Abstract
We reported previously that priming of the respiratory tract with immunobiotic Lactobacillus prior to virus challenge protects mice against subsequent lethal infection with pneumonia virus of mice (PVM). We present here the results of gene microarray which document differential expression of proinflammatory mediators in response to PVM infection alone and those suppressed in response to Lactobacillus plantarum. We also demonstrate for the first time that intranasal inoculation with live or heat-inactivated L. plantarum or Lactobacillus reuteri promotes full survival from PVM infection when administered within 24 h after virus challenge. Survival in response to L. plantarum administered after virus challenge is associated with suppression of proinflammatory cytokines, limited virus recovery, and diminished neutrophil recruitment to lung tissue and airways. Utilizing this post-virus challenge protocol, we found that protective responses elicited by L. plantarum at the respiratory tract were distinct from those at the gastrointestinal mucosa, as mice devoid of the anti-inflammatory cytokine, interleukin (IL)-10, exhibit survival and inflammatory responses that are indistinguishable from those of their wild-type counterparts. Finally, although L. plantarum interacts specifically with pattern recognition receptors TLR2 and NOD2, the respective gene-deleted mice were fully protected against lethal PVM infection by L. plantarum, as are mice devoid of type I interferon receptors. Taken together, L. plantarum is a versatile and flexible agent that is capable of averting the lethal sequelae of severe respiratory infection both prior to and post-virus challenge via complex and potentially redundant mechanisms.
Keywords: Inflammation, Cytokines, Pneumovirus, Pattern-recognition receptors
1. Introduction
Inflammation in response to severe respiratory virus infection can be complex and difficult to manage (Rosenberg and Domachowske, 2012). While antivirals are available for the treatment of specific respiratory viruses, they offer little benefit once symptoms become severe (Chan-Tack et al., 2015) suggesting that virus-induced inflammation contributes broadly to disease pathogenesis. Previous studies of the innate host response to viral lung infections have identified biomarkers of severe disease, high-lighting key pathways for the study of anti-inflammatory and/or immunomodulatory intervention (Schwarze and Mackenzie, 2013; Tabarani et al., 2013; Welliver, 2008).
Toward this end, there is currently significant interest in the immunomodulatory properties of probiotic bacteria (reviewed in Bron et al., 2011; Lebeer et al., 2010). While most of the literature on this subject features bacteria of the genera Lactobacillus and Bifidobacteria and their beneficial actions at the gastrointestinal mucosa, our group has focused on the impact of both live and heat-inactivated preparations of Lactobacillus species administered directly to the respiratory tract. Specifically, we have reported that priming of the respiratory tract of inbred strains of mice with live or heat-inactivated Lactobacillus plantarum results in robust and sustained protection against a subsequent lethal respiratory virus infection in association with profound inflammatory suppression (Gabryszewski et al., 2011; Garcia-Crespo et al., 2013; Percopo et al., 2014a). This set of observations is a unique example of heterologous immunity (also known in other contexts as trained immunity, innate imprinting, and innate memory), which are concepts that explain how the innate immune system alters its responsiveness and offers cross-protection against unrelated pathogens after a primary inflammatory or infectious event (reviewed in Levy and Netea, 2014; Netea et al., 2011; Wissinger et al., 2009; Locati et al., 2013; Ifrim et al., 2014). Using the defined pneumonia virus of mice model of severe pneumovirus infection (PVM; Family Paramyxoviridae, genus Pneumovirus; reviewed in Dyer et al., 2012; Bem et al., 2011), we have reported that Lactobacillus species introduced to the lung are cleared rapidly, and do not colonize the respiratory tract (Garcia-Crespo et al., 2013). In contrast to live or inactivated bacteria, Lactobacillus genomic DNA introduced alone cannot elicit protection against lethal virus infection (Garcia-Crespo et al., 2013). Furthermore, neither B cells nor antibodies are crucial elements of Lactobacillus-mediated protection against the lethal sequelae of this infection (Percopo et al., 2014a). Orally-delivered L. plantarum also has no impact in this setting (Percopo et al., 2014a), a finding that is consistent with the current clinical literature on the role of oral supplementation and its impact on respiratory virus infection (Hao et al., 2011; Esposito et al., 2014; Ozen et al., 2015).
In this manuscript, we demonstrate for the first time that intranasal inoculation with live or inactivated L. plantarum also promotes full survival from acute PVM infection when administered within 24 h after virus challenge, a finding that is also associated with suppression of virus replication and diminished expression of virus-induced proinflammatory cytokines. We have utilized this extended and clinically important means of eliciting protection to explore features of Lactobacillus-mediated protection at the respiratory tract that are unique and distinct from those taking place at gastrointestinal mucosa.
2. Materials and methods
2.1. Mice
Wild-type BALB/c and C57BL/6 mice were obtained from Division of Cancer Therapeutics and Charles River Laboratories, Frederick, MD. Interleukin-10 gene-deleted (IL-10−/−) and Type I interferon receptor gene-deleted (IFNαβR−/−) mice are maintained by the NIAID/Taconic consortium. Toll-like receptor 2 gene-deleted mice (#004650) and nucleotide-binding oligomerization domain-containing protein 2 gene-deleted mice (#005763) were obtained from The Jackson Laboratory. All mouse studies were approved by NIAID and carried out in accordance with NIAID ACUC Guidelines.
2.2. Virus
Mouse passaged stocks of PVM strain J3666 were prepared and stored in liquid nitrogen as previously described (Domachowske et al., 2000). Mice were inoculated intranasally under isoflurane anaesthesia with 50 μL of a 1:10,000 dilution (0.2 TCID50 units for mice on a BALB/c background) or a 1:1000 dilution (2.0 TCID50 units for mice on a C57BL/6 background) in Iscove’s Modified Dulbecco’s medium (IMDM) at time points indicated. Virus titer was determined from cDNA generated from total RNA from mouse lung tissue via a dual standard curve qRT-PCR method targeting the PVM small hydrophobic (SH) gene and mouse GAPDH; this assay generates absolute copy numbers per copy GAPDH (PVMSH/GAPDH) as previously described (Percopo et al., 2014b). Influenza A/HK/68 (H3N2) was passaged in BALB/c mice and stored at 104-fold concentrated stocks at −80 °C.
2.3. Lactobacillus
Cultures of L. plantarum NCIMB 8826 (ATCC BAA-793) or Lactobacillus reuteri F275 (ATCC 23272) were grown overnight in MRS broth at 37 °C. For experiments with live bacteria, cells were washed in sterile phosphate buffered saline (pbs) and re-suspended at 2 × 109 or 2 × 1010 colony forming units (cfu)/mL in sterile pbs with 0.1% bovine serum albumin (bsa) for intranasal inoculation under isoflurane anesthesia using the OD600 vs. cfu/mL determinations reported previously (Gabryszewski et al., 2011). Each mouse received 50 μL of this dilution (=108 or 109 cells or cfu) or 50 μL diluent control (pbs/bsa) per inoculation. In other experiments, L. plantarum or L. reuteri overnight cultures were washed with pbs and heat-inactivated by multiple cycles of freeze/thaw (Gabryszewski et al., 2011) or by heating to 70 °C for 30 min prior to re-suspension at 1011 cells/mL. Concentrated stocks of inactivated L. plantarum and L. reuteri were stored at −80 °C or −20 °C prior to dilution (in pbs/bsa) for inoculation as indicated.
2.4. Cytokine analysis
Cytokine ELISAs (R&D Systems) were performed on clarified homogenates of lung tissue and corrected for total protein by BCA assay (Pierce) as previously described (Gabryszewski et al., 2011).
2.5. Bronchoalveolar lavage (BAL) and cell counts
Cytospin slides (one slide per mouse) were prepared from BAL fluid (1.5 mL in pbs/bsa), fixed and stained with Diff-Quik. Two-hundred (200) to 300 cells were counted per slide.
2.6. Histology
Tissue sections prepared from 10% phosphate-buffered formalin-fixed lung tissue were stained with hematoxylin and eosin (H&E; Histoserv, Germantown, MD).
2.7. Flow cytometry
Lung tissue was harvested and single-cell suspensions were prepared as previously described (Garcia-Crespo et al., 2013; Percopo et al., 2014a). Live/Dead stain (Invitrogen) was added to the cells and Ab binding to Fc receptors was blocked with anti-mouse CD16/CD32. For analysis of T and B cells, lung suspensions were stained with anti-CD45-eF450 (eBioscience), anti-CD3-AF700 (eBioscience), anti-CD4-FITC (eBioscience), anti-CD8a-PE (BD), and anti-CD19-AF647 (BD) in PBS with 0.1% BSA at 4 °C for 30 min and washed with this buffer prior to analysis. For myeloid cell analysis, cells were stained with anti-CD45-AF700 (BD), anti-CD11c-AF488 (BD), anti-SiglecF-PE (BD), anti-GR1-V450 (BD) and anti-MHCII-APC (eBioscience). Natural killer cells were characterized by staining with anti-CD45-eF450 (eBioscience), anti-CD3-AF700 (BD), and anti-CD49/DX5-PE (BD). A minimum of 100,000 events were collected on an LSR II flow cytometer (BD Biosciences) and findings were analyzed in FlowJo 9.2.
2.8. DNA microarray target preparation and analysis
Eight-week old female BALB/c mice (all born on same day and shipped at same time from provider) were inoculated under isoflurane anesthesia with live L. plantarum (50 μL of 2 × 1010 cfu/mL in pbs/bsa) or diluent control on day-14 and again on day-7 and then inoculated with 0.2 TCID50 units (1:10,000 dilution in 50 μL) of PVM on day 14 or vehicle control. Each step of the study, including all mouse inoculations, RNA harvests to DNA microarray target preparation was designed and performed in a manner so as to avoid batch processing effects in the data due to mouse and sample type. Mouse inoculations, tissue harvest, RNA extraction, DNA target preparation batches were balanced between treatment and time. Lung tissues were harvested on days 17, 18, 19 and 20 and were snap frozen in liquid nitrogen. Samples (total 24, 6 mice per group) from mice that received two inoculations of L. plantarum or two inoculations of pbs/bsa diluent prior to virus or vehicle only challenge and harvested on day 19 were processed further for DNA microarray analysis. RNA extraction and target preparation were performed as described (Mackey-Lawrence et al., 2013) for all samples except RNA was extracted using RNeasy 96 well kit (Qiagen, Valencia, CA). Hybridization, fluidics and scanning were performed according to standard Affymetrix protocols (http://www.affymetrix.com) with the whole mouse genome 430 2.0 chip within the Genomics Unit of the Research Technologies Section (NIAID). Command Console (CC v3.1, http://www.Affymetrix.com) software was used to convert the image files to cell intensity data (cel files). All cel files, representing individual samples, were normalized by using the trimmed mean scaling method within expression console (EC v1.2, http://www.Affymetrix.com) to produce the analyzed cel files (chp files) along with the report files. The cel files were input into Partek Genomics Suite software (Partek, Inc. St. Louis, Mo., v6.6-6.12.0907) and quantile-normalized to produce the principal components analysis (PCA) graph and dendrogram. An ANOVA was performed within Partek to obtain multiple test corrected p-values using the false discovery rate method (FDR; (Klipper-Aurbach et al., 1995)) at the 0.05 significance level which were combined with fold change values for each comparison of interest. The array data presented in this publication have been deposited in NCBI’s Gene Expression Omnibus (Edgar et al., 2002) and are accessible through GEO series accession number GSE66721.
2.9. PRR activation assay
Activation of Toll-like receptors (TLRs 2, 3, 4, 5, 7, 8 and 9), NOD-like receptors (NOD 1 and NOD 2), and C-type lectin receptors (CLRs dectin 1a and 1b) were evaluated via a commercial assay in which NF-κB response via a single PRR expressed in HEK293 cells is measured via linked secretory alkaline phosphatase (SEAP; Invivogen). Upon introduction of 20 μL heat-inactivated L. plantarum (108 cells/mL), diluent (negative) control, or TLR/NOD/CLR ligand (positive control) to 200 μL of HEK293 cells (50–75 × 103 in 96 well plate), activation was measured via production of secretory alkaline phosphatase from a reporter gene under control of a promoter element inducible by NF-κB. Alkaline phosphatase produced is measured via detection of colorimetric products at OD = 650 nm. Positive ligands included: TLR2, heat-inactivated Listeria monocytogenes 108 cells/mL; TLR3, poly I:C 1 μg/mL; TLR4, Escherichia coli LPS 1 μg/mL; TLR5, Salmonella typhimurium flagellin 1 μg/mL; TLR7/8, R848 10 μg/mL; TLR9 CpG ODN 2006 1 μg/mL; NOD1, C12-iE-DAP 10 μg/mL; NOD2, L18-MDP 10 μg/mL; Dectin 1a and 1b, beta-glucan from Saccharomyces cerevisiae at 10 ng/mL, curdlan at 100 μg/mL and zymosan at 5 μg/mL.
2.10. Statistical analysis
Findings were analyzed for statistical significance using 1-way analysis of variance (ANOVA), Mann–Whitney U-test, or Log-rank test, as appropriate. Data are presented as mean ± standard deviation.
3. Results
3.1. Priming with L. plantarum results in suppression of a specific cohort of virus-induced proinflammatory mediators
Using a modification of our standard priming protocol (Gabryszewski et al., 2011), BALB/c mice were inoculated intranasally on day-14 and again on day-7 with 109 cells of live L. plantarum in pbs/bsa (50 μL of 2 × 1010 cells/mL) or pbs/bsa diluent control alone. In this experiment, mice are then challenged 21 days later (on day +14) with an otherwise lethal dose of PVM or vehicle only. RNA was isolated from whole lung tissue from 24 mice (four protocols, one time point, six mice per group) and was subjected to whole genome microarray analysis; differential expression of thirty-one (31) soluble proinflammatory mediators identified in this experiment is featured in Table 1. As shown here, PVM infection alone results in increased expression of transcripts encoding numerous inflammatory mediators, including CC and CXC chemokines, including CCL2 and CXCL10. Likewise, PVM infection results in increased expression of acute phase reactants amyloid A1 and A3 (Sandri et al., 2008), granzyme B, a serine protease with antiviral and immunomodulatory properties generated by NK cells, T cells and mast cells (Wensink et al., 2015), lipocalin 2, an immunomodulatory and antimicrobial protein stored in and released from a neutrophil tertiary granules (Dittrich et al., 2013; Flo et al., 2004), and galectins 3 and 9, galactose-binding proteins mediate innate and acquired immunity with roles including targeting the survival of effector cytotoxic T lymphocytes (CTLs), T helper 1 (TH1) cells and targeting regulatory T cells (Rabinovich and Toscano, 2009; Wu et al., 2014).
Table 1.
Significant (0.05, except where noted *) differential gene expression (≥1.5-fold) of virus-induced soluble proinflammatory mediators in response to priming with L. plantarum (Lp). BALB/c mice were inoculated intranasally with L. plantarum (109 cells) or diluent control (pbs/bsa; PBS) on days-14 and -7 (see Gabryszewski et al., 2011), followed by inoculation with pneumonia virus of mice (PVM; 0.2 TCID50 units/50 μL) or vehicle (VEH) control on day +14. Featured is the differential expression of 31 soluble proinflammatory mediators, a subset of the 839 differentially expressed transcripts detected by whole genome microarray from lung tissue evaluated on day 19, 5 days after inoculation of PVM.
Gene symbol | Entrez gene | Description | PBS/PVMa vs. VEH | Lp/PVMb vs. VEH | Lp/PVMc vs. PVM |
---|---|---|---|---|---|
Gzmb | 14939 | Granzyme B | 309.0 | 83.2 | |
Il6 | 16193 | Interleukin 6 | 231.5 | 2.2* | −105.0 |
Ccl2 | 20296 | Chemokine (C-C motif) ligand 2 | 225.3 | 16.6* | −13.6 |
Cxcl10 | 15945 | Chemokine (C-X-C motif) ligand 10 | 75.2 | 6.8 | −11.0 |
Lcn2 | 16819 | Lipocalin 2 | 68.0 | 20.5 | −3.3 |
Cxcl9 | 17329 | Chemokine (C-X-C motif) ligand 9 | 60.8 | 8.1* | |
Cxcl2 | 20310 | Chemokine (C-X-C motif) ligand 2 | 56.7 | 2.8* | −20.0 |
Saa3 | 20210 | Serum amyloid A 3 | 50.3 | 25.1 | |
Saa1 | 20208 | Serum amyloid A 1 | 50.1 | −1.7* | −86.7 |
Ccl7 | 20306 | Chemokine (C-C motif) ligand 7 | 47.2 | 3.4* | −13.8 |
Cxcl1 | 14825 | Chemokine (C-X-C motif) ligand 1 | 47.1 | 14.7 | |
Cxcl11 | 56066 | Chemokine (C-X-C motif) ligand 11 | 32.8 | 1.6* | −21.1 |
Ccl12 | 20293 | Chemokine (C-C motif) ligand 12 | 13.1 | 8.6* | |
Il1rn | 16181 | Interleukin 1 receptor antagonist | 9.8 | 4.3* | |
Lgals3 bp | 19039 | Lectin, galactoside-binding, soluble, 3 binding pro | 9.3 | 8.3 | |
Thbs1 | 21825 | Thrombospondin 1 | 8.9 | 1.6* | −5.5 |
S100a8 | 20201 | S100 calcium binding protein A8 (calgranulin A) | 7.3 | −1.0* | |
Csf1 | 12977 | Colony stimulating factor 1 (macrophage) | 7.2 | 3.6* | |
S100a9 | 20202 | S100 calcium binding protein A9 (calgranulin B) | 6.6 | −1.7* | −11.1 |
Lgals9 | 16859 | Lectin, galactose binding, soluble 9 | 6.2 | 3.0* | |
Cxcl13 | 55985 | Chemokine (C-X-C motif) ligand 13 | 4.0 | 3.9* | |
Ccl5 | 20304 | Chemokine (C-C motif) ligand 5 | 3.1 | 2.9* | |
Ccl9 | 20308 | Chemokine (C-C motif) ligand 9 | 3.0 | 4.6 | |
Cxcl16 | 66102 | Chemokine (C-X-C motif) ligand 16 | 3.0 | 1.7* | |
Lgals3 | 16854 | Lectin, galactose binding, soluble 3 | 2.6 | 2.4 | |
Hmgb1 | 15289 | High mobility group protein B1-like | −2.2 | −1.1* | 2.0 |
Cxcl15 | 20309 | Chemokine (C-X-C motif) ligand 15/IL8 | −2.2 | 1.3* | 2.8 |
Il33 | 77125 | Interleukin 33 | −2.9 | 1.9* | 5.4 |
Cxcl12 | 20315 | Chemokine (C-X-C motif) ligand 12 | −5.9 | −2.1* | 2.8 |
Ccl8 | 20307 | Chemokine (C-C motif) ligand 8 | 3.4* | 16.7 | 5.0 |
Ccl6 | 20305 | Chemokine (C-C motif) ligand 6 | −1.3* | 3.8 | 4.9 |
Mice primed with diluent (PBS) and inoculated with PVM vs. mice primed with diluent and inoculated with vehicle (VEH).
Mice primed with L. plantarum (Lp) and inoculated with PVM vs. mice primed with diluent (PBS) and inoculated with vehicle (VEH).
Mice primed with L. plantarum (Lp) and inoculated with PVM vs. mice primed with diluent (PBS) and inoculated with PVM; only statistically significant differences are shown.
Values not significant (>0.05) over PBS/VEH.
Interestingly, not all pro-inflammatory mediators were up-regulated in response to PVM infection. Specifically, we observed diminished expression of epithelial cytokines and alarmins, HMGB1 and IL-33 in response to PVM infection alone, and priming with L. plantarum counteracts this response to some degree. Release of IL-33 and HMGB1 from host cells and their role in augmenting virus-induced asthma has been considered previously in the literature (Yoo et al., 2013; Kumar et al., 2014), while more recent work (Holtzman et al., 2014) has identified a population of specialized epithelial cells that produces IL-33 as a response to recovery from virus infection. Interestingly, expression of the epithelial-derived chemokine CXCL15 (originally known as lungkine (Rossi et al., 1999; Chen et al., 2001)), is also diminished in response to PVM infection, a response that also is reversed by L. plantarum priming.
Most important with respect to the findings in this manuscript, the microarray findings document that priming of the respiratory tract with L. plantarum prior to virus infection results in a specific anti-inflammatory profile. Among those inflammatory mediators with expression most profoundly suppressed was virus-induced interleukin (IL)-6, with expression diminished 105-fold in response to L. plantarum priming. Other chemokines that undergo profound suppression include CCL2, CXCL10, CXCL2 and CXCL11, with 14, 11, 20 and 21-fold reduced expression, respectively. In contrast, other chemokines, including CCL12, CXCL13, CCL5, CCL9 and CXCL16, experience no significant differential expression in response to L. plantarum priming.
3.2. L. plantarum and L. reuteri administered to the respiratory mucosa after virus challenge also avert the lethal sequelae of infection
In our previous work, we focused on L. plantarum and L. reuteri and their role in preventing the lethal sequelae of respiratory virus infection when administered as a priming agent, prior to virus challenge. Here, BALB/c mice were inoculated with PVM at day 0, followed by intranasal administration L. plantarum or L. reuteri (107 to 109 cells/mouse) at one or more days after virus challenge as outlined in Fig. 1A. Mice that received L. plantarum (107 cells/-mouse) on days 1 and 2 or on days 1, 2, and 3 responded with 40% survival. In contrast, if the first dose was delayed until day 2, and mice received 107 cells on days 2 and 3, no significant survival over background was observed (Fig. 1B). In contrast, mice that received a single inoculum of 108 cells on day 1 alone were fully protected (100% survival) against an otherwise lethal PVM infection (Fig. 1B); we have been able to extend the “window of opportunity” beyond the first 24 h with varying dosing strategies (data not shown). Similar to results obtained with Lactobacillus priming prior to virus infection (Gabryszewski et al., 2011), we show here that live L. plantarum is just as effective as the heat-inactivated in preventing the lethal sequelae of PVM infection when administered post-virus challenge (Fig. 1C). Furthermore, protection elicited post-virus challenge is not a unique property of a given Lactobacillus strain or species, as both live and heat-inactivated L. reuteri were also effective in this role (Fig. 1D).
Fig. 1.
L. plantarum or L. reuteri introduced after virus challenge promote survival in response to an otherwise lethal infection in BALB/c mice. (A) Experimental protocol: BALB/c mice are inoculated with PVM at day 0 followed by intranasal inoculations with live or heat-inactivated (hi) L. plantarum (Lp) or L. reuteri (Lr) at 2 × 108 cells/mL (107 cells/mouse), 2 × 109 cells/mL (108 cells/mouse), 2 × 1010 cells/mL (109 cells/mouse), or diluent control (pbs/bsa) in a 50 μL volume at day (d)1, d2, and/or d3 as indicated. (B) Survival of mice inoculated with hi Lp or pbs/bsa diluent control as in (A); ***p < 0.001, **p < 0.01 vs. pbs/bsa, n = 5–10 mice per group. (C) Survival of mice inoculated with live or hi Lp or pbs/bsa diluent control 108 cells/mouse or pbs/bsa diluent control on d1 and d2 (see (A)); ***p < 0.001, n = 5 mice per group. (D) Survival of mice inoculated with live or hi Lr at 109 cells/mouse or pbs/bsa diluent control on d1; n = 5 mice per group (see (A)), ***p < 0.001.
Heat-inactivated (hi) L. plantarum administered post-virus challenge results in diminished virus recovery (Fig. 2A). Lung tissue from PVM-infected mice that received pbs/bsa diluent control displayed prominent alveolitis and congestion (Fig. 2B, i, at arrows), in contrast to lungs from PVM-infected mice that received hi-L. plantarum on days 1 and 2 after PVM challenge, which were comparatively clear (Fig. 2B, ii). Furthermore, analogous to what we have observed in response to L. plantarum priming (Table 1), survival in response to either heat-inactivated or live L. plantarum administered after PVM challenge resulted in profound suppression of virus-induced cytokines (Fig. 2C and D).
Fig. 2.
L. plantarum introduced after virus challenge limits virus recovery and virus-induced inflammation. (A) Virus titer in lung tissue on day 5 after inoculation with PVM from mice that received intranasal inoculations of hi Lp, 108 cells/mouse on d1 or on d1 and d2; *p < 0.05; **p < 0.01 vs. pbs/bsa. (B) Formalin-fixed, hematoxylin and eosin-stained lung tissue from mice inoculated on d1 and d2 as described in the legend to (A); arrows in panel i. point to areas of inflammation. (C) Proinflammatory cytokines IL-6, CCL2, and CXCL10 in lung tissue of mice inoculated as in the legend to (A); *p < 0.05; **p < 0.01. (D) Proinflammatory cytokines IL-6 and CCL2 in lung tissue on day 5 after inoculation with PVM from mice that received intranasal inoculations of live Lp, 108 cells/mouse on d1 and d2; **p < 0.01.
Administration of hi L. plantarum on days 1 and 2 after PVM challenge also limits neutrophil recruitment to the lungs (Fig. 3). Specifically, we detected 5–6-fold fewer neutrophils at day 5, a reduction from 14 ± 1.5 × 106 to 2.6 ± 0.5 × 106 cells in response to L. plantarum administered post-virus challenge (p < 0.001). We observed no significant differences in total CD4+ T cells, CD8+ T cells, B cells, NK or NKT cells at this time point, although L. plantarum administration did result in increased numbers of eosinophils and dendritic cells.
Fig. 3.
Leukocytes identified in lung tissue of PVM-infected mice that treated with L. plantarum vs. diluent control. Total leukocytes (all CD45+) from single cell suspensions prepared from lungs of PVM-infected (PVM on day 0) BALB/c mice on day 5 after intranasal inoculation of heat-inactivated Lp (108 cells/mouse/inoculum) or diluent (pbs/bsa) on d1 and d2. (A) Alv MΦs, alveolar macrophages (CD11c+SiglecF+); DCs, dendritic cells (CD11c+MHCII+); mono-mΦs, monocyte/macrophages (CD11c+MHCII−); Eos, eosinophils, (SiglecF+CD11c−); PMNs, neutrophils, (Gr1+). (B) NK (natural killer) cells, (CD49b+); NKT cells (CD3+CD49b+). (C) CD4+ T cells; CD8+ T cells; B cells (CD19+); n = 5 per group; *p < 0.05, **p < 0.01.
Mice on the C57BL/6 background respond similarly to Lactobacillus administered after virus challenge. As shown in Fig. 4A, mice are protected in response to 108 or 109 cells of hi L. plantarum administered on days 1 and 2, or 109 cells administered on day 1 alone; protection is also conferred by 109 cells of live L. reuteri (Fig. 4B). Similar to observations in BALB/c mice, protection in response to L. plantarum in C57BL/6 mice is associated with profound suppression of virus-induced neutrophil recruitment (Fig. 4C), diminished virus recovery (Fig. 4D), and suppression of virus-induced proinflammatory cytokines (Fig. 4E and F).
Fig. 4.
L. plantarum introduced after virus challenge promotes survival in association with suppression of virus-induced inflammation in C57BL/6 mice. (A) Survival of mice inoculated with hi L. plantarum (Lp) as in Fig. 1A; ***p < 0.001 vs. pbs/bsa; n = 5–10 mice per group. (B) Survival of mice inoculated with live L. reuteri (Lr) as in Fig. 1A; ***p < 0.001 vs. pbs/bsa; n = 5 mice per group. (C) Leukocytes in bronchoalveolar lavage (BAL) fluid from mice challenged with PVM on day 0 and pbs/bsa or hi Lp (109 cells) on d1; *p < 0.05; mΦs, macrophages; eos, eosinophils; pmns, neutrophils. (D) Virus titer, mice inoculated as described in the legend to (C), *p < 0.05. (E) IL-6 and (F) CCL2 in lung tissue from mice inoculated as described in the legend to (C), *p < 0.05, n = 5 mice per group.
3.3. Protection afforded by L. plantarum is not mediated by IL-10
The cytokine IL-10 is produced by monocytes, Th2 cells and regulatory T cells (Saraiva and O’Garra, 2010) and its role in modulating the anti-inflammatory impact of probiotic bacteria, including Lactobacillus, in the gastrointestinal tract has been explored extensively (Levast et al., 2015; Mengheri, 2008). Interestingly, gene-deletion of IL-10 has no impact on Lactobacillus-mediated protection at the respiratory tract; administration of L. plantarum to the respiratory tract (109 cells/mouse on days 1 and 2 after virus challenge) results in full protection from the lethal sequelae of PVM infection in both wild-type and IL-10−/− mice (Fig. 5A). Administration of L. plantarum to PVM-infected IL-10−/− mice results in diminished virus recovery from lung tissue (Fig. 5B) and prominent suppression of cytokines IL-6 and CCL2 (Fig. 5C).
Fig. 5.
The anti-inflammatory cytokine interleukin-10 (IL-10) does not mediate survival promoted by hi L. plantarum at the respiratory mucosa. (A) Survival of wild-type (wt) BALB/c and IL-10 gene-deleted (IL-10−/−) mice inoculated with PVM at day 0 followed by hi L. plantarum (108 cells/mouse) or diluent control at days (d1) and d2 as shown; ***p < 0.001 vs. mice receiving pbs/bsa, n = 5–15 mice per group. (B) Virus titer in lung tissue of IL-10−/− mice inoculated as in (A) evaluated at d5; ***p < 0.001. (C) Proinflammatory cytokines IL-6 and CCL2 in lung tissue of mice inoculated as in (A) and evaluated on d5; **p < 0.01 n = 5 mice per group.
3.4. L. plantarum interacts in vitro with pattern recognition receptors TLR2 and NOD2
In order to define clearly the specific interactions of Lactobacillus with mucosal immune cells, hi L. plantarum was evaluated in assays designed to identify responses of individual pattern recognition receptors (PRRs) via an NF-kB-linked colorimetric readout (see Materials and Methods). As shown in Fig. 6A, L. plantarum elicits responses that would be anticipated from a gram-positive microorganism, as it activates via both toll-like receptor 2 (TLR2) and nucleotide-binding oligomerization domain-containing protein 2 (NOD2), at 20-fold and 6-fold over diluent control, respectively. We observed no response from any other TLR, from NOD1, or of from C-type lectin-receptors Dectin1a or Dectin1b (data not shown), although positive ligands were uniformly reactive (see Materials and Methods).
Fig. 6.
TLR2 (TLR2−/−) and NOD2 (NOD2−/−) gene-deleted mice respond to L. plantarum and are protected from the lethal sequelae of PVM infection. (A) Activation of PRRs expressed by HEK293 transfectants; detection via products of secretory alkaline phosphatase (A650) in an NF-kB-linked activation assay (Invivogen). Shown are results of triplicates of independent assays of PRRs challenged with 108 cells/mL hi L. plantarum (black filled bars) or control diluent alone (open bars); **p < 0.01 vs. control. (B) Survival of wild-type, TLR2−/− and NOD2−/− mice inoculated with PVM at day 0 followed by hi L. plantarum (109 cells/mouse) at d1 and d2 as in Fig. 1A; ***p < 0.001, ***p < 0.001; **p < 0.02 vs. mice of the same strain receiving diluent control, n = 5–10 mice per group. (C) Virus titer in lung tissue of NOD2−/− mice inoculated with diluent only (−) or with hi L. plantarum (+) as in (B) and evaluated on day 7. (D and E) Expression proinflammatory cytokines IL-6 and CCL2 in lung tissue of NOD2−/− mice inoculated with diluent (−) or with hi L. plantarum (+) as in (B) and evaluated on day 7; *p < 0.05, **p < 0.01; n = 4–5 per group. (F) Survival of wild-type and IFNαβR−/− mice inoculated with PVM at day 0 followed by hi L. plantarum (109 cells/mouse) on d1 and d2 as in Fig. 1A; ***p < 0.001 vs. mice of the same strain receiving diluent control, n = 5–10 mice per group.
3.5. Immunomodulation and protection elicited by L. plantarum in mice with single gene-deletions of TLR2 or NOD2
Wild-type C57BL/6, TLR2−/−, or NOD2−/− mice that were inoculated with virus on day 0 and with L. plantarum (109 cells/mouse) on days 1 and 2 were fully protected from the lethal sequelae of infection compared to their counterparts that received pbs/bsa only (Fig. 6B). Differential virus recovery was not a prominent component of the response to L. plantarum in NOD2−/− mice, results which are similar to those obtained in experiments performed previously with L. plantarum-primed MyD88−/− mice (Gabryszewski et al., 2011; Fig. 6C). However, L. plantarum administration did result in suppression of virus-induced cytokines IL-6 and CCL2 (Fig. 6D and E). Similarly, mice devoid of type I interferon (αβ) receptors also respond to L. plantarum, and are fully protected from the lethal sequelae of PVM infection under the same experimental conditions (Fig. 6F).
4. Discussion
We demonstrate here for the first time that L. plantarum is effective at promoting survival when administered to the respiratory tract after virus challenge. While we and others have explored the priming of the respiratory mucosa with L. plantarum as a means to prevent the negative sequelae of subsequent infection with various viruses (Gabryszewski et al., 2011; Garcia-Crespo et al., 2013; Percopo et al., 2014; Hori et al., 2001; Harata et al., 2010; Izumo et al., 2010; Park et al., 2013; Tomasada et al., 2013; see also Supplemental Fig. 1), this is the first example of a the use of this methodology for definitive intervention after virus exposure has taken place. The fact that we identified a significant “window of opportunity” for complete (100%) post-exposure protection is particularly notable given that, in earlier work, we observed no protection whatsoever in response to live or heat-inactivated L. plantarum administered shortly thereafter, at day 3 post-virus challenge (Gabryszewski et al., 2011). We continue to explore the mechanisms underlying susceptibility (and resistance) to Lactobacillus-mediated suppression.
Similar to what we have observed in response to priming prior to PVM infection, administration of L. plantarum after virus challenge is associated with diminished virus recovery, and with suppression of virus-induced neutrophil recruitment and production of cytokine mediators, including IL-6, CCL2 and CXCL10. Each of these mediators has been identified in clinical studies in association with disease severity [Tabarani et al., 2013; Sow et al., 2011; Lee et al., 2011; Ichikawa et al., 2013; Doyle et al., 2010].
The clinical implications of our findings regarding L. plantarum and its efficacy post-virus challenge, and our interest in its unique antiviral mechanism at the respiratory mucosa, are clear and significant. One is often fully aware of acute exposure to respiratory virus infection, for example, in crowded venues such as public transportation and in office or classroom settings. A 24 h window of opportunity after primary exposure, such as that revealed in this study, is a reasonable time frame in which to administer a preventive agent, particularly one that can be provided simply and efficiently via an intranasal route. This is in contrast to an agent based on mechanisms defined by Lactobacillus priming, which would be designed to provide generalized protection against respiratory virus infection prior to exposure, and might be directed against pathogens for which vaccine strategies have limited efficacy and/or are unavailable.
There are only a few agents currently available for use in preventing the negative sequelae of respiratory virus infection after an initial exposure has taken place. Among these agents are the neuraminidase inhibitors, which are used for both prophylaxis and treatment of influenza; when administered within 48 h of the onset of symptoms, these antivirals may limit duration of disease and ensuing complications (reviewed in Nguyen-Van-Tam et al., 2015) although the overall risk vs. benefits of these drugs have been questioned (Jefferson et al., 2014). With respect to respiratory syncytial virus (RSV), the monoclonal antibody palivizumab directed against the virus F (fusion) protein is highly effective at preventing severe disease among those infants identified at high risk (reviewed in Andabaka et al., 2013) although this does not represent the full measure of the disease burden (Hall et al., 2009). At the same time, treatment options for RSV disease are limited. The antiviral ribavirin is available for only the most severely ill and immunocompromised children (AAP Committee on Infectious Diseases, 1996; Chu and Englund, 2013). A number of drugs are currently in development for the treatment of RSV disease, including antivirals based on siRNA (DeVincenzo et al., 2010) and the cell fusion inhibitor GS-5806 (DeVincenzo et al., 2014). Just recently, Plant et al. (2015) have described a high-throughput screening assay designed to identify agents that inhibit RSV replication. Our groups have focused on the role of the inflammation and its contribution to acute respiratory virus infection, and we have documented the efficacy of combined antiviral/anti-inflammatory approaches in studies carried out using in vivo models (Rosenberg and Domachowske, 2012; Bonville et al., 2003; Bonville et al., 2004).
Among the intriguing questions is the mechanism via which L. plantarum at the respiratory tract limits virus replication in vivo (see Figs. 2A and 4D). Among several possibilities, several groups (Uehara et al., 2007; Birchler et al., 2001; Wang et al., 2003) have reported that epithelial cells respond to gram-positive bacteria and their components by producing beta-defensins, antimicrobial peptide with significant antiviral activity (Lehrer, 2004; Gong et al., 2010). Furthermore, in recent work with a novel recombinant PVM strain J3666 tagged with the red fluorescent protein, mKATE2, we have identified alveolar macrophages (CD45+ SiglecF+ CD11c+) as the major leukocyte population that supports virus replication in vivo. Lactobacillus-priming of the respiratory tract results in a significant reduction in mKATE2+ alveolar macrophages (from 30% to 40% to fewer than 10%) in association with 2-log drop in release of infectious virions (Dyer et al., ms. in revision).
Also clear from these studies, the interactions of Lactobacillus with the local immune environment of the gastrointestinal mucosa are functionally distinct from what we observe in the respiratory tract. Among the most prominent differences, Lactobacillus-mediated protection from inflammatory sequelae has been attributed in large part to the actions of the anti-inflammatory cytokine, IL-10 (reviewed in Claes et al., 2011). For example, Macho Fernandez et al. (2011) showed that peptidoglycan derived Lactobacillus salivarius strain Ls33 served to protect mice from the inflammatory sequelae of chemical colitis via mechanisms that correlated with local production of IL-10. Similarly, Chen et al. (2005) found that inoculation of young mice with Lactobacillus acidophilus stimulated IL-10 expression in conjunction with protection against colitis induced by the bacterial pathogen, Citrobacter rodentium. Recently, Bosch et al. (2012) characterized novel probiotic strains of L. plantarum based on their capacity to induce IL-10 production in vitro from human monocyte THP-1 cells when co-incubated with lipopolysaccharide. In contrast, as we show in Fig. 5, inflammatory suppression and full protection against the lethal sequelae mediated by L. plantarum at the respiratory tract takes place in mice devoid of IL-10, as it does in their wild-type counterparts.
We also explored responses elicited by L. plantarum in mice devoid of pattern recognition receptors, TLR2 and NOD2. While Lactobacillus interacts with TLR2 and NOD2 alone in in vitro signaling assays (Fig. 6), we found that mice with individual gene deletions were protected in both priming (data not shown) and post-virus challenge protocols. The survival responses using priming protocols in TLR2−/− mice (data not shown) may have been anticipated to some extent given our earliest findings with priming in MyD88−/− mice (Gabryszewski et al., 2011); however we had not previously explored the responses of either strain in the post-virus challenge protocol. Likewise, we have also shown that mice devoid of type I IFN receptors (ie, thus devoid of downstream signaling from IFNs resulting from TLR2 activation) are similarly protected by L. plantarum. Among several interpretations, it is conceivable that there may be substantial cross-talk between TLR2 and NOD2 pathways in this setting (Wu et al., 2015; Zeuthen et al., 2008; Borm et al., 2008; Netea et al., 2005; Pavot et al., 2014; Watanabe et al., 2006), a possibility that requires further consideration.
Among other possibilities, Lactobacillus species may elicit protection via interactions with another, fully independent group of PRRs, the peptidoglycan recognition proteins (PGLYRPs), a family of evolutionarily ancient secretory/bactericidal proteins that interact with bacterial cell wall peptidoglycan (Royet et al., 2011). Of particular note is PGLYRP-1, which is expressed primarily in neutrophil granules, and was recently identified by Read et al. (2015) as a ligand for the orphan neutrophil and monocyte/macrophage Ig superfamily receptor TREM-1, which augments inflammatory and innate immune responses triggered by exogenous pathogens by stimulating release of pro-inflammatory mediators (Ford and McVicar, 2009). Of particular interest, PGLYP-1 acts either independently or in combination with PGN to promote signaling at this receptor. In our previous work (Garcia-Crespo et al., 2013), we found that delivery of high concentrations of peptidoglycan resulted in prolonged survival (t½ = 9.0 vs. 10.5 days, *p < 0.05) upon subsequent challenge with PVM, although not 100% protection that we readily observe in response to Lactobacillus. As such, PGLYRPs are unlikely to function alone to promote Lactobacillus-mediated protection. However, the fact that we observe even a minor response to PGN suggests that we cannot rule out the possibility of some contributions from these receptors.
5. Conclusions
In summary, L. plantarum is a versatile and flexible agent for prevention of lethal sequelae of respiratory virus infection via its actions at the respiratory mucosa. These interactions are distinct from those elicited by Lactobacillus species at the gastrointestinal mucosa, and do not rely on signaling mediated by IL-10. We demonstrated previously that priming of the respiratory mucosa with L. plantarum results in robust and sustained protection against lethal infection with PVM; we demonstrate here that administration of heat-inactivated L. plantarum within 24 h after virus challenge also results in full survival from an otherwise lethal virus challenge, a finding of significant clinical potential. In vitro signaling assays indicate that L. plantarum interacts with PRRs NOD2 and TLR2, yet mice devoid of each of these individual PRRs remain fully protected against lethal PVM infection by L. plantarum. Taken together, these results speak to the possibility of (a) significant redundancy of these critical responses and/or (b) mechanisms that complement, bypass or supersede PRR responses.
Supplementary Material
Acknowledgments
The authors thank Dr. Kimmo Virtaneva of the Genomics Unit, RTS, for his assistance with experimental design and sample processing, and Dr. Kirk M. Druey (MSTS/LAD/NIAID) for critical review of this manuscript. This research was funded by Division of Intramural Research AI000943 to H.F.R. and Collaborative Research and Development Agreement 2013-0510 between NIAID and GlaxoSmithKline.
Abbreviations
- PVM
pneumonia virus of mice
- Lp
Lactobacillus plantarum
- Lr
Lactobacillus reuteri
Appendix A. Supplementary data
Supplementary data associated with this article can be found, in the online version, at http://dx.doi.org/10.1016/j.antiviral.2015.07.001.
References
- American Academy of Pediatrics Committee on Infectious Diseases. Reassessment of the indications for ribavirin therapy in respiratory syncytial virus infections. Pediatrics. 1996;97:137–140. [PubMed] [Google Scholar]
- Andabaka T, Nickerson JR, Rojas-Reyes MX, Rueda JD, Bacic Vrca V, Barsic B. Monoclonal antibody for reducing the risk of respiratory syncytial virus infection in children. Cochrane Database Syst Rev. 2013;4:CD006602. doi: 10.1002/14651858.CD006602.pub4. [DOI] [PubMed] [Google Scholar]
- Bem RA, Domachowske JB, Rosenberg HF. Animal models of human respiratory syncytial virus disease. Am J Physiol Lung Cell Mol Physiol. 2011;301:L148–L156. doi: 10.1152/ajplung.00065.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Birchler T, Seibl R, Büchner K, Loeliger S, Reinhard S, Hossle JP, Aguzzi A, Lauener RP. Human Toll-like receptor 2 mediates induction of the antimicrobial peptide human beta-defensin 2 in response to bacterial lipoprotein. Eur J Immunol. 2001;31:3131–3137. doi: 10.1002/1521-4141(200111)31:11<3131::aid-immu3131>3.0.co;2-g. [DOI] [PubMed] [Google Scholar]
- Bonville CA, Easton AJ, Rosenberg HF, Domachowske JB. Altered pathogenesis of severe pneumovirus infection in response to combined antiviral and specific immunomodulatory agents. J Virol. 2003;77:1237–1244. doi: 10.1128/JVI.77.2.1237-1244.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bonville CA, Lau VK, DeLeon JM, Gao JL, Easton AJ, Rosenberg HF, Domachowske JB. Functional antagonism of chemokine receptor CCR1 reduces mortality in acute pneumovirus infection in vivo. J Virol. 2004;78:7984–7989. doi: 10.1128/JVI.78.15.7984-7989.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Borm ME, van Bodegraven AA, Mulder CJ, Kraal G, Bouma G. The effect of NOD2 activation on TLR2-mediated cytokine responses is dependent on activation dose and NOD2 genotype. Genes Immun. 2008;9:274–278. doi: 10.1038/gene.2008.9. [DOI] [PubMed] [Google Scholar]
- Bosch M, Rodriguez M, Garcia F, Fernandez E, Fuentes MC, Cune J. Probiotic properties of Lactobacillus plantarum CECT 7315 and CECT 7316 isolated from the faeces of healthy children. Lett Appl Microbiol. 2012;54:240–246. doi: 10.1111/j.1472-765X.2011.03199.x. [DOI] [PubMed] [Google Scholar]
- Bron PA, van Barlen P, Kleerebezem M. Emerging insights into the interaction between probiotics and the host intestinal mucosa. Nat Rev Microbiol. 2011;10:66–78. doi: 10.1038/nrmicro2690. [DOI] [PubMed] [Google Scholar]
- Chan-Tack KM, Kim IC, Moruf A, Birnkrant DB. Clinical experience with intravenous zanamivir under an Emergency IND program in the United States (2011–2014) Antivir Ther. 2015 doi: 10.3851/IMP2944. in press. [DOI] [PubMed] [Google Scholar]
- Chen SC, Mehrad B, Deng JC, Vassileva G, Manfra DJ, Cook DN, Wiekowski MT, Zlotnik A, Standiford TJ, Lira SA. Impaired pulmonary host defense in mice lacking expression of the CXC chemokine lungkine. J Immunol. 2001;166:3362–3368. doi: 10.4049/jimmunol.166.5.3362. [DOI] [PubMed] [Google Scholar]
- Chen CC, Louis S, Shi HN, Walker WA. Preinoculation with the probiotic Lactobacillus acidophilus early in life effectively inhibits murine Citrobacter rodentium colitis. Pediatr Res. 2005;58:1185–1191. doi: 10.1203/01.pdr.0000183660.39116.83. [DOI] [PubMed] [Google Scholar]
- Chu HY, Englund JA. Respiratory syncytial virus disease: prevention and treatment. Curr Top Microbiol Immunol. 2013;372:235–288. doi: 10.1007/978-3-642-38919-1_12. [DOI] [PubMed] [Google Scholar]
- Claes IJ, De Keersmaecker SC, Vanderleyden J, LeBeer S. Lessons from probiotic-host interaction studies in murine models of experimental colitis. Mol Nutr Food Res. 2011;55:1441–1453. doi: 10.1002/mnfr.201100139. [DOI] [PubMed] [Google Scholar]
- DeVincenzo J, Lambkin-Williams R, Wilkinson T, Cehelski J, Nochur S, Walsh E, Meyers R, Gollob J, Vaishnaw A. A randomized, double-blind, placebo-controlled study of an RNAi-based therapy directed against respiratory syncytial virus. Proc Natl Acad Sci USA. 2010;107:8800–8805. doi: 10.1073/pnas.0912186107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DeVincenzo JP, Whitley RJ, Mackman RL, Scaglioni-Weinlich C, Harrison L, Farrell E, McBride S, Lambkin-Williams R, Jordan R, Xin Y, Ramanathan S, O’Riordan T, Lewis SA, Li X, Toback SL, Lin SL, Chien JW. Oral GS-5806 activity in a respiratory syncytial virus challenge study. N Engl J Med. 2014;371:711–722. doi: 10.1056/NEJMoa1401184. [DOI] [PubMed] [Google Scholar]
- Dittrich AM, Meyer HA, Hamelmann E. The role of lipocalins in airway disease. Clin Exp Allergy. 2013;43:503–511. doi: 10.1111/cea.12025. [DOI] [PubMed] [Google Scholar]
- Domachowske JB, Bonville CA, Dyer KD, Easton AJ, Rosenberg HF. Pulmonary eosinophilia and production of MIP-1alpha are prominent responses to infection with pneumonia virus of mice. Cell Immunol. 2000;200:98–104. doi: 10.1006/cimm.2000.1620. [DOI] [PubMed] [Google Scholar]
- Doyle WJ, Casselbrant ML, Li-Korotky HS, Doyle AP, Lo CY, Turner R, Cohen S. The interleukin 6–174 C/C genotype predicts greater rhinovirus illness. J Infect Dis. 2010;201:199–206. doi: 10.1086/649559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dyer KD, Garcia-Crespo KE, Glineur S, Domachowske JB, Rosenberg HF. The pneumonia virus of mice (PVM) model of acute respiratory infection. Viruses. 2012;4:3494–3510. doi: 10.3390/v4123494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Edgar R, Domrachev M, Lash AE. Gene expression omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002;30:207–210. doi: 10.1093/nar/30.1.207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Esposito S, Rigante D, Principi N. Do children’s upper respiratory tract infections benefit from probiotics? BMC Infect Dis. 2014;14:194. doi: 10.1186/1471-2334-14-194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Flo TH, Smith KD, Sato S, Rodriguez DJ, Holmes MA, Strong RK, Akira S, Aderem A. Lipocalin 2 mediates an innate immune response to bacterial infection by sequestrating iron. Nature. 2004;432:917–921. doi: 10.1038/nature03104. [DOI] [PubMed] [Google Scholar]
- Ford JW, McVicar DW. TREM and TREM-like receptors in inflammation and disease. Curr Opin Immunol. 2009;21:38–46. doi: 10.1016/j.coi.2009.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gabryszewski SJ, Bachar O, Dyer KD, Percopo CM, Killoran KE, Domachowske JB, Rosenberg HF. Lactobacillus-mediated priming of the respiratory mucosa protects against lethal pneumovirus infection. J Immunol. 2011;186:1151–1161. doi: 10.4049/jimmunol.1001751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garcia-Crespo KE, Chan CC, Gabryszewski SJ, Percopo CM, Rigaux P, Dyer KD, Domachowske JB, Rosenberg HF. Lactobacillus priming and respiratory virus infection: heterologous immunity and protection against lethal pneumovirus infection. Antiviral Res. 2013;97:270–279. doi: 10.1016/j.antiviral.2012.12.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gong T, Jiang Y, Wang Y, Yang D, Li W, Zhang Q, Feng W, Wang B, Jiang Z, Li M. Recombinant mouse beta-defensin 2 inhibits infection by influenza A by blocking its entry. Arch Virol. 2010;155:491–498. doi: 10.1007/s00705-010-0608-1. [DOI] [PubMed] [Google Scholar]
- Hall CB, Weinberg GA, Iwane MK, Blumkin AK, Edwards KM, Staat MA, Auinger P, Griffin MR, Poehling KA, Erdman D, Grijalva CG, Zhu Y, Szilagyi P. The burden of respiratory syncytial virus infection in young children. N Engl J Med. 2009;360:588–598. doi: 10.1056/NEJMoa0804877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hao Q, Lu Z, Don BR, Huang CQ, Wu T. Probiotics for preventing acute upper respiratory tract infections. Cochrane Database Syst Rev. 2011;9:CD006895. doi: 10.1002/14651858.CD006895.pub2. [DOI] [PubMed] [Google Scholar]
- Harata G, He F, Hiruta N, Kawase M, Kubota A, Hiramatsu M, Yasui H. Intranasal administration of Lactobacillus rhamnosus GG protects mice from H1N1 influenza virus infection by regulating respiratory immune responses. Lett Appl Microbiol. 2010;50:597–602. doi: 10.1111/j.1472-765X.2010.02844.x. [DOI] [PubMed] [Google Scholar]
- Holtzman MJ, Byers DE, Brett JA, Patel AC, Agapov E, Jin X, Wu K. Linking acute infection to chronic lung disease. The role of IL-33-expressing epithelial progenitor cells. Ann Am Thorac Soc Suppl. 2014;5:S287–S291. doi: 10.1513/AnnalsATS.201402-056AW. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hori T, Kiyoshima J, Hida K, Yasui H. Effect of intranasal administration of Lactobacillus casei Shirota on influenza virus infection of upper respiratory tract in mice. Clin Diagn Lab Immunol. 2001;8:593–597. doi: 10.1128/CDLI.8.3.593-597.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ichikawa A, Kuba K, Morita M, Chida S, Tezuka H, Hara H, Sasaki T, Ohteki T, Ranieri VM, dos Santos CC, Kawaoka Y, Akira S, Luster AD, Lu B, Penninger JM, Uhlig S, Slutsky AS, Imai Y. CXCL10-CXCR3 enhances the development of neutrophil-mediated fulminant lung injury of viral and nonviral origin. Am J Respir Crit Care Med. 2013;187:65–77. doi: 10.1164/rccm.201203-0508OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ifrim DC, Quintin J, Joosten LAB, Jacobs C, Jansen T, Jacobs L, Gow NAR, Williams DL, van der Meer JWM, Netea MG. Trained immunity or tolerance: opposing functional programs induced in human monocytes after engagement of various pattern recognition receptors. Clin Vaccine Immunol. 2014;21:534–545. doi: 10.1128/CVI.00688-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Izumo T, Maekawa T, Ida M, Noguchi A, Kitagawa Y, Shibata H, Yasui H, Kito Y. Effect of intranasal administration of Lactobacillus pentosus S-PT84 on influenza virus infection in mice. Int Immunopharmacol. 2010;10:1101–1106. doi: 10.1016/j.intimp.2010.06.012. [DOI] [PubMed] [Google Scholar]
- Jefferson T, Jones MA, Doshi P, Del Mar CB, Hama R, Thompson MJ, Spencer EA, Onakpoya I, Mahtani KR, Nunam D, Howick J, Heneghan CJ. Neuraminidase inhibitors for preventing and treating influenza in healthy adults and children. Cochrane Database Syst Rev. 2014;4:CD008965. doi: 10.1002/14651858.CD008965.pub3. [DOI] [PubMed] [Google Scholar]
- Klipper-Aurbach Y, Wasserman M, Braunspiegel-Weintrob N, Borstein D, Peleg S, Assa S, Karp M, Benjamini Y, Hochbert Y, Laron Z. Mathematical formulae for the prediction of the residual beta cell function during the first two years of disease in children and adolescents with insulin-dependent diabetes mellitus. Med Hypotheses. 1995;45:486–490. doi: 10.1016/0306-9877(95)90228-7. [DOI] [PubMed] [Google Scholar]
- Kumar RK, Foster PS, Rosenberg HF. Respiratory viral infection, epithelial cytokines, and innate lymphoid cells in asthma exacerbations. J Leukoc Biol. 2014;96:391–396. doi: 10.1189/jlb.3RI0314-129R. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lebeer S, Vanderleyden J, De Keersmaecker SCJ. Host interactions of probiotic bacterial surface molecules: comparison with commensals and pathogens. Nat Rev Microbiol. 2010;8:171–184. doi: 10.1038/nrmicro2297. [DOI] [PubMed] [Google Scholar]
- Lee N, Chan PK, Wong CK, Wong KT, Choi KW, Joynt GM, Lam P, Chan MC, Wong BC, Lui GC, Sin WW, Wong RY, Lam WY, Yeung AC, Leung TF, So HY, Yu AW, Sung JJ, Hui DS. Viral clearance and inflammatory response patterns in adults hospitalized for pandemic 2009 influenza A (H1N1) virus pneumonia. Antivir Ther. 2011;16:237–247. doi: 10.3851/IMP1722. [DOI] [PubMed] [Google Scholar]
- Lehrer RI. Primate defensins. Nat Rev Microbiol. 2004;2:727–738. doi: 10.1038/nrmicro976. [DOI] [PubMed] [Google Scholar]
- Levast B, Li Z, Madrenas J. The role of IL-10 in microbiome-associated immune modulation and disease tolerance. Cytokine. 2015 doi: 10.1016/j.cyto.2014.11.027. In press. [DOI] [PubMed] [Google Scholar]
- Levy O, Netea MG. Innate immune memory: implications for development of pediatric immunomodulatory agents and adjuvanted vaccines. Pediatr Res. 2014;75:184–188. doi: 10.1038/pr.2013.214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Locati M, Mantovani A, Sica A. Macrophage activation and polarization as an adaptive component of innate immunity. Adv Immunol. 2013;120:163–184. doi: 10.1016/B978-0-12-417028-5.00006-5. [DOI] [PubMed] [Google Scholar]
- Macho Fernandez E, Valenti V, Rockel C, Hermann C, Pot B, Boneca IG, Grangette C. Anti-inflammatory capacity of selected lactobacilli in experimental colitis is driven by NOD2-mediated recognition of a specific peptidoglycan-derived muropeptide. Gut. 2011;60:1050–1059. doi: 10.1136/gut.2010.232918. [DOI] [PubMed] [Google Scholar]
- Mackey-Lawrence NM, Guo X, Sturdevant DE, Virtaneva K, Hernandez MM, Houpt E, Sher A, Petri WA., Jr Effect of the leptin receptor Q223R polymorphism on the host transcriptome following infection of Entamoeba histolytica. Infect Immun. 2013;81:1460–1470. doi: 10.1128/IAI.01383-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mengheri E. Health, probiotics, and inflammation. J Clin Gastroenterol. 2008;42(Suppl 3)(pt 2):S177–S178. doi: 10.1097/MCG.0b013e31817eedc4. [DOI] [PubMed] [Google Scholar]
- Netea MG, Ferwerda G, de Jong DJ, Jansen T, Jacobs L, Kramer M, Naber TH, Drenth JP, Girardin SE, Kullberg BJ, Adema GJ, Van der Meer JW. Nucleotide-binding oligomerization domain-2 modulates specific TLR pathways for the induction of cytokine release. J Immunol. 2005;174:6518–6523. doi: 10.4049/jimmunol.174.10.6518. [DOI] [PubMed] [Google Scholar]
- Netea MG, Quintin J, van der Meer JWM. Trained immunity: a memory for innate host defense. Cell Host Microbe. 2011;9:355–361. doi: 10.1016/j.chom.2011.04.006. [DOI] [PubMed] [Google Scholar]
- Nguyen-Van-Tam JS, Venkatesan S, Muthuri SG, Myles PR. Neuraminidase inhibitors: who, when, where. Clin Microbiol Infect. 2015;21:222–225. doi: 10.1016/j.cmi.2014.11.020. [DOI] [PubMed] [Google Scholar]
- Ozen M, Sandal GK, Dinleyici EC. Probiotics for the prevention of pediatric upper respiratory infections: a systematic review. Expert Opin Biol Ther. 2015;15:9–20. doi: 10.1517/14712598.2015.980233. [DOI] [PubMed] [Google Scholar]
- Park MK, Ngo V, Kwon YM, Lee YT, Yoo S, Cho YH, Hong SM, Hwang HS, Ko EJ, Jung YJ, Moon DW, Jeong EJ, Kim MC, Lee YN, Jang JH, Oh JS, Kim CH, Kang SM. Lactobacillus plantarum DK119 as a probiotic confers protection against influenza virus by modulating innate immunity. PLoS ONE. 2013;9:e75368. doi: 10.1371/journal.pone.0075368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pavot V, Rochereau N, Resseguier J, Gutjahr A, Genin C, Tiraby G, Perouzel E, Lioux T, Vernejoul F, Verrier B, Paul S. Cutting edge: new chimeric NOD2/TLR2 adjuvant drastically increases vaccine immunogenicity. J Immunol. 2014;193:5781–5785. doi: 10.4049/jimmunol.1402184. [DOI] [PubMed] [Google Scholar]
- Percopo CM, Dyer KD, Garcia-Crespo KE, Gabryszewski SJ, Shaffer AL, 3rd, Domachowske JB, Rosenberg HF. B cells are not essential for Lactobacillus-mediated protection against lethal pneumovirus infection. J Immunol. 2014a;192:5265–5272. doi: 10.4049/jimmunol.1400087. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Percopo CM, Dyer KD, Karpe KA, Domachowske JB, Rosenberg HF. Chapter 23: eosinophils and respiratory virus infection: a qRT-PCR dual-standard curve method for determining virus recovery from mouse lung tissue. In: Walsh GA, editor. Methods in Molecular Biology. Humana Press; 2014b. [DOI] [PubMed] [Google Scholar]
- Plant H, Stacey C, Tiong-Yip C-L, Walsh J, Yu Q, Rich K. High-throughput hit screening cascade to identify respiratory syncytial virus (RSV) inhibitors. J Biomol Screen. 2015 doi: 10.1177/1087057115569428. in press. [DOI] [PubMed] [Google Scholar]
- Rabinovich GA, Toscano MA. Turning “sweet” on immunity: galectin-glycan interactions in immune tolerance and inflammation. Nat Rev Immunol. 2009;9:338–352. doi: 10.1038/nri2536. [DOI] [PubMed] [Google Scholar]
- Read CB, Kuijper JL, Hjorth SA, Heipel MD, Tang X, Fleetwood AJ, Dantzler JL, Grell SN, Kastrup J, Wang C, Brandt CS, Hansen AJ, Wagtmann NR, Xu W, Stennicke VW. Cutting edge: identification of neutrophil PGLYRP1 as a ligand for TREM-1. J Immunol. 2015;194:1417–1421. doi: 10.4049/jimmunol.1402303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rosenberg HF, Domachowske JB. Inflammatory responses to respiratory syncytial virus (RSV) infection and development of immunomodulatory pharmacotherapeutics. Curr Med Chem. 2012;19:1424–1431. doi: 10.2174/092986712799828346. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rossi DL, Hurst SD, Xu Y, Wang W, Menon S, Coffman RL, Zlotnik A. Lungkine, a novel CXC chemokine specifically expressed by lung bronchoepithelial cells. J Immunol. 1999;162:5490–5497. [PubMed] [Google Scholar]
- Royet J, Gupta D, Dziarski R. Peptidoglycan recognition proteins: modulators of the microbiome and inflammation. Nat Rev Immunol. 2011;11:837–851. doi: 10.1038/nri3089. [DOI] [PubMed] [Google Scholar]
- Sandri S, Rodriguez D, Gomes E, Monteiro HP, Russo M, Campa A. Is serum amyloid A an endogenous TLR2 agonist? J Leukoc Biol. 2008;83:1174–1180. doi: 10.1189/jlb.0407203. [DOI] [PubMed] [Google Scholar]
- Saraiva M, O’Garra A. The regulation of IL-10 production by immune cells. Nat Rev Immunol. 2010;10:170–181. doi: 10.1038/nri2711. [DOI] [PubMed] [Google Scholar]
- Schwarze J, Mackenzie KJ. Novel insights into immune and inflammatory responses to respiratory viruses. Thorax. 2013;68:108–110. doi: 10.1136/thoraxjnl-2012-202291. [DOI] [PubMed] [Google Scholar]
- Sow FB, Gallup JM, Krishnan S, Patera AC, Suzich J, Ackermann MR. Respiratory syncytial virus infection is associated with an altered innate immunity and a heightened pro-inflammatory response in the lungs of preterm lambs. Respir Res. 2011;12:106. doi: 10.1186/1465-9921-12-106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tabarani CM, Bonville CA, Suryadevara M, Branigan P, Wang D, Huang D, Rosenberg HF, Domachowske JB. Novel inflammatory markers, clinical risk factors, and virus type associated with severe respiratory syncytial virus infection. Pediatr Infect Dis J. 2013;32:e437–e442. doi: 10.1097/INF.0b013e3182a14407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tomasada Y, Chiba E, Zelaya H, Takahashi T, Tsykida K, Kitazawa H, Alvarez S, Villena J. Nasally administered Lactobacillus rhamnosus strains differentially modulate respiratory antiviral immune responses and induce protection against respiratory syncytial virus infection. BMC Immunol. 2013;14:40. doi: 10.1186/1471-2172-14-40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Uehara A, Fujimoto Y, Fukase K, Takada H. Various human epithelial cells express functional Toll-like receptors, NOD1 and NOD2 to produce anti-microbial peptides, but not proinflammatory cytokines. Mol Immunol. 2007;44:3100–3111. doi: 10.1016/j.molimm.2007.02.007. [DOI] [PubMed] [Google Scholar]
- Wang X, Zhang Z, Louboutin JP, Moser C, Weiner DJ, Wilson JM. Airway epithelia regulate expression of human beta-defensin 2 through Toll-like receptor 2. FASEB J. 2003;17:1727–1729. doi: 10.1096/fj.02-0616fje. [DOI] [PubMed] [Google Scholar]
- Watanabe T, Kitani A, Murray PJ, Wakatsuki Y, Fuss IJ, Strober WJ. Nucleotide binding oligomerization domain 2 deficiency leads to dysregulated TLR2 signaling and induction of antigen-specific colitis. Immunity. 2006;25:473–485. doi: 10.1016/j.immuni.2006.06.018. [DOI] [PubMed] [Google Scholar]
- Welliver RC., Sr The immune response to respiratory syncytial virus infection: friend or foe? Clin Rev Allergy Immunol. 2008;34:163–173. doi: 10.1007/s12016-007-8033-2. [DOI] [PubMed] [Google Scholar]
- Wensink AC, Hack CE, Bovenschen N. Granzymes regulate proinflammatory cytokine responses. J Immunol. 2015;194:491–497. doi: 10.4049/jimmunol.1401214. [DOI] [PubMed] [Google Scholar]
- Wissinger E, Goulding J, Hussell T. Immune homeostasis in the respiratory tract and its impact on heterologous infection. Semin Immunol. 2009;21:147–155. doi: 10.1016/j.smim.2009.01.005. [DOI] [PubMed] [Google Scholar]
- Wu C, Thalhamer T, Franca RF, Xiao S, Wang C, Hotta C, Zhu C, Hirashima M, Anderson AC, Kuchroo VK. Galectin-9-CD44 interaction enhances stability and function of adaptive regulatory T cells. Immunity. 2014;41:270–282. doi: 10.1016/j.immuni.2014.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu J, Zhang Y, Xin Z, Wu X. The crosstalk between TLR2 and NOD2 in Aspergillus fumigatus keratitis. Mol Immunol. 2015;64:235–243. doi: 10.1016/j.molimm.2014.11.021. [DOI] [PubMed] [Google Scholar]
- Yoo JK, Kim TS, Hufford MM, Braciale TJ. Viral infection of the lung: host response and sequelae. J Allergy Clin Immunol. 2013;132:1263–1276. doi: 10.1016/j.jaci.2013.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zeuthen LH, Fink LN, Frokiaer H. Toll-like receptor 2 and nucleotide-binding oligomerization domain-2 play divergent roles in the recognition of gut-derived lactobacilli and bifidobacteria in dendritic cells. Immunology. 2008;124:489–502. doi: 10.1111/j.1365-2567.2007.02800.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.