bacteria at 8 hpi, infected NK cells were permeabilized or not and then exposed to Coxiella-specific antiserum. Afterwards, cells were analyzed by immunofluorescence microscopy (left). The Western blot (middle) of infected KY-2 cells (0 to 72 hpi; MOI, 10) shows coxHSP60 in cell lysates (intracellular) and culture supernatants (extracellular). GAPDH served as a loading control. coxHSP60 signals were determined by densitometric analysis (right). The signal of total coxHSP60 at 24 hpi was set to 100. (c) KY-2 cells were infected or not with Coxiella (0 to 72 hpi; MOI, 10) and stained for Coxiella (green) and DNA (4′,6-diamidino-2-phenylindole [DAPI]; blue). (d) Flow cytometric analysis of C. burnetii infection in KY-2 cells (0 to 72 hpi; MOI, 10). The plot displays the relative amount of infected cells. The maximum mean value at 24 hpi was set to 1 (**, P < 0.01, and ***, P < 0.001, versus 24 hpi; n = 3; mean ± SD). (e) Analysis of necrotic/apoptotic KY-2 cells during Coxiella infection (0 to 72 hpi; MOI, 10) was performed via trypan blue staining (not significant [n.s.] versus 0 hpi; n = 3; mean ± SD). (f) Long-term time-lapse recording of Coxiella release from infected NK cells. A total of 1 × 105 NK cells were infected with C. burnetii (tdTomato; MOI, 30; washed 3 hpi), and LCI was started at 24 hpi. Bacterial movement was tracked at 37°C. Trajectories were determined from the image series. The timestamp is relative to the start of the respective image series and thus starts at 0 min. The upper and lower rows correspond to starting time points at 24 hpi and 26 hpi, respectively. The differently colored lines (green and magenta) show the individual Coxiella trajectories over periods of 510 and 800 min. The corresponding time-lapse movies are included in the supplemental material (Movies S1 and S2).
FIG 2
FIG 2
Short-interval time-lapse image series of C. burnetii-release from an infected NK cell. A total of 1 × 105 NK cells were infected with C. burnetii (tdTomato; MOI, 30; washed 3 hpi) and overlaid with 0.6% low-melting-point-agarose-containing KY-2 cell culture medium at 20 hpi. The time-lapse image series shows a period of about 21.5 min, starting at about 31.5 hpi. Bacterial movement during release was tracked at 37°C, and the maximum z-projection of the fluorescence channel (C. burnetii, red) was merged with a single representative plane of the bright-field channel. Trajectories are shown as green lines. The first row displays selected bright-field images of the LCI analysis. The second row shows the overlay of the selected bright-field images with maximum z-projection of red fluorescent bacteria. The third and fourth rows depict corresponding detail views of the bacterial exit process as bright-field images and as overlays with red fluorescent bacteria. The time-lapse movie is included in the supplemental material (Movie S3).
FIG 3
FIG 3
Activation of KY-2 cells during C. burnetii infection. (a) Flow cytometric analysis of NK cell degranulation via staining of surface CD107a/LAMP1 (left) on noninfected and C. burnetii-infected KY-2 cells (24 hpi; MOI, 10). KY-2 cell endocytosis was inhibited 12 hpi by MDC to stabilize surface-expressed CD107a/LAMP1. Cells were first stained for surface CD107a/LAMP1 and then fixed. After mild permeabilization, pretreated cells were stained for intracellular bacteria. The corresponding histogram plot (right) shows the relative increase of the surface CD107a/LAMP1 (noninfected controls were set to 1 (*, P < 0.05 versus control [noninfected]; n = 3; mean ± SD). (b) Secretion of Grzm B (left) and IFN-γ (right) of infected KY-2 cells (0 to 72 hpi; MOI, 10) measured by ELISA (**, P < 0.01, and ****, P < 0.0001, versus noninfected control; n = 3; mean ± SD). (c) Analysis of PKCϴ phosphoactivation during KY-2 cell infection. KY-2 cells were infected or not with C. burnetii (MOI, 10) for 72 h and analyzed by Western blots probed for P-PKCϴ, PKCϴ, and coxHSP60 (top). GAPDH served as a loading control. After densitometric analysis (bottom), the P-PKCϴ/PKCϴ ratio was plotted (gray histogram) and compared to the control of noninfected KY-2 cells (black histogram; controls were set to 1; *, P < 0.05, versus noninfected control; n = 3; ± SD). (d) Western blot analysis of coxHSP60 in infected KY-2 cells (MOI 10) (top) and culture supernatants (bottom) in the absence or presence of PKCϴ inhibitor sotrastaurin (250 nM) 24 hpi and 72 hpi. GAPDH and Ponceau-S staining served as loading controls for cell lysates and culture supernatants, respectively.
FIG 4
FIG 4
Characterization of intracellular C. burnetii in infected KY-2 cells. (a) TEM of noninfected (control) and C. burnetii-infected (24 hpi and 72 hpi; MOI, 10) KY-2 cells. Arrowheads indicate cell surface membrane-associated Coxiella structures. (b) TEM of C. burnetii-infected (24 hpi; MOI, 10) KY-2 cells. Arrowheads indicate electron-dense intracellular Coxiella structures associated with secretory granules of KY-2 cells. (c) Immunofluorescence analysis of perforin (green) and C. burnetii (red) in infected (24 hpi; MOI, 10) and noninfected KY-2 cells. DNA was stained via DAPI (blue).
FIG 5
FIG 5
Characterization of extracellular C. burnetii and C. psittaci released by infected KY-2 cells. (a) The left image depicts an immunofluorescence analysis showing colocalization of perforin (green) and KY-2 cell-released C. burnetii (red) 24 hpi (MOI, 10). A TEM analysis of NK cell-released bacterial structures (72 hpi; MOI, 10) is shown in the middle image. Black stars indicate intact C. burnetii, whereas black diamonds mark largely deformed, aberrant bacterial structures. The TEM analysis of purified C. burnetii stocks from infected L929 cells containing LCVs and SCVs of C. burnetii is shown in the right image. (b) The first image depicts a TEM analysis of NK cell-released chlamydial structures. The inset highlights an immunofluorescence analysis showing colocalization of perforin (green) and KY-2-released C. psittaci (red) at 24 hpi (MOI, 10). A TEM analysis of an infectious C. psittaci stock is shown in the second image from the left. The two images on the right show enlarged TEM pictures of Chlamydia, either released from NK cells by degranulation or harvested from an infectious stock culture.
FIG 6
FIG 6
C. burnetii survives the cellular defense (granule uptake and release) of infected NK cells. (a) C. burnetii and C. psittaci were preincubated in citrate buffer at pH 4 and 5 or in Tris buffer at pH 7 (control) for 18 h at 4°C. After infection (MOI, 10) of suitable reporter cells (L929 and BGM; 72 hpi), infectivity was measured by flow cytometry (top). Bacterium-positive cells (green) were identified and gated (left) as described in the legend to Fig. 1. The corresponding histogram plots (right) show the number of positive cells (controls were set to 1 [infected without preincubation]; *, P < 0.05, **, P < 0.01, and ***, P < 0.001, versus control; n = 3; mean ± SD). (b) Western blot analysis of cell infection after treatment of purified Chlamydia or Coxiella with Grzm B. Both pathogens were incubated with Grzm B or left untreated. The bacteria were then washed and used for the infection of reporter cells (L929 for Coxiella and BGM for Chlamydia; 72 hpi; MOI, 30). Bacterial HSP60 (bactHSP60) signal intensities were measured by densitometric analysis (left). GAPDH served as a loading control. The obtained results of three independent experiments are depicted as a histogram plot (right) (signals of untreated samples were set to 1; n.s., ***, P < 0.001, versus untreated samples; n = 3; mean ± SD). (c) Flow cytometry of the infectivity of culture supernatants (sup.) from KY-2 cells. KY-2 cells were infected with bacteria (Chlamydia or Coxiella; MOI, 10 [10 IFU/cell]) or not for 72 h. GEs were determined by qPCR. To compare the infectivity of original stocks and NK cell-released bacteria, equal amounts of GEs (Coxiella and Chlamydia infections, 70 GEs/cell) were used in parallel experiments for reporter cell infection (L929 and BGM). Bacterium-positive cells (green) were identified and gated (upper panel) as described in the legend to Fig. 1. The corresponding histogram plot (bottom) shows the relative number of infected reporter cells (controls were set to 1 [original stocks]; *, P < 0.05, and ***, P < 0.001, versus controls; n = 3; mean ± SD).
FIG 7
FIG 7
Growth/replication of Coxiella released from KY-2 cells. (a) Flow cytometry of the Coxiella growth/replication in L929 cells infected with bacteria expulsed by KY-2 (top). NK cells were infected with C. burnetii (MOI, 10 [10 IFU/cell]) or not for 72 h. Bacteria from corresponding culture supernatants were centrifuged and washed. GEs of the pellet fractions were determined by qPCR, and 70 GEs/cell were used for L929 cell infection and analyzed for different time points (control, 24 to 72 h). Bacterium-positive cells (green) were identified and gated (top) as described in the legend to Fig. 1. The corresponding histogram plots (bottom) show the relative number of infected reporter cells (control, 24 to 72 hpi) (values obtained for 72 hpi were set to 1; **, P < 0.01, and ***, P < 0.001, versus controls; n = 3; mean ± SD). (b) Immunofluorescence analysis of C. burnetii (green) in infected and reinfected (70 GEs/cell) L929 cells (72 hpi). DNA was stained via DAPI (blue). (c) Successive reinfection of L929 cells. Centrifuged/washed bacteria expulsed by infected KY-2 cells were used for an L929 cell reinfection assay (1. reinfection, 70 GEs/cell). After 72 h, one-half of the infected cells was examined for infection by flow cytometry. The other half was homogenized to harvest Coxiella for a second reinfection round in L929 cells (2. reinfection). As with the 1. reinfections, flow cytometry was performed after 72 hpi.
FIG 8
FIG 8
C. burnetii infection of primary NK cells. (a) Western blot of infected primary NK cells (0 to 72 hpi; MOI, 10) showing coxHSP60 in cell lysates (intracellular) and culture supernatants (extracellular) (left). GAPDH served as a loading control. coxHSP60 signals were determined by densitometric analysis (right). The signal of total coxHSP60 at 24 hpi was set to 100. (b) Flow cytometric analysis of C. burnetii infection in KY-2 cells (0 to 72 hpi; MOI, 10). The depicted plot displays the relative amount of infected cells. The maximum value at 24 hpi was set to 1 (**, P < 0.01, and ***, P < 0.001, versus 24 hpi; n = 3; ± SD). (c) Necrotic/apoptotic KY-2 cells during Coxiella infection (0 to 72 hpi; MOI, 10) were identified by trypan blue staining (n.s. versus 0 hpi; n = 3; mean ± SD). (d) Western blot and protein staining of coxHSP60 in infected primary NK cells (MOI, 10) as well as culture supernatants in the presence of sotrastaurin (72 hpi). Ponceau-S staining served as a loading control. (e) Immunofluorescence microscopy shows colocalization of perforin (green) and C. burnetii (red) in infected primary NK cells (24 hpi; MOI 20) (left) and for NK cell-released C. burnetii (12 hpi, MOI 20) (right). DNA was stained with DAPI (blue). (f) Flow cytometry of the infectivity of culture supernatants from primary NK cells. Primary NK cells were infected with bacteria (Chlamydia or Coxiella; MOI, 10) or not for 72 h. GEs were determined by qPCR. Equal amounts of GEs (Coxiella and Chlamydia infections, 70 GEs/cell) were used in parallel experiments for reporter cell infection. The corresponding histogram plot shows the relative number of infected reporter cells (controls were set to 1 [original stocks]; **, P < 0.01, and ***, P < 0.001, versus controls; n = 3; mean ± SD).
FIG 9
FIG 9
Growth/replication of Coxiella released from primary NK cells. (a) Flow cytometry of the Coxiella growth/replication in L929 cells infected with bacteria expulsed by primary NK cells (top). NK cells were infected with C. burnetii (MOI, 10 [10 IFU/cell]) or not for 72 h. GEs were determined by qPCR, and 70 GEs/cell were used for L929 cell infection and analyzed for different periods (control, 24 to 72 h). Bacterium-positive cells (green) were identified and gated (top) as described in the legend to Fig. 1. The corresponding histogram plots (bottom) show the relative number of infected reporter cells (control, 24 to 72 hpi) (values obtained for 72 hpi were set to 1; **, P < 0.01, and ***, P < 0.001, versus controls; n = 3; mean ± SD). (b) Immunofluorescence analysis of C. burnetii (green) in infected and reinfected (70 GEs/cell) L929 cells (72 hpi). DNA was stained via DAPI (blue). c) Successive reinfection of L929 cells. Centrifuged/washed bacteria expulsed by infected primary NK cells were used for an L929 reinfection assay (1. Reinfection, 70 GEs/cell). After 72 h, one-half of the infected cells was examined for infection by flow cytometry. The other half was homogenized to harvest Coxiella for a second reinfection round in L929 cells (2. reinfection). As with the 1. reinfections, flow cytometry was performed after 72 hpi.
FIG 10
FIG 10
Working model of bacterial escape from cellular self-defense of C. burnetii-infected NK cells. The depicted model shows the transient C. burnetii-infection of NK cells triggering activation, cytokine secretion, bacterial granule fusion, and release of infectious Coxiella.

References

    1. Topham NJ, Hewitt EW. 2009. Natural killer cell cytotoxicity: how do they pull the trigger? Immunology 128:7–15. doi:10.1111/j.1365-2567.2009.03123.x.
    1. Jiao L, Gao X, Joyee AG, Zhao L, Qiu H, Yang M, Fan Y, Wang S, Yang X. 2011. NK cells promote type 1 T cell immunity through modulating the function of dendritic cells during intracellular bacterial infection. J Immunol 187:401–411. doi:10.4049/jimmunol.1002519.
    1. Li J, Dong X, Zhao L, Wang X, Wang Y, Yang X, Wang H, Zhao W. 2016. Natural killer cells regulate Th1/Treg and Th17/Treg balance in chlamydial lung infection. J Cell Mol Med 20:1339–1351. doi:10.1111/jcmm.12821.
    1. Tseng CT, Rank RG. 1998. Role of NK cells in early host response to chlamydial genital infection. Infect Immun 66:5867–5875. doi:10.1128/IAI.66.12.5867-5875.1998.
    1. Radomski N, Franzke K, Matthiesen S, Karger A, Knittler MR. 2019. NK cell-mediated processing of Chlamydia psittaci drives potent anti-bacterial Th1 immunity. Sci Rep 9:4799. doi:10.1038/s41598-019-41264-4.
    1. Paolini R, Bernardini G, Molfetta R, Santoni A. 2015. NK cells and interferons. Cytokine Growth Factor Rev 26:113–120. doi:10.1016/j.cytogfr.2014.11.003.
    1. Hewitt EW. 2003. The MHC class I antigen presentation pathway: strategies for viral immune evasion. Immunology 110:163–169. doi:10.1046/j.1365-2567.2003.01738.x.
    1. Shafer WM, Pohl J, Onunka VC, Bangalore N, Travis J. 1991. Human lysosomal cathepsin G and granzyme B share a functionally conserved broad spectrum antibacterial peptide. J Biol Chem 266:112–116.
    1. Trapani JA, Smyth MJ. 2002. Functional significance of the perforin/granzyme cell death pathway. Nat Rev Immunol 2:735–747. doi:10.1038/nri911.
    1. Smyth MJ, Kelly JM, Sutton VR, Davis JE, Browne KA, Sayers TJ, Trapani JA. 2001. Unlocking the secrets of cytotoxic granule proteins. J Leukoc Biol 70:18–29.
    1. Dotiwala F, Sen Santara S, Binker-Cosen AA, Li B, Chandrasekaran S, Lieberman J. 2017. Granzyme B disrupts central metabolism and protein synthesis in bacteria to promote an immune cell death program. Cell 171:1125–1137.e11. doi:10.1016/j.cell.2017.10.004.
    1. Bertzbach LD, van Haarlem DA, Hartle S, Kaufer BB, Jansen CA. 2019. Marek’s disease virus infection of natural killer cells. Microorganisms 7:588. doi:10.3390/microorganisms7120588.
    1. Paul S, Lal G. 2017. The molecular mechanism of natural killer cells function and its importance in cancer immunotherapy. Front Immunol 8:1124. doi:10.3389/fimmu.2017.01124.
    1. Pegram HJ, Andrews DM, Smyth MJ, Darcy PK, Kershaw MH. 2011. Activating and inhibitory receptors of natural killer cells. Immunol Cell Biol 89:216–224. doi:10.1038/icb.2010.78.
    1. Cooper MA, Colonna M, Yokoyama WM. 2009. Hidden talents of natural killers: NK cells in innate and adaptive immunity. EMBO Rep 10:1103–1110. doi:10.1038/embor.2009.203.
    1. Zwirner NW, Domaica CI. 2010. Cytokine regulation of natural killer cell effector functions. Biofactors 36:274–288. doi:10.1002/biof.107.
    1. Chalifour A, Jeannin P, Gauchat JF, Blaecke A, Malissard M, N'Guyen T, Thieblemont N, Delneste Y. 2004. Direct bacterial protein PAMP recognition by human NK cells involves TLRs and triggers alpha-defensin production. Blood 104:1778–1783. doi:10.1182/blood-2003-08-2820.
    1. Marcenaro E, Ferranti B, Falco M, Moretta L, Moretta A. 2008. Human NK cells directly recognize Mycobacterium bovis via TLR2 and acquire the ability to kill monocyte-derived DC. Int Immunol 20:1155–1167. doi:10.1093/intimm/dxn073.
    1. Lindemann RA. 1988. Bacterial activation of human natural killer cells: role of cell surface lipopolysaccharide. Infect Immun 56:1301–1308. doi:10.1128/IAI.56.5.1301-1308.1988.
    1. Hart OM, Athie-Morales V, O'Connor GM, Gardiner CM. 2005. TLR7/8-mediated activation of human NK cells results in accessory cell-dependent IFN-gamma production. J Immunol 175:1636–1642. doi:10.4049/jimmunol.175.3.1636.
    1. Qiu F, Maniar A, Diaz MQ, Chapoval AI, Medvedev AE. 2011. Activation of cytokine-producing and antitumor activities of natural killer cells and macrophages by engagement of Toll-like and NOD-like receptors. Innate Immun 17:375–387. doi:10.1177/1753425910372000.
    1. Anel A, Aguilo JI, Catalan E, Garaude J, Rathore MG, Pardo J, Villalba M. 2012. Protein kinase c-theta (pkc-theta) in natural killer cell function and anti-tumor immunity. Front Immunol 3:187. doi:10.3389/fimmu.2012.00187.
    1. Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. 2008. Functions of natural killer cells. Nat Immunol 9:503–510. doi:10.1038/ni1582.
    1. Vasilevsky S, Greub G, Nardelli-Haefliger D, Baud D. 2014. Genital Chlamydia trachomatis: understanding the roles of innate and adaptive immunity in vaccine research. Clin Microbiol Rev 27:346–370. doi:10.1128/CMR.00105-13.
    1. Zhong GM, de la Maza LM. 1988. Activation of mouse peritoneal macrophages in vitro or in vivo by recombinant murine gamma interferon inhibits the growth of Chlamydia trachomatis serovar L1. Infect Immun 56:3322–3325. doi:10.1128/IAI.56.12.3322-3325.1988.
    1. Roan NR, Starnbach MN. 2008. Immune-mediated control of Chlamydia infection. Cell Microbiol 10:9–19. doi:10.1111/j.1462-5822.2007.01069.x.
    1. Eldin C, Melenotte C, Mediannikov O, Ghigo E, Million M, Edouard S, Mege JL, Maurin M, Raoult D. 2017. From Q fever to Coxiella burnetii infection: a paradigm change. Clin Microbiol Rev 30:115–190. doi:10.1128/CMR.00045-16.
    1. McQuiston JH, Childs JE. 2002. Q fever in humans and animals in the United States. Vector Borne Zoonotic Dis 2:179–191. doi:10.1089/15303660260613747.
    1. Shannon JG, Heinzen RA. 2009. Adaptive immunity to the obligate intracellular pathogen Coxiella burnetii. Immunol Res 43:138–148. doi:10.1007/s12026-008-8059-4.
    1. Voth DE, Heinzen RA. 2007. Lounging in a lysosome: the intracellular lifestyle of Coxiella burnetii. Cell Microbiol 9:829–840. doi:10.1111/j.1462-5822.2007.00901.x.
    1. Maurin M, Raoult D. 1999. Q fever. Clin Microbiol Rev 12:518–553. doi:10.1128/CMR.12.4.518.
    1. Frankel D, Richet H, Renvoise A, Raoult D. 2011. Q fever in France, 1985–2009. Emerg Infect Dis 17:350–356. doi:10.3201/eid1703.100882.
    1. Shannon JG, Howe D, Heinzen RA. 2005. Virulent Coxiella burnetii does not activate human dendritic cells: role of lipopolysaccharide as a shielding molecule. Proc Natl Acad Sci U S A 102:8722–8727. doi:10.1073/pnas.0501863102.
    1. Benoit M, Barbarat B, Bernard A, Olive D, Mege JL. 2008. Coxiella burnetii, the agent of Q fever, stimulates an atypical M2 activation program in human macrophages. Eur J Immunol 38:1065–1070. doi:10.1002/eji.200738067.
    1. Capo C, Lindberg FP, Meconi S, Zaffran Y, Tardei G, Brown EJ, Raoult D, Mege JL. 1999. Subversion of monocyte functions by Coxiella burnetii: impairment of the cross-talk between alphavbeta3 integrin and CR3. J Immunol 163:6078–6085.
    1. Colombo MI, Gutierrez MG, Romano PS. 2006. The two faces of autophagy: Coxiella and Mycobacterium. Autophagy 2:162–164. doi:10.4161/auto.2827.
    1. Zhang G, Samuel JE. 2004. Vaccines against Coxiella infection. Expert Rev Vaccines 3:577–584. doi:10.1586/14760584.3.5.577.
    1. Zhang G, Russell-Lodrigue KE, Andoh M, Zhang Y, Hendrix LR, Samuel JE. 2007. Mechanisms of vaccine-induced protective immunity against Coxiella burnetii infection in BALB/c mice. J Immunol 179:8372–8380. doi:10.4049/jimmunol.179.12.8372.
    1. Andoh M, Zhang G, Russell-Lodrigue KE, Shive HR, Weeks BR, Samuel JE. 2007. T cells are essential for bacterial clearance, and gamma interferon, tumor necrosis factor alpha, and B cells are crucial for disease development in Coxiella burnetii infection in mice. Infect Immun 75:3245–3255. doi:10.1128/IAI.01767-06.
    1. Read AJ, Erickson S, Harmsen AG. 2010. Role of CD4+ and CD8+ T cells in clearance of primary pulmonary infection with Coxiella burnetii. Infect Immun 78:3019–3026. doi:10.1128/IAI.00101-10.
    1. Chen C, Dow C, Wang P, Sidney J, Read A, Harmsen A, Samuel JE, Peters B. 2011. Identification of CD4+ T cell epitopes in C. burnetii antigens targeted by antibody responses. PLoS One 6:e17712. doi:10.1371/journal.pone.0017712.
    1. Turco J, Thompson HA, Winkler HH. 1984. Interferon-gamma inhibits growth of Coxiella burnetii in mouse fibroblasts. Infect Immun 45:781–783. doi:10.1128/IAI.45.3.781-783.1984.
    1. Grieshaber S, Swanson JA, Hackstadt T. 2002. Determination of the physical environment within the Chlamydia trachomatis inclusion using ion-selective ratiometric probes. Cell Microbiol 4:273–283. doi:10.1046/j.1462-5822.2002.00191.x.
    1. Lampe MF, Rohan LC, Skinner MC, Stamm WE. 2004. Susceptibility of Chlamydia trachomatis to excipients commonly used in topical microbicide formulations. Antimicrob Agents Chemother 48:3200–3202. doi:10.1128/AAC.48.8.3200-3202.2004.
    1. Gong Z, Luna Y, Yu P, Fan H. 2014. Lactobacilli inactivate Chlamydia trachomatis through lactic acid but not H2O2. PLoS One 9:e107758. doi:10.1371/journal.pone.0107758.
    1. Mahmoud EA, Svensson LO, Olsson SE, Mardh PA. 1995. Antichlamydial activity of vaginal secretion. Am J Obstet Gynecol 172:1268–1272. doi:10.1016/0002-9378(95)91491-9.
    1. Yasin B, Pang M, Wagar EA, Lehrer RI. 2002. Examination of Chlamydia trachomatis infection in environments mimicking normal and abnormal vaginal pH. Sex Transm Dis 29:514–519. doi:10.1097/00007435-200209000-00004.
    1. Schramm N, Bagnell CR, Wyrick PB. 1996. Vesicles containing Chlamydia trachomatis serovar L2 remain above pH 6 within HEC-1B cells. Infect Immun 64:1208–1214. doi:10.1128/IAI.64.4.1208-1214.1996.
    1. Heinzen RA, Scidmore MA, Rockey DD, Hackstadt T. 1996. Differential interaction with endocytic and exocytic pathways distinguish parasitophorous vacuoles of Coxiella burnetii and Chlamydia trachomatis. Infect Immun 64:796–809. doi:10.1128/IAI.64.3.796-809.1996.
    1. Rzomp KA, Scholtes LD, Briggs BJ, Whittaker GR, Scidmore MA. 2003. Rab GTPases are recruited to chlamydial inclusions in both a species-dependent and species-independent manner. Infect Immun 71:5855–5870. doi:10.1128/iai.71.10.5855-5870.2003.
    1. Sun HS, Eng EW, Jeganathan S, Sin AT, Patel PC, Gracey E, Inman RD, Terebiznik MR, Harrison RE. 2012. Chlamydia trachomatis vacuole maturation in infected macrophages. J Leukoc Biol 92:815–827. doi:10.1189/jlb.0711336.
    1. Beron W, Gutierrez MG, Rabinovitch M, Colombo MI. 2002. Coxiella burnetii localizes in a Rab7-labeled compartment with autophagic characteristics. Infect Immun 70:5816–5821. doi:10.1128/iai.70.10.5816-5821.2002.
    1. Schulze-Luehrmann J, Eckart RA, Olke M, Saftig P, Liebler-Tenorio E, Lührmann A. 2016. LAMP proteins account for the maturation delay during the establishment of the Coxiella burnetii-containing vacuole. Cell Microbiol 18:181–194. doi:10.1111/cmi.12494.
    1. Newton HJ, Roy CR. 2011. The Coxiella burnetii Dot/Icm system creates a comfortable home through lysosomal renovation. mBio 2:e00226-11. doi:10.1128/mBio.00226-11.
    1. Lührmann A, Newton HJ, Bonazzi M. 2017. Beginning to understand the role of the type IV secretion system effector proteins in Coxiella burnetii pathogenesis. Curr Top Microbiol Immunol 413:243–268. doi:10.1007/978-3-319-75241-9_10.
    1. Howe D, Shannon JG, Winfree S, Dorward DW, Heinzen RA. 2010. Coxiella burnetii phase I and II variants replicate with similar kinetics in degradative phagolysosome-like compartments of human macrophages. Infect Immun 78:3465–3474. doi:10.1128/IAI.00406-10.
    1. Hackstadt T, Williams JC. 1981. Biochemical stratagem for obligate parasitism of eukaryotic cells by Coxiella burnetii. Proc Natl Acad Sci U S A 78:3240–3244. doi:10.1073/pnas.78.5.3240.
    1. Maurin M, Benoliel AM, Bongrand P, Raoult D. 1992. Phagolysosomes of Coxiella burnetii-infected cell lines maintain an acidic pH during persistent infection. Infect Immun 60:5013–5016. doi:10.1128/IAI.60.12.5013-5016.1992.
    1. Newton HJ, McDonough JA, Roy CR. 2013. Effector protein translocation by the Coxiella burnetii Dot/Icm type IV secretion system requires endocytic maturation of the pathogen-occupied vacuole. PLoS One 8:e54566. doi:10.1371/journal.pone.0054566.
    1. Mezouar S, Vitte J, Gorvel L, Ben Amara A, Desnues B, Mege JL. 2019. Mast cell cytonemes as a defense mechanism against Coxiella burnetii. mBio 10:e02669-18. doi:10.1128/mBio.02669-18.
    1. Elliott A, Peng Y, Zhang G. 2013. Coxiella burnetii interaction with neutrophils and macrophages in vitro and in SCID mice following aerosol infection. Infect Immun 81:4604–4614. doi:10.1128/IAI.00973-13.
    1. Baca OG, Akporiaye ET, Aragon AS, Martinez IL, Robles MV, Warner NL. 1981. Fate of phase I and phase II Coxiella burnetii in several macrophage-like tumor cell lines. Infect Immun 33:258–266. doi:10.1128/IAI.33.1.258-266.1981.
    1. Schoenlaub L, Elliott A, Freches D, Mitchell WJ, Zhang G. 2015. Role of B cells in host defense against primary Coxiella burnetii infection. Infect Immun 83:4826–4836. doi:10.1128/IAI.01073-15.
    1. Howe D, Melnicakova J, Barak I, Heinzen RA. 2003. Maturation of the Coxiella burnetii parasitophorous vacuole requires bacterial protein synthesis but not replication. Cell Microbiol 5:469–480. doi:10.1046/j.1462-5822.2003.00293.x.
    1. Clark SE, Filak HC, Guthrie BS, Schmidt RL, Jamieson A, Merkel P, Knight V, Cole CM, Raulet DH, Lenz LL. 2016. Bacterial manipulation of NK cell regulatory activity increases susceptibility to Listeria monocytogenes infection. PLoS Pathog 12:e1005708. doi:10.1371/journal.ppat.1005708.
    1. Tato CM, Martins GA, High FA, DiCioccio CB, Reiner SL, Hunter CA. 2004. Cutting Edge: innate production of IFN-gamma by NK cells is independent of epigenetic modification of the IFN-gamma promoter. J Immunol 173:1514–1517. doi:10.4049/jimmunol.173.3.1514.
    1. Rebbig A, Matthiesen S, Lührmann A, Knittler MR. 2018. Flow cytometry as a new complementary tool to study Coxiella burnetii in cell cultures. J Microbiol Methods 151:39–43. doi:10.1016/j.mimet.2018.05.022.
    1. Latomanski EA, Newton HJ. 2018. Interaction between autophagic vesicles and the Coxiella-containing vacuole requires CLTC (clathrin heavy chain). Autophagy 14:1710–1725. doi:10.1080/15548627.2018.1483806.
    1. Vodkin MH, Williams JC. 1988. A heat shock operon in Coxiella burnetii produces a major antigen homologous to a protein in both Mycobacteria and Escherichia coli. J Bacteriol 170:1227–1234. doi:10.1128/jb.170.3.1227-1234.1988.
    1. Elsa J, Duron O, Severine B, Gonzalez-Acuna D, Sidi-Boumedine K. 2015. Molecular methods routinely used to detect Coxiella burnetii in ticks cross-react with Coxiella-like bacteria. Infect Ecol Epidemiol 5:29230. doi:10.3402/iee.v5.29230.
    1. Evenou JP, Wagner J, Zenke G, Brinkmann V, Wagner K, Kovarik J, Welzenbach KA, Weitz-Schmidt G, Guntermann C, Towbin H, Cottens S, Kaminski S, Letschka T, Lutz-Nicoladoni C, Gruber T, Hermann-Kleiter N, Thuille N, Baier G. 2009. The potent protein kinase C-selective inhibitor AEB071 (sotrastaurin) represents a new class of immunosuppressive agents affecting early T-cell activation. J Pharmacol Exp Ther 330:792–801. doi:10.1124/jpet.109.153205.
    1. Krzewski K, Coligan JE. 2012. Human NK cell lytic granules and regulation of their exocytosis. Front Immunol 3:335. doi:10.3389/fimmu.2012.00335.
    1. Rak GD, Mace EM, Banerjee PP, Svitkina T, Orange JS. 2011. Natural killer cell lytic granule secretion occurs through a pervasive actin network at the immune synapse. PLoS Biol 9:e1001151. doi:10.1371/journal.pbio.1001151.
    1. Burkhardt JK, Hester S, Lapham CK, Argon Y. 1990. The lytic granules of natural killer cells are dual-function organelles combining secretory and pre-lysosomal compartments. J Cell Biol 111:2327–2340. doi:10.1083/jcb.111.6.2327.
    1. Samanta D, Gilk SD. 2017. Measuring pH of the Coxiella burnetii parasitophorous vacuole. Curr Protoc Microbiol 47:6C.3.1–6C.3.11. doi:10.1002/cpmc.38.
    1. Heinzen RA, Hackstadt T, Samuel JE. 1999. Developmental biology of Coxiella burnetii. Trends Microbiol 7:149–154. doi:10.1016/s0966-842x(99)01475-4.
    1. Gomes MS, Paul S, Moreira AL, Appelberg R, Rabinovitch M, Kaplan G. 1999. Survival of Mycobacterium avium and Mycobacterium tuberculosis in acidified vacuoles of murine macrophages. Infect Immun 67:3199–3206. doi:10.1128/IAI.67.7.3199-3206.1999.
    1. Veras PS, de Chastellier C, Moreau MF, Villiers V, Thibon M, Mattei D, Rabinovitch M. 1994. Fusion between large phagocytic vesicles: targeting of yeast and other particulates to phagolysosomes that shelter the bacterium Coxiella burnetii or the protozoan Leishmania amazonensis in Chinese hamster ovary cells. J Cell Sci 107:3065–3076.
    1. Veras PS, Moulia C, Dauguet C, Tunis CT, Thibon M, Rabinovitch M. 1995. Entry and survival of Leishmania amazonensis amastigotes within phagolysosome-like vacuoles that shelter Coxiella burnetii in Chinese hamster ovary cells. Infect Immun 63:3502–3506. doi:10.1128/IAI.63.9.3502-3506.1995.
    1. Coleman SA, Fischer ER, Howe D, Mead DJ, Heinzen RA. 2004. Temporal analysis of Coxiella burnetii morphological differentiation. J Bacteriol 186:7344–7352. doi:10.1128/JB.186.21.7344-7352.2004.
    1. Larson CL, Beare PA, Howe D, Heinzen RA. 2013. Coxiella burnetii effector protein subverts clathrin-mediated vesicular trafficking for pathogen vacuole biogenesis. Proc Natl Acad Sci U S A 110:E4770–E4779. doi:10.1073/pnas.1309195110.
    1. McCaul TF, Williams JC. 1981. Developmental cycle of Coxiella burnetii: structure and morphogenesis of vegetative and sporogenic differentiations. J Bacteriol 147:1063–1076. doi:10.1128/JB.147.3.1063-1076.1981.
    1. Ginsburg I, Neeman N, Duchan Z, Sela MN, James J, Lahav M. 1975. The effect of leukocyte hydrolases on bacteria: IV. The role played by artificial enzyme “cocktails” and tissue enzymes in bacteriolysis. Inflammation 1:41–56. doi:10.1007/BF00918058.
    1. Bangalore N, Travis J, Onunka VC, Pohl J, Shafer WM. 1990. Identification of the primary antimicrobial domains in human neutrophil cathepsin G. J Biol Chem 265:13584–13588.
    1. Amano K, Williams JC, McCaul TF, Peacock MG. 1984. Biochemical and immunological properties of Coxiella burnetii cell wall and peptidoglycan-protein complex fractions. J Bacteriol 160:982–988. doi:10.1128/JB.160.3.982-988.1984.
    1. Samanta D, Clemente TM, Schuler BE, Gilk SD. 2019. Coxiella burnetii type 4B secretion system-dependent manipulation of endolysosomal maturation is required for bacterial growth. PLoS Pathog 15:e1007855. doi:10.1371/journal.ppat.1007855.
    1. Di Russo Case E, Samuel JE. 2016. Contrasting lifestyles within the host cell. Microbiol Spectr 4:VMBF-0014-2015. doi:10.1128/microbiolspec.VMBF-0014-2015.
    1. Tassi I, Cella M, Presti R, Colucci A, Gilfillan S, Littman DR, Colonna M. 2008. NK cell-activating receptors require PKC-theta for sustained signaling, transcriptional activation, and IFN-gamma secretion. Blood 112:4109–4116. doi:10.1182/blood-2008-02-139527.
    1. Aguilo JI, Garaude J, Pardo J, Villalba M, Anel A. 2009. Protein kinase C-theta is required for NK cell activation and in vivo control of tumor progression. J Immunol 182:1972–1981. doi:10.4049/jimmunol.0801820.
    1. Bonnema JD, Rivlin KA, Ting AT, Schoon RA, Abraham RT, Leibson PJ. 1994. Cytokine-enhanced NK cell-mediated cytotoxicity. Positive modulatory effects of IL-2 and IL-12 on stimulus-dependent granule exocytosis. J Immunol 152:2098–2104.
    1. Vishwanath S, Hackstadt T. 1988. Lipopolysaccharide phase variation determines the complement-mediated serum susceptibility of Coxiella burnetii. Infect Immun 56:40–44. doi:10.1128/IAI.56.1.40-44.1988.
    1. Shannon JG, Howe D, Heinzen RA. 2005. Lack of dendritic cell maturation following infection by Coxiella burnetii synthesizing different lipopolysaccharide chemotypes. Ann N Y Acad Sci 1063:154–160. doi:10.1196/annals.1355.024.
    1. Islam A, Lockhart M, Stenos J, Graves S. 2013. The attenuated Nine Mile phase II clone 4/RSA439 strain of Coxiella burnetii is highly virulent for severe combined immunodeficient (SCID) mice. Am J Trop Med Hyg 89:800–803. doi:10.4269/ajtmh.12-0653.
    1. Kishimoto RA, Rozmiarek H, Larson EW. 1978. Experimental Q fever infection in congenitally athymic nude mice. Infect Immun 22:69–71. doi:10.1128/IAI.22.1.69-71.1978.
    1. Ammerdorffer A, Schoffelen T, Gresnigt MS, Oosting M, den Brok MH, Abdollahi-Roodsaz S, Kanneganti TD, de Jong DJ, van Deuren M, Roest HJ, Rebel JM, Netea MG, Joosten LA, Sprong T. 2015. Recognition of Coxiella burnetii by Toll-like receptors and nucleotide-binding oligomerization domain-like receptors. J Infect Dis 211:978–987. doi:10.1093/infdis/jiu526.
    1. Martinez J, Huang X, Yang Y. 2010. Direct TLR2 signaling is critical for NK cell activation and function in response to vaccinia viral infection. PLoS Pathog 6:e1000811. doi:10.1371/journal.ppat.1000811.
    1. Athie-Morales V, O’Connor GM, Gardiner CM. 2008. Activation of human NK cells by the bacterial pathogen-associated molecular pattern muramyl dipeptide. J Immunol 180:4082–4089. doi:10.4049/jimmunol.180.6.4082.
    1. Hackstadt T, Caldwell HD. 1985. Effect of proteolytic cleavage of surface-exposed proteins on infectivity of Chlamydia trachomatis. Infect Immun 48:546–551. doi:10.1128/IAI.48.2.546-551.1985.
    1. Gutiérrez-Martín CB, Ojcius DM, Hsia R, Hellio R, Bavoil PM, Dautry-Varsat A. 1997. Heparin-mediated inhibition of Chlamydia psittaci adherence to HeLa cells. Microb Pathog 22:47–57. doi:10.1006/mpat.1996.0090.
    1. Ting LM, Hsia RC, Haidaris CG, Bavoil PM. 1995. Interaction of outer envelope proteins of Chlamydia psittaci GPIC with the HeLa cell surface. Infect Immun 63:3600–3608. doi:10.1128/IAI.63.9.3600-3608.1995.
    1. Su H, Watkins NG, Zhang YZ, Caldwell HD. 1990. Chlamydia trachomatis-host cell interactions: role of the chlamydial major outer membrane protein as an adhesin. Infect Immun 58:1017–1025. doi:10.1128/IAI.58.4.1017-1025.1990.
    1. Buttrum L, Ledbetter L, Cherla R, Zhang Y, Mitchell WJ, Zhang G. 2018. Both major histocompatibility complex class I (MHC-I) and MHC-II molecules are required, while MHC-I appears to play a critical role in host defense against primary Coxiella burnetii infection. Infect Immun 86:e00602-17. doi:10.1128/IAI.00602-17.
    1. Thiery J, Keefe D, Saffarian S, Martinvalet D, Walch M, Boucrot E, Kirchhausen T, Lieberman J. 2010. Perforin activates clathrin- and dynamin-dependent endocytosis, which is required for plasma membrane repair and delivery of granzyme B for granzyme-mediated apoptosis. Blood 115:1582–1593. doi:10.1182/blood-2009-10-246116.
    1. Gorvel L, Textoris J, Banchereau R, Ben Amara A, Tantibhedhyangkul W, von Bargen K, Ka MB, Capo C, Ghigo E, Gorvel JP, Mege JL. 2014. Intracellular bacteria interfere with dendritic cell functions: role of the type I interferon pathway. PLoS One 9:e99420. doi:10.1371/journal.pone.0099420.
    1. Karlhofer FM, Orihuela MM, Yokoyama WM. 1995. Ly-49-independent natural killer (NK) cell specificity revealed by NK cell clones derived from p53-deficient mice. J Exp Med 181:1785–1795. doi:10.1084/jem.181.5.1785.
    1. Schafer W, Eckart RA, Schmid B, Cagkoylu H, Hof K, Muller YA, Amin B, Lührmann A. 2017. Nuclear trafficking of the anti-apoptotic Coxiella burnetii effector protein AnkG requires binding to p32 and Importin-alpha1. Cell Microbiol 19:e12634. doi:10.1111/cmi.12634.
    1. Gibson DG. 2011. Enzymatic assembly of overlapping DNA fragments. Methods Enzymol 498:349–361. doi:10.1016/B978-0-12-385120-8.00015-2.
    1. Omsland A, Beare PA, Hill J, Cockrell DC, Howe D, Hansen B, Samuel JE, Heinzen RA. 2011. Isolation from animal tissue and genetic transformation of Coxiella burnetii are facilitated by an improved axenic growth medium. Appl Environ Microbiol 77:3720–3725. doi:10.1128/AEM.02826-10.
    1. Omsland A, Cockrell DC, Howe D, Fischer ER, Virtaneva K, Sturdevant DE, Porcella SF, Heinzen RA. 2009. Host cell-free growth of the Q fever bacterium Coxiella burnetii. Proc Natl Acad Sci U S A 106:4430–4434. doi:10.1073/pnas.0812074106.
    1. Goellner S, Schubert E, Liebler-Tenorio E, Hotzel H, Saluz HP, Sachse K. 2006. Transcriptional response patterns of Chlamydophila psittaci in different in vitro models of persistent infection. Infect Immun 74:4801–4808. doi:10.1128/IAI.01487-05.
    1. Schachter J, Wyrick PB. 1994. Culture and isolation of Chlamydia trachomatis. Methods Enzymol 236:377–390. doi:10.1016/0076-6879(94)36028-6.
    1. Fiegl D, Kägebein D, Liebler-Tenorio EM, Weisser T, Sens M, Gutjahr M, Knittler MR. 2013. Amphisomal route of MHC class I cross-presentation in bacteria-infected dendritic cells. J Immunol 190:2791–2806. doi:10.4049/jimmunol.1202741.
    1. Schindelin J, Arganda-Carreras I, Frise E, Kaynig V, Longair M, Pietzsch T, Preibisch S, Rueden C, Saalfeld S, Schmid B, Tinevez JY, White DJ, Hartenstein V, Eliceiri K, Tomancak P, Cardona A. 2012. Fiji: an open-source platform for biological-image analysis. Nat Methods 9:676–682. doi:10.1038/nmeth.2019.

Source: PubMed

3
Abonneren