Alveolar Macrophages Treated With Bacillus subtilis Spore Protect Mice Infected With Respiratory Syncytial Virus A2

Ji Eun Hong, Yoon-Chul Kye, Sung-Moo Park, In Su Cheon, Hyuk Chu, Byung-Chul Park, Yeong-Min Park, Jun Chang, Jae-Ho Cho, Man Ki Song, Seung Hyun Han, Cheol-Heui Yun, Ji Eun Hong, Yoon-Chul Kye, Sung-Moo Park, In Su Cheon, Hyuk Chu, Byung-Chul Park, Yeong-Min Park, Jun Chang, Jae-Ho Cho, Man Ki Song, Seung Hyun Han, Cheol-Heui Yun

Abstract

Respiratory syncytial virus (RSV) is a major pathogen that infects lower respiratory tract and causes a common respiratory disease. Despite serious pathological consequences with this virus, effective treatments for controlling RSV infection remain unsolved, along with poor innate immune responses induced at the initial stage of RSV infection. Such a poor innate defense mechanism against RSV leads us to study the role of alveolar macrophage (AM) that is one of the primary innate immune cell types in the respiratory tract and may contribute to protective responses against RSV infection. As an effective strategy for enhancing anti-viral function of AM, this study suggests the intranasal administration of Bacillus subtilis spore which induces expansion of AM in the lung with activation and enhanced production of inflammatory cytokines along with several genes associated with M1 macrophage differentiation. Such effect by spore on AM was largely dependent on TLR-MyD88 signaling and, most importantly, resulted in a profound reduction of viral titers and pathological lung injury upon RSV infection. Taken together, our results suggest a protective role of AM in RSV infection and its functional modulation by B. subtilis spore, which may be a useful and potential therapeutic approach against RSV.

Keywords: Bacillus subtilis; MyD88; alveolar macrophage activation; respiratory syncytial virus; spore.

Figures

FIGURE 1
FIGURE 1
Pre-treatment with spore through intranasal route reduces the disease severity following RSV infection and induces the population change of alveolar macrophages (AM) and enhances antiviral effector molecules. Mice were administered with 1 × 109 CFU of spore via i.n. route at 5 days prior to RSV infection with 2 × 106 PFU per mouse (n = 3). (A) Body weight was monitored daily after the infection and (B) viral load in the lungs was analyzed by plaque assay at 4 days post-infection (DPI). Change of various innate immune cells in the (C) post-lavaged lung and (D) BAL fluid was analyzed by flow cytometry at 0 to 4 DPI. Empty and filled circles indicate PBS and spore pre-treated mice, respectively. (E) IFN-β and IL-12p40 in BAL fluid were measured by ELISA. Data are expressed as mean ± S.E.M. for the group. Significant differences from results with the PBS control are ∗P < 0.05; ∗∗P < 0.01; and ∗∗∗P < 0.001, respectively.
FIGURE 2
FIGURE 2
Mice selectively depleted with AMs fail to protect RSV infection. Mice were injected i.t. with clodronate-encapsulated liposome twice on days 1 and 3 prior to RSV infection. (A) Body weight was monitored daily after the infection and (B) viral load in the lungs was analyzed by plaque assay at 4 DPI (n = 3). At 4 DPI, perfused lungs were stained with H&E (C) for histological examination by microscopy at 200 × magnifications and (D–G) scored for histopathology. “Cont” indicates the mice injected with control liposome and “Clod” indicates the mice injected with clodronate-encapsulated liposome. Arrows indicated as follows; orange, epithelium thickness and destruction; green, pulmonary edema; red, inflammatory cells; and black, cell death. Data are expressed as mean ± S.E.M. for the group. Significant differences from results with the PBS control are ∗P < 0.05 and ∗∗P < 0.01, respectively.
FIGURE 3
FIGURE 3
Alveolar macrophages pre-treated with spore extend their antiviral effects. MH-S cells were stimulated with spore for 24 h, with a ratio of MH-S:Spore at = 1:0, 1:1, 1:10, and 1:50. Then, the cells were washed and infected with 1 MOI RSV for 12 h. (A) The viral load was analyzed by using plaque assay and (B) the production of IL-12p40 and IL-6 was measured by ELISA. Data are expressed as mean ± S.E.M. of three replicates. ∗ and ∗∗ indicate significant differences at P < 0.05 and P < 0.01, respectively.
FIGURE 4
FIGURE 4
Alveolar macrophages in mice intranasally administered with spore play a key role in RSV infection. Mice were intranasally administered with spore at 5 days prior to RSV infection and injected with control or clodronate-encapsulated liposome through intratracheal on days 3 and 1 before the infection and sacrifice on 4 DPI. (A) Body weight was monitored daily after the infection and (B) viral load in the lung was examined at 4 DPI, respectively (n = 3). (C) At 4 DPI, perfused lungs were stained with H&E for histological examination by microscopy at 200 × magnifications and (D–G) scored for histopathology. Arrows indicated are as follows; orange, epithelium thickness and destruction; green, pulmonary edema; red, inflammatory cells; and black, cell death. Significant differences from results with the PBS control are ∗P < 0.05; ∗∗P < 0.01; and ∗∗∗P < 0.001, respectively.
FIGURE 5
FIGURE 5
Protective mechanism of spore pre-treated mice infected with RSV is dependent on MyD88 signaling in AMs. Wild type or MyD88 knockout mice were administered i.n. with spore at 5 days prior to RSV infection. (A) Body weight was monitored daily after the infection (n = 3). At 4 DPI, (B) viral load in the lungs was analyzed by plaque assay and absolute number of AMs in (C) post lavaged lungs, and (D) BAL cells were acquired by flow cytometry. Values with different letters (a, b, c, d) are significantly different one from another (P < 0.05). (E) Blood-perfused lungs were stained with H&E for histological examination by microscopy at 200× magnification and (F–I) scored for histopathology at 4 DPI. Arrows indicated are as follow; orange, epithelium thickness and destruction; green, pulmonary edema; red, inflammatory cells; and black, cell death. Significant differences from results with the PBS control are ∗P < 0.05; ∗∗P < 0.01; and ∗∗∗P < 0.001, respectively.
FIGURE 6
FIGURE 6
Bone marrow-derived macrophages pre-treated with spore develop antiviral effects through MyD88-dependent pathway. Wild type or MyD88 knockout BMMs were treated with spore for 24 h at ratio of spore per number of cells for 0, 1, 10, and 50. Then, the cells were washed and infected with 1 MOI virus for additional 12 h. (A) Viral titers from wild type or MyD88 knockout BMMs were measured by plaque assay and (B) the production of IL-12p40 was measured by ELISA. Data are expressed as mean ± S.E.M. for 3 independent experiments. ∗, ∗∗, and ∗∗∗ indicate significant differences at P < 0.05, P < 0.01, and P < 0.001, respectively.

References

    1. Barros M. H., Hauck F., Dreyer J. H., Kempkes B., Niedobitek G. (2013). Macrophage polarisation: an immunohistochemical approach for identifying M1 and M2 macrophages. PLoS One 8:e80908. 10.1371/journal.pone.0080908
    1. Colenutt C., Cutting S. M. (2014). Use of Bacillus subtilis PXN21 spores for suppression of Clostridium difficile infection symptoms in a murine model. FEMS Microbiol. Lett. 358 154–161. 10.1111/1574-6968.12468
    1. Copenhaver A. M., Casson C. N., Nguyen H. T., Fung T. C., Duda M. M., Roy C. R., et al. (2014). Alveolar macrophages and neutrophils are the primary reservoirs for Legionella pneumophila and mediate cytosolic surveillance of type IV secretion. Infect. Immun. 82 4325–4336. 10.1128/IAI.01891-14
    1. Garcia-Crespo K. E., Chan C. C., Gabryszewski S. J., Percopo C. M., Rigaux P., Dyer K. D., et al. (2013). Lactobacillus priming of the respiratory tract: heterologous immunity and protection against lethal pneumovirus infection. Antiv. Res. 97 270–279. 10.1016/j.antiviral.2012.12.022
    1. Geissmann F., Manz M. G., Jung S., Sieweke M. H., Merad M., Ley K. (2010). Development of monocytes, macrophages, and dendritic cells. Science 327 656–661. 10.1126/science.1178331
    1. Ghoneim H. E., Thomas P. G., Mccullers J. A. (2013). Depletion of alveolar macrophages during influenza infection facilitates bacterial superinfections. J. Immunol. 191 1250–1259. 10.4049/jimmunol.1300014
    1. Ginhoux F. (2014). Fate PPAR-titioning: PPAR-gamma ’instructs’ alveolar macrophage development. Nat. Immunol. 15 1005–1007. 10.1038/ni.3011
    1. Gomez M. I., Prince A. (2008). Airway epithelial cell signaling in response to bacterial pathogens. Pediatr. Pulmonol. 43 11–19. 10.1002/ppul.20735
    1. Goritzka M., Makris S., Kausar F., Durant L. R., Pereira C., Kumagai Y., et al. (2015). Alveolar macrophage-derived type I interferons orchestrate innate immunity to RSV through recruitment of antiviral monocytes. J. Exp. Med. 212 699–714. 10.1084/jem.20140825
    1. Guilliams M., De Kleer I., Henri S., Post S., Vanhoutte L., De Prijck S., et al. (2013). Alveolar macrophages develop from fetal monocytes that differentiate into long-lived cells in the first week of life via GM-CSF. J. Exp. Med. 210 1977–1992. 10.1084/jem.20131199
    1. Hashimoto D., Miller J., Merad M. (2011). Dendritic cell and macrophage heterogeneity in vivo. Immunity 35 323–335. 10.1016/j.immuni.2011.09.007
    1. Herold S., Mayer K., Lohmeyer J. (2011). Acute lung injury: how macrophages orchestrate resolution of inflammation and tissue repair. Front. Immunol. 2:65. 10.3389/fimmu.2011.00065
    1. Hong H. A., Huang J. M., Khaneja R., Hiep L. V., Urdaci M. C., Cutting S. M. (2008). The safety of Bacillus subtilis and Bacillus indicus as food probiotics. J. Appl. Microbiol. 105 510–520. 10.1111/j.1365-2672.2008.03773.x
    1. Jang J. E., Lee J. B., Kim K. H., Park S. M., Shim B. S., Cheon I. S., et al. (2011). Evaluation of protective efficacy of respiratory syncytial virus vaccine against A and B subgroup human isolates in Korea. PLoS One 6:e23797. 10.1371/journal.pone.0023797
    1. Kiso M., Takano R., Sakabe S., Katsura H., Shinya K., Uraki R., et al. (2013). Protective efficacy of orally administered, heat-killed Lactobacillus pentosus b240 against influenza A virus. Sci. Rep. 3:1563. 10.1038/srep01563
    1. Kolli D., Gupta M. R., Sbrana E., Velayutham T. S., Chao H., Casola A., et al. (2014). Alveolar macrophages contribute to the pathogenesis of human metapneumovirus infection while protecting against respiratory syncytial virus infection. Am. J. Respir Cell Mol. Biol. 51 502–515. 10.1165/rcmb.2013-0414OC
    1. Kumagai Y., Takeuchi O., Kato H., Kumar H., Matsui K., Morii E., et al. (2007). Alveolar macrophages are the primary interferon-alpha producer in pulmonary infection with RNA viruses. Immunity 27 240–252. 10.1016/j.immuni.2007.07.013
    1. Kurt-Jones E. A., Popova L., Kwinn L., Haynes L. M., Jones L. P., Tripp R. A., et al. (2000). Pattern recognition receptors TLR4 and CD14 mediate response to respiratory syncytial virus. Nat. Immunol. 1 398–401. 10.1038/80833
    1. Lee I. K., Kye Y. C., Kim G., Kim H. W., Gu M. J., Umboh J., et al. (2016). Stress, nutrition, and intestinal immune responses in pigs – A review. Asian-Australas J. Anim. Sci. 29 1075–1082. 10.5713/ajas.16.0118
    1. Lee Y. N., Youn H. N., Kwon J. H., Lee D. H., Park J. K., Yuk S. S., et al. (2013). Sublingual administration of Lactobacillus rhamnosus affects respiratory immune responses and facilitates protection against influenza virus infection in mice. Antiv. Res. 98 284–290. 10.1016/j.antiviral.2013.03.013
    1. Lefevre M., Racedo S. M., Denayrolles M., Ripert G., Desfougeres T., Lobach A. R., et al. (2017). Safety assessment of Bacillus subtilis CU1 for use as a probiotic in humans. Regul. Toxicol. Pharmacol. 83 54–65. 10.1016/j.yrtph.2016.11.010
    1. Lefevre M., Racedo S. M., Ripert G., Housez B., Cazaubiel M., Maudet C., et al. (2015). Probiotic strain Bacillus subtilis CU1 stimulates immune system of elderly during common infectious disease period: a randomized, double-blind placebo-controlled study. Immun. Ageing 12:24. 10.1186/s12979-015-0051-y
    1. Marr N., Turvey S. E., Grandvaux N. (2013). Pathogen recognition receptor crosstalk in respiratory syncytial virus sensing: a host and cell type perspective. Trends Microbiol. 21 568–574. 10.1016/j.tim.2013.08.006
    1. Martinez F. O., Gordon S. (2014). The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep. 6:13. 10.12703/P6-13
    1. Maus U. A., Koay M. A., Delbeck T., Mack M., Ermert M., Ermert L., et al. (2002). Role of resident alveolar macrophages in leukocyte traffic into the alveolar air space of intact mice. Am. J. Physiol. Lung Cell Mol. Physiol. 282 L1245–L1252. 10.1152/ajplung.00453.2001
    1. McGill J., Heusel J. W., Legge K. L. (2009). Innate immune control and regulation of influenza virus infections. J. Leukoc. Biol. 86 803–812. 10.1189/jlb.0509368
    1. McKenney P. T., Driks A., Eichenberger P. (2013). The Bacillus subtilis endospore: assembly and functions of the multilayered coat. Nat. Rev. Microbiol. 11 33–44. 10.1038/nrmicro2921
    1. Meng J., Stobart C. C., Hotard A. L., Moore M. L. (2014). An overview of respiratory syncytial virus. PLoS Pathog. 10:e1004016. 10.1371/journal.ppat.1004016
    1. Mgbemena V., Segovia J., Chang T. H., Bose S. (2013). KLF6 and iNOS regulates apoptosis during respiratory syncytial virus infection. Cell Immunol. 283 1–7. 10.1016/j.cellimm.2013.06.002
    1. Murawski M. R., Bowen G. N., Cerny A. M., Anderson L. J., Haynes L. M., Tripp R. A., et al. (2009). Respiratory syncytial virus activates innate immunity through Toll-like receptor 2. J. Virol. 83 1492–1500. 10.1128/JVI.00671-08
    1. Murray P. J., Allen J. E., Biswas S. K., Fisher E. A., Gilroy D. W., Goerdt S., et al. (2014). Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity 41 14–20. 10.1016/j.immuni.2014.06.008
    1. Nair H., Nokes D. J., Gessner B. D., Dherani M., Madhi S. A., Singleton R. J., et al. (2010). Global burden of acute lower respiratory infections due to respiratory syncytial virus in young children: a systematic review and meta-analysis. Lancet 375 1545–1555. 10.1016/S0140-6736(10)60206-1
    1. Park M. K., Ngo V., Kwon Y. M., Lee Y. T., Yoo S., Cho Y. H., et al. (2013). Lactobacillus plantarum DK119 as a probiotic confers protection against influenza virus by modulating innate immunity. PLoS One 8:e75368. 10.1371/journal.pone.0075368
    1. Pribul P. K., Harker J., Wang B., Wang H., Tregoning J. S., Schwarze J., et al. (2008). Alveolar macrophages are a major determinant of early responses to viral lung infection but do not influence subsequent disease development. J. Virol. 82 4441–4448. 10.1128/JVI.02541-07
    1. Ragupathy S., Esmaeili F., Paschoud S., Sublet E., Citi S., Borchard G. (2014). Toll-like receptor 2 regulates the barrier function of human bronchial epithelial monolayers through atypical protein kinase C zeta, and an increase in expression of claudin-1. Tissue Barriers 2 e29166. 10.4161/tisb.29166
    1. Schneider C., Nobs S. P., Heer A. K., Kurrer M., Klinke G., Van Rooijen N., et al. (2014a). Alveolar macrophages are essential for protection from respiratory failure and associated morbidity following influenza virus infection. PLoS Pathog. 10:e1004053. 10.1371/journal.ppat.1004053
    1. Schneider C., Nobs S. P., Kurrer M., Rehrauer H., Thiele C., Kopf M. (2014b). Induction of the nuclear receptor PPAR-gamma by the cytokine GM-CSF is critical for the differentiation of fetal monocytes into alveolar macrophages. Nat. Immunol. 15 1026–1037. 10.1038/ni.3005
    1. Seo S. U., Kwon H. J., Ko H. J., Byun Y. H., Seong B. L., Uematsu S., et al. (2011). Type I interferon signaling regulates Ly6C(hi) monocytes and neutrophils during acute viral pneumonia in mice. PLoS Pathog. 7:e1001304. 10.1371/journal.ppat.1001304
    1. Shi T., Mcallister D. A., O’brien K. L., Simoes E. A. F., Madhi S. A., Gessner B. D., et al. (2017). Global, regional, and national disease burden estimates of acute lower respiratory infections due to respiratory syncytial virus in young children in 2015: a systematic review and modelling study. Lancet 390 946–958. 10.1016/S0140-6736(17)30938-8
    1. Shibata Y., Berclaz P. Y., Chroneos Z. C., Yoshida M., Whitsett J. A., Trapnell B. C. (2001). GM-CSF regulates alveolar macrophage differentiation and innate immunity in the lung through PU.1. Immunity 15 557–567. 10.1016/S1074-7613(01)00218-7
    1. Shim Y. R., Lee H. K. (2015). Caspase-1 independent viral clearance and adaptive immunity against mucosal respiratory syncytial virus infection. Immune Netw. 15 73–82. 10.4110/in.2015.15.2.73
    1. Shirey K. A., Pletneva L. M., Puche A. C., Keegan A. D., Prince G. A., Blanco J. C., et al. (2010). Control of RSV-induced lung injury by alternatively activated macrophages is IL-4R alpha-TLR4-, and IFN-beta-dependent. Mucosal Immunol. 3 291–300. 10.1038/mi.2010.6
    1. Snelgrove R. J., Goulding J., Didierlaurent A. M., Lyonga D., Vekaria S., Edwards L., et al. (2008). A critical function for CD200 in lung immune homeostasis and the severity of influenza infection. Nat. Immunol. 9 1074–1083. 10.1038/ni.1637
    1. Song M., Hong H. A., Huang J. M., Colenutt C., Khang D. D., Nguyen T. V., et al. (2012). Killed Bacillus subtilis spores as a mucosal adjuvant for an H5N1 vaccine. Vaccine 30 3266–3277. 10.1016/j.vaccine.2012.03.016
    1. Steinwede K., Tempelhof O., Bolte K., Maus R., Bohling J., Ueberberg B., et al. (2011). Local delivery of GM-CSF protects mice from lethal pneumococcal pneumonia. J. Immunol. 187 5346–5356. 10.4049/jimmunol.1101413
    1. Tan I. S., Ramamurthi K. S. (2014). Spore formation in Bacillus subtilis. Environ. Microbiol. Rep. 6 212–225. 10.1111/1758-2229.12130
    1. Tate M. D., Pickett D. L., Van Rooijen N., Brooks A. G., Reading P. C. (2010). Critical role of airway macrophages in modulating disease severity during influenza virus infection of mice. J. Virol. 84 7569–7580. 10.1128/JVI.00291-10
    1. Ugonna K., Bingle C. D., Plant K., Wilson K., Everard M. L. (2014). Macrophages are required for dendritic cell uptake of respiratory syncytial virus from an infected epithelium. PLoS One 9:e91855. 10.1371/journal.pone.0091855
    1. Yaqoob P. (2014). Ageing, immunity and influenza: a role for probiotics? Proc. Nutr. Soc. 73 309–317. 10.1017/S0029665113003777
    1. Zhang X., Goncalves R., Mosser D.M. (2008). The isolation and characterization of murine macrophages. Curr. Protoc. Immunol. 14:Unit 14.11.

Source: PubMed

Подписаться