Rotavirus (RV) causes significant morbidity and mortality in children worldwide. The intestinal microbiota plays an important role in modulating host–pathogen interactions, but little is known about the impact of commonly used probiotics on human RV (HRV) infection. In this study, we compared the immunomodulatory effects of Gram-positive (Lactobacillus rhamnosus strain GG [LGG]) and Gram-negative (Escherichia coli Nissle [EcN]) probiotic bacteria on virulent human rotavirus (VirHRV) infection and immunity using neonatal gnotobiotic piglets. Gnotobiotic piglets were colonized with EcN, LGG, or EcN+LGG or uncolonized and challenged with VirHRV. Mean peak virus shedding titers and mean cumulative fecal scores were significantly lower in EcN-colonized compared with LGG-colonized or uncolonized piglets. Reduced viral shedding titers were correlated with significantly reduced small intestinal HRV IgA Ab responses in EcN-colonized compared with uncolonized piglets post-VirHRV challenge. However the total IgA levels post-VirHRV challenge in the intestine and pre-VirHRV challenge in serum were significantly higher in EcN-colonized than in LGG-colonized piglets. In vitro treatment of mononuclear cells with these probiotics demonstrated that EcN, but not LGG, induced IL-6, IL-10, and IgA, with the latter partially dependent on IL-10. However, addition of exogenous recombinant porcine IL-10 + IL-6 to mononuclear cells cocultured with LGG significantly enhanced IgA responses. The greater effectiveness of EcN in moderating HRV infection may also be explained by the binding of EcN but not LGG to Wa HRV particles or HRV 2/4/6 virus-like particles but not 2/6 virus-like particles. Results suggest that EcN and LGG differentially modulate RV infection and B cell responses.

Rotavirus (RV) is a leading cause of diarrhea. It causes an estimated 480,000 deaths in children <5 years of age in developing countries (1). The efficacy of the available RV vaccines is low in developing countries compared with developed countries (2). Many factors, such as malnutrition, micronutrient deficiencies, and breastfeeding (35), are implicated in the lower efficacy of enteric vaccines in impoverished countries. In addition to the aforementioned factors, recent studies have also shown a role for the intestinal microbiota in modulating enteric viral infections and oral vaccine responses (6, 7). Ablation of the intestinal microbiota reduced the severity of RV infection and modulated RV-induced adaptive immunity in mice (8). A higher abundance of Clostridiales, Enterobacteriales, and Pseudomonadales was associated with poor oral poliovirus vaccine responses in infants, whereas higher bifidobacteria abundance was positively correlated with greater oral poliovirus vaccine-specific T cell and Ab responses (9). Previous studies also showed a direct role of commensals in enhancing enteric viral infections, including poliovirus (10) and mouse mammary tumor virus (11) infections. Thus, the composition of the microbiota or certain members of commensal microbial communities play a significant role in modulating viral infections and host immunity to pathogens and vaccines.

Probiotics are increasingly used to enhance oral vaccine responses and to treat some enteric infections (12) as well as various inflammatory diseases of the gastrointestinal (GI) tract in children (13). Among probiotics, Gram-positive (G+) probiotics such as Lactobacillus spp or Bifidobacteria spp have been administrated in randomized human clinical trials (14, 15) and experimental studies (1619) to reduce the severity of RV-induced diarrhea. Among G+ probiotics, Lactobacillus rhamnosus strain GG (LGG) has been extensively investigated for its beneficial health effects such as shortening the duration of human RV (HRV) diarrhea and enhancing HRV-specific immune responses in children (15, 20). However, mechanisms of action of LGG on HRV infection and whether LGG has any superior probiotic effects on HRV infection and immunity compared with a G- probiotic such as Escherichia coli Nissle (EcN) are largely unknown. G+ and G-negative (G−) probiotics/commensals differ in microbe-associated molecular patterns and cell wall constituents, which may differentially influence neonatal immune maturation and susceptibility to HRV infections. In addition, E. coli is one of the first species to colonize newborn babies (21). EcN is widely used to treat inflammatory disorders such as ulcerative colitis in humans (22). Beneficial effects of EcN are mediated through enhancing intestinal barrier function (23) and moderating inflammatory disorders (24). Furthermore, similar to other probiotics, EcN has antimicrobial and immunomodulatory properties, such as inhibition of pathogenic bacterial invasion of epithelial cells (25), induction of β-defensin in epithelial cells (26), and modulation of T cell proliferation (27). However, the role of EcN in the maturation of Ab responses, EcN direct effects on HRV pathogenesis, and comparative effects of G+ and G− probiotics on HRV infection and immunity are unknown.

Gnotobiotic (Gn) piglets are an ideal model to delineate the direct beneficial effects of probiotics on enteric viral infections and virus-induced B cell responses. For instance, Gn piglets are susceptible to HRV diarrhea (28). Furthermore, piglets receive no Abs in utero because of the epitheliochorial placenta of the sow, eliminating the maternal Ab influence on neonatal immune responses (29). In addition, fetal as well as newborn piglets have a functional, although immature, immune system, and they are capable of mounting immune responses to environmental Ags, commensal microorganisms and pathogens (30, 31). In this study, we compared the effects of G− EcN and G+ LGG probiotics on virulent HRV (VirHRV) infection and B cell responses in the Gn piglet model.

For colonization of the piglets with the probiotics, LGG ATCC 53103 (American Type Culture Collection, Manassas, VA) was prepared as described previously (17). EcN (supplied by Dr. U. Sonnenborn, Department of Biological Research, Ardeypharm, Herdecke, Germany) was streaked onto Luria–Bertani (LB) agar plates and incubated overnight at 37°C. Subsequently, a single colony from the streaked plate was picked and inoculated into 6 ml LB broth. After overnight incubation, the number of EcN colony forming units (CFU) in the suspension was estimated by spectrophotometry at 600 nm, based on comparison with predetermined standard growth curve.

All animal experiments were approved by the Institutional Animal Care and Use Committee at Ohio State University. Near-term sows (Landrace × Yorkshire × Duroc cross-bred) were purchased from the Ohio State University swine center facility. Cesarean-derived Gn piglets from the sows were maintained in sterile isolators as described previously (32). Prior to probiotic colonization, sterility was verified by aerobic and anaerobic culturing of rectal swabs collected from the piglets. Subsequently, these piglets were assigned to one of the following four groups: 1) EcN colonized, 2) LGG colonized, 3) EcN and LGG dual-colonized (EcN+LGG), and 4) uncolonized piglets. For monocolonization of probiotics, 6-d-old piglets were colonized with 105 CFU/pig, and for dual colonization of EcN and LGG probiotics, piglets were colonized with 1:1 ratio of EcN and LGG at a total of 105 CFU/pig. Rectal swabs were collected weekly to assess the colonization status of each probiotics in the piglets as described previously (33). Total viable counts of LGG were determined using deMan, Rogosa, and Sharpe agar, and EcN were enumerated after plating on LB agar. Subsequently all piglets were challenged with VirHRV (1 × 106 fluorescent focus units). Post-VirHRV challenge, rectal swabs were collected to assess HRV shedding. Fecal scores were recorded as previously described (34), and the mean cumulative fecal score was calculated as (daily fecal scores from postchallenge days [PCD] 1–7)/n to assess the severity of diarrhea (35). Serum samples were collected to assess the impact of probiotics and VirHRV challenge on total Ig responses. All piglets were euthanized at postbacterial colonization day (PBCD) 36/post-HRV challenge day (PCD) 21 and blood, duodenum, ileum, and spleen were collected to isolate mononuclear cells (MNCs) as described previously (Fig. 1A) (17). To determine the intestinal Ab responses, small intestinal contents were collected, and protease inhibitors were incorporated.

FIGURE 1.

(A) Schematic diagram of the experimental design showing time points for probiotic colonization, VirHRV Wa challenge, and euthanasia. (B) Fecal probiotic bacterial shedding from probiotic colonized piglets. Different letters indicate significant differences (p < 0.05) at the same time point in fecal probiotics counts among treatment groups, whereas the same letters indicate no significant difference. The values that are statistically similar are in the same box. (C) Probiotic colonization levels at different segments of the intestinal tract. Different letters indicate significant differences (p < 0.05) at the same tissue in probiotics counts among treatment groups, whereas the same letters indicate no significant difference.

FIGURE 1.

(A) Schematic diagram of the experimental design showing time points for probiotic colonization, VirHRV Wa challenge, and euthanasia. (B) Fecal probiotic bacterial shedding from probiotic colonized piglets. Different letters indicate significant differences (p < 0.05) at the same time point in fecal probiotics counts among treatment groups, whereas the same letters indicate no significant difference. The values that are statistically similar are in the same box. (C) Probiotic colonization levels at different segments of the intestinal tract. Different letters indicate significant differences (p < 0.05) at the same tissue in probiotics counts among treatment groups, whereas the same letters indicate no significant difference.

Close modal

A cell culture immunofluorescence assay was used to detect shedding of HRV in rectal swab fluids as described previously (34). Ab-secreting cells (ASCs) in duodenal MNCs were determined as described previously (36). HRV-specific Ab responses and total Ig levels were determined by ELISA (37). Flow cytometry staining to identify CD79β+CD21CD2 B cells was performed as described previously (18, 38).

Bacterial specific Ab responses were determined as previously described with some modifications (39, 40). EcN and LGG were prepared as previously described for probiotic colonization. Wells of a 96-well ELISA plate were coated with 100 μl 1 × 107 bacteria diluted in 0.05 M bicarbonate buffer and incubated at 4°C overnight. After overnight incubation, bacteria were fixed to the plates by adding 100 μl of 80% acetone per well for 10 min. Plates were washed twice with PBS-0.05% Tween 20, and then, 200 μl of a 4% nonfat milk solution was added to each well and incubated at 37°C for 2 h. Following plate washing, diluted serum samples were added and incubated at 37°C for 1 h. Plates were washed five times and HRP-conjugated goat anti-pig IgA Ab (0.3 μg/ml; AbD Serotec), or HRP-conjugated goat anti-pig IgG Ab (catalog number 04-14-02; KPL) was added and incubated at 37°C for 1 h. Last, after washing, tetramethylbenzidine substrate (KPL) was added to each well, and the reaction was stopped with 1 M phosphoric acid.

The effects of probiotics on HRV binding to the human colonic epithelial (Caco-2) cells were assessed as previously described using a cell–virus binding assay with modifications (41). Briefly, Caco-2 cells were maintained in DMEM: nutrient mixture F-12 (DMEM/F-12) medium (Thermo Fisher Scientific) containing 15% FBS at 37°C in the presence of 5% CO2. Confluent Caco-2 cell monolayers in 6-well cell culture plates (1 × 106 cells) were washed once with MEM. Subsequently, cells were incubated with individual probiotic bacteria at a cell:bacteria ratio of 1:100 at 37°C for 4 h with gentle rocking. In the presence of bacteria, cells were cooled on ice for 10 min. Subsequently, HRV, at a multiplicity of infection 3.0, was added to the cells–bacterial mixture and then incubated at 4°C for 1 h with gentle rocking. After this incubation, cells were washed twice with cold MEM. After adding cold MEM, cells were subjected to two rounds of freeze-thaw to release bound virus (42), and concentrations of recovered virus in collected samples were determined by ELISA. Probiotics adhesion to Caco-2 cells was performed as described previously (43). Briefly, cells were incubated with individual probiotic bacteria at a cell:bacteria ratio of 1:10 at 37°C for 90 min and 4 h. After washing three times, cells were detached from wells by treating with 0.05% trypsin-EDTA (Thermo Fisher Scientific) for 20 min. Subsequently, serially diluted suspensions were plated on agar plates. and bacteria adhered to the Caco-2 cells were quantified.

Direct binding of each probiotic to HRV was assessed by flow cytometry under in vitro conditions. Probiotic bacteria were first stained with green fluorescent nucleic acid dye SYTO 9 at 5 μM concentration (Life Technologies) (44). Subsequently, stained bacteria were incubated with either semipurified HRV (45) or Alexa Fluor 647 (Life Technologies)–conjugated human rotavirus like particles (VLP) (46) at 37°C for 1.5 h. Subsequently, bacteria were washed three times using sterile PBS to remove unbound virus and VLPs. To detect bacterial bound HRV, bacteria were incubated with Alexa Fluor 647–conjugated anti-RV mAb (clone RG23B9C5H11) or isotype control at 4°C for 45 min. After washing, bacteria were acquired by a BD Accuri C6 flow cytometer, and data were analyzed using the instrument software.

Splenic and ileal MNCs were cultured with EcN and LGG at 1:10 ratio (MNC:bacteria) for 24 h as described previously (18). Culture supernatants were collected to quantify IL-6 and IL-10 cytokines as described previously (17). To assess the effects of IL-6 and IL-10 on IgA responses, splenic MNCs were cocultured with heat-killed EcN and LGG in the presence or absence of blocking Abs to IL-6 (10 μg/ml; R&D Systems) and IL-10 (10 μg/ml; R&D Systems) or recombinant porcine IL-6 (10 μg/ml; R&D Systems) and IL-10 (10 μg/ml; R&D Systems) cytokines. After 7 d, culture supernatants were collected to quantify total IgA levels by ELISA.

Mean days to onset of virus shedding, mean duration of virus shedding, average peak virus shedding titer and mean duration of diarrhea, log-transformed Ab titers, total Ig levels, and ASC were analyzed by Kruskal–Wallis rank-sum test. Mean cumulative fecal scores were analyzed by the area under the curve as described previously (17). Statistical analyzes were performed by using GraphPad Prism 5 software (Graph Pad Software).

Colonization of EcN, LGG, and EcN+LGG in the Gn piglets was determined by quantifying CFUs in fecal samples weekly and in different sections of the GI tract at the end of the study (PBCD36/PCD21). Except at PBCD22/PCD7, no significant differences in fecal bacterial CFUs were observed between EcN and LGG piglets (Fig. 1B). In EcN+LGG piglets, fecal LGG counts were consistently lower compared with EcN CFU counts, which could be due to interference of EcN on the colonization of LGG in the EcN+LGG dually colonized piglets. Relative comparisons of LGG and EcN colonization in EcN+LGG piglets revealed a significant reduction in LGG compared with EcN in the duodenum, jejunum, ileum, cecum, and colon. In the probiotic colonized piglets, VirHRV infection had no significant effect on fecal bacterial CFU counts at all post-VirHRV challenge times assessed as compared with pre-VirHRV challenge (PBCD15/PCD0). Enumeration of each probiotic in different sections of the GI tract revealed that LGG and EcN colonized all sections of the GI tract of each monocolonized piglet examined at PBCD36/PCD21 (Fig. 1C). Translocation of probiotics to the mesenteric lymph nodes, spleen, and liver was observed in all the probiotic colonized piglets, but no septicemia or clinical signs of bacteremia were observed in the piglets.

The effects of LGG, EcN and LGG+EcN colonization on VirHRV infection were assessed. Fecal virus shedding was confirmed for all VirHRV challenged pigs. However, mean peak virus shedding titers and mean cumulative fecal scores were significantly lower in EcN compared with LGG-colonized or uncolonized piglets (Table I). Furthermore, LGG-colonized piglets had similar mean peak virus shedding titers as in the uncolonized piglets. Furthermore, EcN+LGG cocolonized piglets had 3-fold lower average peak virus shedding titers and significantly reduced cumulative mean fecal scores compared with uncolonized piglets. In addition, the mean duration of diarrhea was 3.4 d shorter in EcN+LGG-colonized compared with uncolonized piglets.

Table I.
Summary of HRV shedding and diarrhea scores in the four different treatment groups
TreatmentnVirus Sheddinga
Diarrheab
% ShedMean Duration DayscAvg Peak Titer Shedc (FFU/ml)% with DiarrheaMean Duration DayscMean Cumulative Fecal Scorec,d
EcN+VirHRV 100 3.6 0.95 × 104 (e) 66 1.5 (e) 8.2 (e) 
LGG+VirHRV 100 4.5 6.8 × 104 (f) 100 3.6 (e, f) 11.0 (f, g) 
EcN+LGG+VirHRV 100 2.8 1.9 × 104 (e, f) 71 1.4 (e) 8.3 (e, g) 
Control+VirHRV 100 4.2 7.0 × 104 (f) 100 4.8 (f) 12.6 (f) 
TreatmentnVirus Sheddinga
Diarrheab
% ShedMean Duration DayscAvg Peak Titer Shedc (FFU/ml)% with DiarrheaMean Duration DayscMean Cumulative Fecal Scorec,d
EcN+VirHRV 100 3.6 0.95 × 104 (e) 66 1.5 (e) 8.2 (e) 
LGG+VirHRV 100 4.5 6.8 × 104 (f) 100 3.6 (e, f) 11.0 (f, g) 
EcN+LGG+VirHRV 100 2.8 1.9 × 104 (e, f) 71 1.4 (e) 8.3 (e, g) 
Control+VirHRV 100 4.2 7.0 × 104 (f) 100 4.8 (f) 12.6 (f) 
a

Determined by cell culture immunofluorescence assay.

b

Pigs with fecal score > 1 were considered diarrheic. Fecal consistency was scored as follows: 0, normal; 1, pasty/semiliquid; and 2, liquid.

c

Means with different letters (e–g) in the same column differ significantly (determined by Kruskal–Wallis rank sum test, p ≤ 0.05).

d

Area under the curve indicates diarrhea severity.

FFU, fluorescent-forming unit.

The effects of selected probiotics on HRV-specific Ab responses were assessed. Consistent with decreased fecal HRV shedding and diarrhea, EcN+VirHRV piglets had significantly lower small intestinal IgA HRV Ab titers (Fig. 2A) and duodenal IgA and IgG HRV ASCs (Fig. 2B, 2C) as compared with uncolonized piglets at PBCD36/PCD21. The reduced HRV-specific Ab titers were correlated with the reduced virus shedding titers (data not shown). Regardless of similar mean peak virus shedding titers between LGG- and uncolonized-piglets, intestinal HRV specific IgA and IgG Ab responses were significantly lower in LGG-colonized compared with uncolonized-piglets post-VirHRV challenge (Fig 2). Intestinal virus-specific IgA Ab responses were comparable between the EcN+LGG+VirHRV and uncolonized+VirHRV groups regardless of the reduced virus shedding titers and a significant reduction in mean fecal scores in the former group compared with the later one.

FIGURE 2.

EcN and LGG probiotic colonization significantly modulated the small intestinal HRV-specific Ab responses at the post-VirHRV challenge. Geometric mean titers (GMT) of HRV-specific-IgA Ab titers in intestinal contents (A) and HRV-specific IgA IgG ASCs responses in duodenum (B and C) of Gn pigs challenged with or without probiotic colonization at post-VirHRV challenge (PBCD36/PCD21) (one-way ANOVA followed by Duncan’s multiple range test on log10 transferred titers, *p < 0.05). Error bars indicate SEM.

FIGURE 2.

EcN and LGG probiotic colonization significantly modulated the small intestinal HRV-specific Ab responses at the post-VirHRV challenge. Geometric mean titers (GMT) of HRV-specific-IgA Ab titers in intestinal contents (A) and HRV-specific IgA IgG ASCs responses in duodenum (B and C) of Gn pigs challenged with or without probiotic colonization at post-VirHRV challenge (PBCD36/PCD21) (one-way ANOVA followed by Duncan’s multiple range test on log10 transferred titers, *p < 0.05). Error bars indicate SEM.

Close modal

The impact of probiotic colonization and VirHRV infection on intestinal B cell responses was assessed. Small intestinal total IgA levels (Fig. 3A) and total IgA-secreting cells (Fig. 3B) were significantly higher in EcN piglets compared with LGG-colonized piglets post-VirHRV challenge. Furthermore, the comparable intestinal total IgA responses between EcN ± LGG+VirHRV and VirHRV groups also indicate the immunostimulatory effects of HRV on the B cell responses (Fig. 3A, 3B).

FIGURE 3.

EcN enhanced small intestinal total IgA responses at PBCD36/PCD21. Total IgA levels in the small intestinal contents (A) and total IgA Ig secreting cell responses in the duodenum (B) of the Gn pigs challenged with or without probiotic colonization at post-HRV challenge (PBCD36/PCD21) (one-way ANOVA followed by Duncan’s multiple range test on log10 transferred titers, *p < 0.05). Error bars indicate SEM.

FIGURE 3.

EcN enhanced small intestinal total IgA responses at PBCD36/PCD21. Total IgA levels in the small intestinal contents (A) and total IgA Ig secreting cell responses in the duodenum (B) of the Gn pigs challenged with or without probiotic colonization at post-HRV challenge (PBCD36/PCD21) (one-way ANOVA followed by Duncan’s multiple range test on log10 transferred titers, *p < 0.05). Error bars indicate SEM.

Close modal

The effect of EcN and LGG monocolonization on serum total IgA and IgG responses was also assessed. No differences in total serum IgA or IgG levels were observed in any group at PBCD7 (Fig. 4). However, the EcN and EcN+LGG piglets had significantly higher total serum IgA (Fig. 4A) and IgG (Fig. 4B) levels compared with uncolonized or LGG-monocolonized piglets at PBCD15/PCD0. Furthermore, significantly higher total IgA responses were also observed in EcN+LGG piglets compared with the other three groups at PBCD29/PCD14. This indicates a positive interaction among EcN, LGG, and VirHRV in inducing total serum IgA responses. Furthermore, induction of EcN- but not LGG-specific IgA Ab responses may also have partially contributed to the higher total IgA Ab responses in EcN and EcN+LGG piglets (Supplemental Fig. 1). In addition, significant activation of B cells by EcN was also evident in our study in which higher frequencies of Ab-forming B cells (CD79β+CD21CD2 B cells) was observed in EcN and EcN+LGG compared with LGG alone–treated MNCs in vitro (Supplemental Fig. 2). Notably, total IgG responses were similar between LGG and uncolonized piglets at pre- and post-VirHRV challenge time points. This suggests that the LGG colonization alone was not sufficient to induce markedly increased systemic total IgG responses. In uncolonized piglets, VirHRV challenge increased total IgA responses, but not total IgG responses, as evident by higher IgA responses at various post-VirHRV challenge days compared with pre-VirHRV challenge (PBCD15/PCD0).

FIGURE 4.

EcN ± LGG colonization significantly increased serum total IgA and IgG responses pre-VirHRV challenge (PBCD15/PCD0). (A and B) Serum total IgA and IgG levels were measured by ELISA at indicated time points. Different letters indicate significant differences (p < 0.05) at the same time point in total Ig levels among treatment groups, whereas the same letters indicate no significant difference. The values that are statistically similar are in the same box (one-way ANOVA followed by Duncan’s multiple range test, p < 0.05). Error bars indicate SEM.

FIGURE 4.

EcN ± LGG colonization significantly increased serum total IgA and IgG responses pre-VirHRV challenge (PBCD15/PCD0). (A and B) Serum total IgA and IgG levels were measured by ELISA at indicated time points. Different letters indicate significant differences (p < 0.05) at the same time point in total Ig levels among treatment groups, whereas the same letters indicate no significant difference. The values that are statistically similar are in the same box (one-way ANOVA followed by Duncan’s multiple range test, p < 0.05). Error bars indicate SEM.

Close modal

EcN or EcN+LGG but not LGG colonization significantly enhanced systemic as well as intestinal total IgA responses pre-VirHRV challenge. This might be due to the EcN probiotic–dependent stimulation of IgA-inducing cytokines such as IL-6 and IL-10 as we assessed by coculturing MNCs with the live probiotics. Significantly higher IL-6 and IL-10 levels were observed in EcN and EcN+LGG compared with LGG-treated MNCs (Fig. 5A).

FIGURE 5.

(A) Effect of EcN and LGG on IL-6 and IL-10 cytokine responses in splenic and ileal MNCS under in vitro condition. Splenic and ileal MNCs from control Gn piglets were cocultured with indicated probiotics for 24 h at 37°C, and supernatants were collected for IL-6 and IL-10 determination by ELISA. Different alphabetical letters above the bars indicate significant differences (p < 0.05) among treatment groups, whereas the same letters indicate no significant difference. (B) Blocking Abs to IL-6 and IL-10 were added to MNC+probiotic coculture to determine their effects on total IgA responses. (C) Effect of recombinant porcine IL-6 and IL-10 on total IgA responses in MNCs cocultured with the probiotics. Different letters above the bars indicate significant differences (p < 0.05) among treatment groups within each probiotic treatment, whereas the same letters indicate no significant difference. Results are mean ± SEM (n = 4, one-way ANOVA, *p < 0.05).

FIGURE 5.

(A) Effect of EcN and LGG on IL-6 and IL-10 cytokine responses in splenic and ileal MNCS under in vitro condition. Splenic and ileal MNCs from control Gn piglets were cocultured with indicated probiotics for 24 h at 37°C, and supernatants were collected for IL-6 and IL-10 determination by ELISA. Different alphabetical letters above the bars indicate significant differences (p < 0.05) among treatment groups, whereas the same letters indicate no significant difference. (B) Blocking Abs to IL-6 and IL-10 were added to MNC+probiotic coculture to determine their effects on total IgA responses. (C) Effect of recombinant porcine IL-6 and IL-10 on total IgA responses in MNCs cocultured with the probiotics. Different letters above the bars indicate significant differences (p < 0.05) among treatment groups within each probiotic treatment, whereas the same letters indicate no significant difference. Results are mean ± SEM (n = 4, one-way ANOVA, *p < 0.05).

Close modal

Treatment of MNCs with probiotics for 7 d revealed that total IgA responses were higher in culture supernatants from EcN ± LGG compared with LGG alone treated MNCs (Fig. 5B). Significantly higher levels of total IgA were observed in culture supernatants from EcN treated MNCs compared with that of LGG treated MNCs. The addition of porcine IL-10 Ab, but not porcine IL-6 Ab, significantly reduced total IgA levels (Fig. 5B). Thus, it appears that IL-10 plays a significant role in the EcN-induced IgA responses. We also examined whether the addition of recombinant porcine IL-10, IL-6, and IL-10+IL-6 in MNC cocultured with LGG had any enhancing effect on total IgA responses. The addition of IL-10 and IL-6+IL-10 cytokines induced higher and significantly higher total IgA levels, respectively, in the cytokine-treated MNC+LGG cocultures compared with LGG alone–treated MNCs (Fig. 5C).

The observed variable effects of probiotics on virus shedding and diarrhea severity might be caused by a difference in their interference with virus attachment to epithelial cells. Preincubation of Caco-2 epithelial cells with EcN, but not with LGG, significantly reduced (∼50% reduction) cellular attachment of HRV (Fig. 6A). Furthermore, a significantly higher percentage of EcN binds to Caco-2 cells compared with a negligible percentage of LGG that binds to Caco-2 cells (Fig. 6B). These results suggest that EcN potentially directly interferes with HRV attachment to target (epithelial) cells, subsequently resulting in lower virus shedding and diarrhea.

FIGURE 6.

(A) Confluent Caco-2 cell monolayers in 6-well cell culture plates were incubated with individual probiotic bacteria at 37°C for 4 h. In the presence of bacteria, HRV at a multiplicity of infection 3.0 was added to the cells–bacterial mixture and then incubated at 4°C for 1 h. After washing, cells were subjected to two rounds of freeze-thaw to release bound virus, and concentrations of virus in collected samples were determined by ELISA. Results are expressed as mean percentage virus bound to probiotic treated Caco-2 cells relative to the no-probiotic–treated control cells (n = 4 independent experiments, one-way ANOVA, *p < 0.05). (B) Caco-2 cell monolayers were incubated with individual probiotic bacteria at 37°C for 90 min and 4 h. Subsequently, percent bacteria adhered to cells were quantified (n = 2 independent experiments, t test, *p < 0.05). (C) Analysis of Wa HRV binding to EcN and LGG. SYTO 9–stained probiotic bacteria were incubated with semipurified HRV. After washing, bacterial-bound HRV was detected by incubating with Alexa Fluor 647 (Life Technologies)–conjugated RV mAb (clone RG23B9C5H11). The percentage of virus bound to bacteria was assessed by flow cytometry. (D) Determination of rotavirus VLP 2/6/4 and VLP2/6 binding to EcN and LGG probiotics. SYTO 9-stained probiotic bacteria were incubated with Alexa Fluor 647–conjugated (Life Technologies) human rotavirus-like particles (VLP). After washing, the percentage of VLP bound to bacteria was assessed by flow cytometry. Results are mean ± SEM (n = 4, t test, *p < 0.05, ***p = 0.0004).

FIGURE 6.

(A) Confluent Caco-2 cell monolayers in 6-well cell culture plates were incubated with individual probiotic bacteria at 37°C for 4 h. In the presence of bacteria, HRV at a multiplicity of infection 3.0 was added to the cells–bacterial mixture and then incubated at 4°C for 1 h. After washing, cells were subjected to two rounds of freeze-thaw to release bound virus, and concentrations of virus in collected samples were determined by ELISA. Results are expressed as mean percentage virus bound to probiotic treated Caco-2 cells relative to the no-probiotic–treated control cells (n = 4 independent experiments, one-way ANOVA, *p < 0.05). (B) Caco-2 cell monolayers were incubated with individual probiotic bacteria at 37°C for 90 min and 4 h. Subsequently, percent bacteria adhered to cells were quantified (n = 2 independent experiments, t test, *p < 0.05). (C) Analysis of Wa HRV binding to EcN and LGG. SYTO 9–stained probiotic bacteria were incubated with semipurified HRV. After washing, bacterial-bound HRV was detected by incubating with Alexa Fluor 647 (Life Technologies)–conjugated RV mAb (clone RG23B9C5H11). The percentage of virus bound to bacteria was assessed by flow cytometry. (D) Determination of rotavirus VLP 2/6/4 and VLP2/6 binding to EcN and LGG probiotics. SYTO 9-stained probiotic bacteria were incubated with Alexa Fluor 647–conjugated (Life Technologies) human rotavirus-like particles (VLP). After washing, the percentage of VLP bound to bacteria was assessed by flow cytometry. Results are mean ± SEM (n = 4, t test, *p < 0.05, ***p = 0.0004).

Close modal

The reduced virus shedding and the significant reduction in fecal scores in EcN compared with LGG piglets may potentially be caused by direct binding of EcN to HRV. Analysis of the interaction between HRV and the probiotic bacteria revealed that a significantly higher percentage of EcN binds to HRV Wa strain compared with a negligible percentage of LGG that binds to HRV (Fig. 6C). Similarly, only EcN, but not LGG, binds to HRV VLP 2/4/6 particles. In comparison, the inner capsid VLP (VLP2/6) binding to EcN and LGG was very low or undetectable (Fig. 6D). Thus, VP4 of HRV might be mediating the interaction between Wa HRV and EcN.

The role of intestinal commensals in modulating viral infections has been increasingly recognized in recent studies (7). In this study, we compared anti-infectious and immunomodulatory effects of G+, G− probiotics, and their combination on RV infection and immunity. A significant protective effect against HRV infection was observed in EcN-colonized, compared with LGG-colonized or uncolonized piglets. EcN+LGG colonization resulted in 3-fold reduction in fecal virus shedding and induced marked B cell responses in the Gn piglets.

EcN colonization alone or cocolonization of EcN with LGG reduced fecal HRV shedding and the severity of diarrhea post-VirHRV challenge. The significant reduction in the intestinal and serum HRV-specific Ab responses in EcN-colonized piglets compared with control piglets was consistent with the reduction in fecal HRV shedding titers and diarrhea in the EcN group. The EcN-induced partial protection against HRV infection likely accounts for the significantly lower HRV-specific intestinal IgA Ab in the EcN piglets compared with the uncolonized group post-VirHRV challenge. A potential mechanism that might underlie EcN-induced partial protection against VirHRV infection is through the direct interference by EcN on HRV infection or indirectly through modulating host immunity. In support of the first possibility, our finding of HRV or VLP2/4/6 (but not VLP2/6) binding to EcN, but not to LGG, suggests that the partial protection conferred by EcN may be mediated by direct interaction between HRV and EcN. This observation is further supported by our in vitro virus–epithelial cell binding assay in which a significant reduction in virus attachment to epithelial cells was observed in EcN compared with LGG-treated cells. Furthermore, EcN had a higher capacity for adhesion to epithelial cells than LGG, which coincided with the EcN-induced reduction in virus attachment to Caco-2 cells in vitro. Rotavirus capsid is composed of three concentric layers, and among the viral proteins, VP2 is present in the internal core layer, VP6 is part of the intermediate layer, and VP4 is present in the form of spikes in the external (VP7) layer (47). Bacteria or bacterial derived ligands have been shown to interact with viruses and the interaction modulated viral pathogenesis (10, 11, 48). Furthermore, binding of EcN with VLP2/4/6 but not with VLP2/6 suggests that EcN mainly interacts with VP4, the major viral cell attachment protein of HRV (49). In addition, EcN may enhance innate immunity, which may also confer protective effects against VirHRV infection. A recent study showed that administration of flagellin, a TLR5 ligand derived from the flagella of bacteria, prevented RV infection via activation of innate immunity in a mouse model (50). Similar to the effect of EcN on HRV infection, EcN also inhibited the invasion of an intestinal cell line by several bacterial pathogens such as Salmonella enterica serovar Typhimurium, Yersinia enterocolitica, Shigella flexneri, Legionella pneumophila, and Listeria monocytogenes (25). Apart from the effects on pathogens, EcN also has anti-inflammatory properties as observed in treatment of T cells with EcN, which resulted in decreased secretion of inflammatory cytokines such as TNF-α and MCP-1 (51). In addition, our findings of reduced HRV diarrhea severity was also consistent with human clinical trials in which EcN treatment markedly reduced symptoms of infectious diarrhea in children and young infants (52, 53). Collectively, we showed that EcN had a significant protective effect on HRV shedding and diarrhea severity.

EcN and EcN+LGG colonization significantly increased serum total Ig responses in the Gn piglets pre-VirHRV challenge. Serum total IgA and IgG levels remained unchanged in uncolonized piglets pre-VirHRV challenge, which indicates a requirement of the intestinal microbes (including enteric viruses) for induction of B cell responses. The enhanced B cell responses in EcN-colonized piglets might be due to either direct activation the B cells by EcN or through stimulating APCs. Similar to our study, colonization of benign commensal E. coli (G58-1) alone in Gn piglets significantly enhanced serum total Ig levels (54). In another study, monoassociation of piglets with E. coli O83 resulted in increased frequency of dendritic cells and T cells in the small intestinal lamina propria (55). LPS of G− bacteria are a well-known B cell mitogen (56), and LPS also induces B cell maturation via the TLR4 signaling pathway (57). Furthermore, EcN supplementation significantly enhanced systemic total IgM and cellular proliferative responses in preterm infants (40). Although both EcN and EcN+LGG piglets had statistically similar mean peak virus shedding titers, HRV-specific IgA Ab responses were higher in EcN+LGG compared with EcN alone–colonized piglets. Potential synergistic immunostimulatory effects of microbe-associated molecular patterns from these two probiotics through multiple TLRs (58) and 2-fold higher virus shedding titers might have contributed to the higher HRV specific IgA responses in the EcN+LGG compared with EcN alone–colonized piglets. Thus, EcN has an immunostimulatory effect on both the systemic and intestinal immune systems.

LGG colonization had minimal effects on induction of total Ig responses in comparison with EcN. Specifically, serum total IgA and IgG levels were comparable between LGG colonized and uncolonized piglets at PBCD15/PCD0 (pre-VirHRV challenge). A similar trend was also observed for the intestinal HRV-specific Ab responses post-VirHRV challenge. Considering the comparable virus shedding titers between the LGG and uncolonized piglets, one would expect similar or higher small intestinal HRV-specific IgA Ab responses in LGG-colonized piglets compared with uncolonized piglets. Rather, we observed significantly lower HRV-specific intestinal and total IgA responses in LGG compared with uncolonized piglets post-VirHRV challenge. The mechanisms for lower induction of Ab responses in LGG-colonized piglets is unclear, but our in vitro studies indicated that LGG, but not EcN, failed to induce cytokines such as IL-10 and IL-6, which are well-known inducers of IgA Ab responses (5961). This observation is further supported by the significantly enhanced total IgA responses after addition of porcine IL-6 and IL-10 cytokines to the MNC–LGG cocultures. These findings are consistent with a previous study in which G+ lactobacilli bacteria failed to induce IL-6 and IL-10 cytokines, but E. coli induced higher levels of these cytokines in dendritic cells (62). Similarly, Hessle et al. (63) observed that various G+ commensal bacteria are less potent inducers of IL-10 compared with that of G− commensal bacteria. In an earlier study, we also observed a significant reduction in ileal IL-10 cytokine–secreting cell numbers in Lactobacillus acidophilus and Lactobacillus reuteri dual-colonized piglets compared with uncolonized piglets post-VirHRV infection (64). A previous study also showed that treatment of T cells with EcN conditioned medium upregulated expression of IL-10 (27). Thus, it appears that LGG alone may be a less potent inducer of IgA responses in comparison with EcN. Several earlier studies (14, 15) reported beneficial effects of LGG on RV infection and RV vaccine–induced immunity. One potential explanation for the discordance of present results from the previous studies (using conventional animal models or human subjects) is that LGG exert its beneficial effects through interaction with other members of the intestinal microbiota. In support this possibility, a recent study showed that LGG supplementation stabilized VirHRV-induced changes in the gut microbiota in Gn piglets transplanted with infant fecal microbiota (65). Furthermore, LGG supplementation significantly enhanced frequencies of CD4+IFNγ+ T cells but had no effect on HRV specific Ab responses in human microbiota colonized, HRV-vaccinated piglets compared with noncolonized, HRV-vaccinated Gn piglets (66). Apart from the effects on microbiota composition, LGG supplementation significantly altered functionality of resident intestinal commensals in human (67). Thus, we speculate that LGG might promote enhanced cellular or innate immunity in the presence of complex microbiota, which might have contributed the beneficial effects observed for LGG on HRV infections in children (15). In the absence of a complex microbiota in our Gn piglets, LGG may have only minimal beneficial effects on RV infection and immunity.

The differences in adherence properties of LGG and EcN in the intestinal microenvironment might also have contributed to the observed differential effects on immune responses. Our in vitro probiotics adherence assay to epithelial cells also suggests that there might be a variation in colonization properties of the probiotics which might have resulted in the observed differential immunomodulatory effects. We observed consistently reduced levels of LGG in EcN+LGG cocolonized compared with LGG alone–colonized piglets. Similarly, EcN introduction in the Gn mice colonized with E. coli K-12, Lactobacillus johnsonii, and Bifidobacterium longum resulted in complete elimination of E. coli K-12 and L. johnsonii (68). The mechanism mediating this EcN dominance over LGG is not clear. One potential explanation might be that factors such as the ability of EcN to produce microcin and also the presence of several different iron uptake systems (69) in its genome might have conferred a competitive edge to EcN over LGG in EcN+LGG-cocolonized piglets. Our study has implications for the use of probiotics to treat RV infections to effectively manage an active HRV outbreak. We envisage that supplementation of EcN probiotic might be a cost-effective therapy to ameliorate the severity of RV infections. One caveat of this study is use of the Gn piglets instead of conventional animals to determine beneficial effects of the probiotics on RV infection and immunity. Although our approach is advantageous in terms of eliminating the confounding effects of the intestinal microbiota and defining the effects of individual or combined probiotics on HRV infections, these findings have to be further validated using conventionalized animals such as Gn piglets colonized with human fecal microbiota. Furthermore, our study indicates that EcN directly interacts with HRV, and it remains to be determined whether EcN has a positive or negative effect on responses to HRV vaccine. In summary, compared with the G+ LGG probiotic, the G− EcN had greater beneficial effects in ameliorating VirHRV infection and promoting the development of the neonatal intestinal and systemic Ab responses.

We thank the technical assistance of Dr. Juliette Hanson, R. Wood, J. Ogg, Thavamathi Annamalai, and Kyle T. Scheuer.

This work was supported by National Institute of Allergy and Infectious Diseases/National Institutes of Health Grant R01 A1099451 (to L.J.S.) and federal funds appropriated to the Ohio Agricultural Research and Development Center of Ohio State University.

The online version of this article contains supplemental material.

Abbreviations used in this article:

ASC

Ab-secreting cell

EcN

Escherichia coli Nissle

G−

Gram-negative

G+

Gram-positive

GI

gastrointestinal

Gn

gnotobiotic

HRV

human rotavirus

LB

Luria–Bertani

LGG

Lactobacillus rhamnosus strain GG

MNC

mononuclear cell

PBCD

postbacterial colonization day

PCD

postchallenge day

RV

rotavirus

VirHRV

virulent human rotavirus

VLP

virus-like particle.

1
Tate
J. E.
,
Burton
A. H.
,
Boschi-Pinto
C.
,
Steele
A. D.
,
Duque
J.
,
Parashar
U. D.
WHO-coordinated Global Rotavirus Surveillance Network
.
2012
.
2008 estimate of worldwide rotavirus-associated mortality in children younger than 5 years before the introduction of universal rotavirus vaccination programmes: a systematic review and meta-analysis.
Lancet Infect. Dis.
12
:
136
141
.
2
Viboud, C., B. A. Lopman, V. E. Pitzer, R. Sarkar, B. Gladstone, M. Patel, J. Glasser, M. Gambhir, C. Atchison, and B. T. Grenfell. 2012. Understanding reduced rotavirus vaccine efficacy in low socio-economic settings. PloS One 7: Article ID e41720, 41727 pages
.
3
Ferreira
R. B.
,
Antunes
L. C. M.
,
Finlay
B. B.
.
2010
.
Should the human microbiome be considered when developing vaccines?
PLoS Pathog.
6
:
e1001190
.
4
Qadri
F.
,
Bhuiyan
T. R.
,
Sack
D. A.
,
Svennerholm
A. M.
.
2013
.
Immune responses and protection in children in developing countries induced by oral vaccines.
Vaccine
31
:
452
460
.
5
Valdez
Y.
,
Brown
E. M.
,
Finlay
B. B.
.
2014
.
Influence of the microbiota on vaccine effectiveness.
Trends Immunol.
35
:
526
537
.
6
Wilks
J.
,
Golovkina
T.
.
2012
.
Influence of microbiota on viral infections.
PLoS Pathog.
8
:
e1002681
.
7
Robinson
C. M.
,
Pfeiffer
J. K.
.
2014
.
Viruses and the microbiota.
Ann. Rev. Virol.
1
:
55
69
.
8
Uchiyama
R.
,
Chassaing
B.
,
Zhang
B.
,
Gewirtz
A. T.
.
2014
.
Antibiotic treatment suppresses rotavirus infection and enhances specific humoral immunity.
J. Infect. Dis.
210
:
171
182
.
9
Huda
M. N.
,
Lewis
Z.
,
Kalanetra
K. M.
,
Rashid
M.
,
Ahmad
S. M.
,
Raqib
R.
,
Qadri
F.
,
Underwood
M. A.
,
Mills
D. A.
,
Stephensen
C. B.
.
2014
.
Stool microbiota and vaccine responses of infants.
Pediatrics
134
:
e362
e372
.
10
Kuss
S. K.
,
Best
G. T.
,
Etheredge
C. A.
,
Pruijssers
A. J.
,
Frierson
J. M.
,
Hooper
L. V.
,
Dermody
T. S.
,
Pfeiffer
J. K.
.
2011
.
Intestinal microbiota promote enteric virus replication and systemic pathogenesis.
Science
334
:
249
252
.
11
Kane
M.
,
Case
L. K.
,
Kopaskie
K.
,
Kozlova
A.
,
MacDearmid
C.
,
Chervonsky
A. V.
,
Golovkina
T. V.
.
2011
.
Successful transmission of a retrovirus depends on the commensal microbiota.
Science
334
:
245
249
.
12
Szajewska
H.
,
Mrukowicz
J. Z.
.
2001
.
Probiotics in the treatment and prevention of acute infectious diarrhea in infants and children: a systematic review of published randomized, double-blind, placebo-controlled trials.
J. Pediatr. Gastroenterol. Nutr.
33
(
Suppl. 2
):
S17
S25
.
13
Sanders
M. E.
,
Guarner
F.
,
Guerrant
R.
,
Holt
P. R.
,
Quigley
E. M.
,
Sartor
R. B.
,
Sherman
P. M.
,
Mayer
E. A.
.
2013
.
An update on the use and investigation of probiotics in health and disease.
Gut
62
:
787
796
.
14
Majamaa
H.
,
Isolauri
E.
,
Saxelin
M.
,
Vesikari
T.
.
1995
.
Lactic acid bacteria in the treatment of acute rotavirus gastroenteritis.
J. Pediatr. Gastroenterol. Nutr.
20
:
333
338
.
15
Kaila
M.
,
Isolauri
E.
,
Soppi
E.
,
Virtanen
E.
,
Laine
S.
,
Arvilommi
H.
.
1992
.
Enhancement of the circulating antibody secreting cell response in human diarrhea by a human Lactobacillus strain.
Pediatr. Res.
32
:
141
144
.
16
Zhang
W.
,
Azevedo
M. S.
,
Wen
K.
,
Gonzalez
A.
,
Saif
L. J.
,
Li
G.
,
Yousef
A. E.
,
Yuan
L.
.
2008
.
Probiotic Lactobacillus acidophilus enhances the immunogenicity of an oral rotavirus vaccine in gnotobiotic pigs.
Vaccine
26
:
3655
3661
.
17
Chattha
K. S.
,
Vlasova
A. N.
,
Kandasamy
S.
,
Rajashekara
G.
,
Saif
L. J.
.
2013
.
Divergent immunomodulating effects of probiotics on T cell responses to oral attenuated human rotavirus vaccine and virulent human rotavirus infection in a neonatal gnotobiotic piglet disease model.
J. Immunol.
191
:
2446
2456
.
18
Kandasamy
S.
,
Chattha
K. S.
,
Vlasova
A. N.
,
Rajashekara
G.
,
Saif
L. J.
.
2014
.
Lactobacilli and Bifidobacteria enhance mucosal B cell responses and differentially modulate systemic antibody responses to an oral human rotavirus vaccine in a neonatal gnotobiotic pig disease model.
Gut Microbes
5
:
639
651
.
19
Vlasova
A. N.
,
Chattha
K. S.
,
Kandasamy
S.
,
Liu
Z.
,
Esseili
M.
,
Shao
L.
,
Rajashekara
G.
,
Saif
L. J.
.
2013
.
Lactobacilli and bifidobacteria promote immune homeostasis by modulating innate immune responses to human rotavirus in neonatal gnotobiotic pigs.
PLoS One
8
:
e76962
.
20
Guandalini
S.
,
Pensabene
L.
,
Zikri
M. A.
,
Dias
J. A.
,
Casali
L. G.
,
Hoekstra
H.
,
Kolacek
S.
,
Massar
K.
,
Micetic-Turk
D.
,
Papadopoulou
A.
, et al
.
2000
.
Lactobacillus GG administered in oral rehydration solution to children with acute diarrhea: a multicenter European trial.
J. Pediatr. Gastroenterol. Nutr.
30
:
54
60
.
21
Nowrouzian
F.
,
Hesselmar
B.
,
Saalman
R.
,
Strannegard
I. L.
,
Aberg
N.
,
Wold
A. E.
,
Adlerberth
I.
.
2003
.
Escherichia coli in infants’ intestinal microflora: colonization rate, strain turnover, and virulence gene carriage.
Pediatr. Res.
54
:
8
14
.
22
Kruis
W.
,
Fric
P.
,
Pokrotnieks
J.
,
Lukás
M.
,
Fixa
B.
,
Kascák
M.
,
Kamm
M. A.
,
Weismueller
J.
,
Beglinger
C.
,
Stolte
M.
, et al
.
2004
.
Maintaining remission of ulcerative colitis with the probiotic Escherichia coli Nissle 1917 is as effective as with standard mesalazine.
Gut
53
:
1617
1623
.
23
Hering
N. A.
,
Richter
J. F.
,
Fromm
A.
,
Wieser
A.
,
Hartmann
S.
,
Günzel
D.
,
Bücker
R.
,
Fromm
M.
,
Schulzke
J. D.
,
Troeger
H.
.
2014
.
TcpC protein from E. coli Nissle improves epithelial barrier function involving PKCζ and ERK1/2 signaling in HT-29/B6 cells.
Mucosal Immunol.
7
:
369
378
.
24
Rembacken
B. J.
,
Snelling
A. M.
,
Hawkey
P. M.
,
Chalmers
D. M.
,
Axon
A. T.
.
1999
.
Non-pathogenic Escherichia coli versus mesalazine for the treatment of ulcerative colitis: a randomised trial.
Lancet
354
:
635
639
.
25
Altenhoefer
A.
,
Oswald
S.
,
Sonnenborn
U.
,
Enders
C.
,
Schulze
J.
,
Hacker
J.
,
Oelschlaeger
T. A.
.
2004
.
The probiotic Escherichia coli strain Nissle 1917 interferes with invasion of human intestinal epithelial cells by different enteroinvasive bacterial pathogens.
FEMS Immunol. Med. Microbiol.
40
:
223
229
.
26
Schlee
M.
,
Wehkamp
J.
,
Altenhoefer
A.
,
Oelschlaeger
T. A.
,
Stange
E. F.
,
Fellermann
K.
.
2007
.
Induction of human β-defensin 2 by the probiotic Escherichia coli Nissle 1917 is mediated through flagellin.
Infect. Immun.
75
:
2399
2407
.
27
Sturm
A.
,
Rilling
K.
,
Baumgart
D. C.
,
Gargas
K.
,
Abou-Ghazalé
T.
,
Raupach
B.
,
Eckert
J.
,
Schumann
R. R.
,
Enders
C.
,
Sonnenborn
U.
, et al
.
2005
.
Escherichia coli Nissle 1917 distinctively modulates T-cell cycling and expansion via Toll-like receptor 2 signaling.
Infect. Immun.
73
:
1452
1465
.
28
Saif
L.
,
Ward
L.
,
Yuan
L.
,
Rosen
B.
,
To
T.
.
1996
.
The Gnotobiotic Piglet as a Model for Studies of Disease Pathogenesis and Immunity to Human Rotaviruses.
Springer
,
Vienna, Austria
.
29
Butler
J. E.
,
Lager
K. M.
,
Splichal
I.
,
Francis
D.
,
Kacskovics
I.
,
Sinkora
M.
,
Wertz
N.
,
Sun
J.
,
Zhao
Y.
,
Brown
W. R.
, et al
.
2009
.
The piglet as a model for B cell and immune system development.
Vet. Immunol. Immunopathol.
128
:
147
170
.
30
Butler
J. E.
,
Sun
J.
,
Weber
P.
,
Ford
S. P.
,
Rehakova
Z.
,
Sinkora
J.
,
Lager
K.
.
2001
.
Antibody repertoire development in fetal and neonatal piglets. IV. Switch recombination, primarily in fetal thymus, occurs independent of environmental antigen and is only weakly associated with repertoire diversification.
J. Immunol.
167
:
3239
3249
.
31
Šinkora
M.
,
Šinkorova
J.
,
Butler
J. E.
.
2002
.
B cell development and VDJ rearrangement in the fetal pig.
Vet. Immunol. Immunopathol.
87
:
341
346
.
32
Meyer
R. C.
,
Bohl
E. H.
,
Kohler
E. M.
.
1964
.
Procurement and maintenance of germ-free seine for microbiological investigations.
Appl. Microbiol.
12
:
295
300
.
33
Kumar
A.
,
Vlasova
A. N.
,
Liu
Z.
,
Chattha
K. S.
,
Kandasamy
S.
,
Esseili
M.
,
Zhang
X.
,
Rajashekara
G.
,
Saif
L. J.
.
2014
.
In vivo gut transcriptome responses to Lactobacillus rhamnosus GG and Lactobacillus acidophilus in neonatal gnotobiotic piglets.
Gut Microbes
5
:
152
164
.
34
Ward
L. A.
,
Rosen
B. I.
,
Yuan
L.
,
Saif
L. J.
.
1996
.
Pathogenesis of an attenuated and a virulent strain of group A human rotavirus in neonatal gnotobiotic pigs.
J. Gen. Virol.
77
:
1431
1441
.
35
Yuan
L.
,
Kang
S. Y.
,
Ward
L. A.
,
To
T. L.
,
Saif
L. J.
.
1998
.
Antibody-secreting cell responses and protective immunity assessed in gnotobiotic pigs inoculated orally or intramuscularly with inactivated human rotavirus.
J. Virol.
72
:
330
338
.
36
Yuan
L.
,
Ward
L. A.
,
Rosen
B. I.
,
To
T. L.
,
Saif
L. J.
.
1996
.
Systematic and intestinal antibody-secreting cell responses and correlates of protective immunity to human rotavirus in a gnotobiotic pig model of disease.
J. Virol.
70
:
3075
3083
.
37
Parreño
V.
,
Hodgins
D. C.
,
de Arriba
L.
,
Kang
S. Y.
,
Yuan
L.
,
Ward
L. A.
,
T. L.
,
Saif
L. J.
.
1999
.
Serum and intestinal isotype antibody responses to Wa human rotavirus in gnotobiotic pigs are modulated by maternal antibodies.
J. Gen. Virol.
80
:
1417
1428
.
38
Sinkora
M.
,
Stepanova
K.
,
Butler
J. E.
,
Francis
D.
,
Santiago-Mateo
K.
,
Potockova
H.
,
Karova
K.
,
Sinkorova
J.
.
2011
.
Ileal Peyer’s patches are not necessary for systemic B cell development and maintenance and do not contribute significantly to the overall B cell pool in swine.
J. Immunol.
187
:
5150
5161
.
39
Zhang
W.
,
Azevedo
M. S.
,
Gonzalez
A. M.
,
Saif
L. J.
,
Van Nguyen
T.
,
Wen
K.
,
Yousef
A. E.
,
Yuan
L.
.
2008
.
Influence of probiotic Lactobacilli colonization on neonatal B cell responses in a gnotobiotic pig model of human rotavirus infection and disease.
Vet. Immunol. Immunopathol.
122
:
175
181
.
40
Cukrowska
B.
,
LodInová-ZádnIková
R.
,
Enders
C.
,
Sonnenborn
U.
,
Schulze
J.
,
Tlaskalová-Hogenová
H.
.
2002
.
Specific proliferative and antibody responses of premature infants to intestinal colonization with nonpathogenic probiotic E. coli strain Nissle 1917.
Scand. J. Immunol.
55
:
204
209
.
41
Graham
K. L.
,
Halasz
P.
,
Tan
Y.
,
Hewish
M. J.
,
Takada
Y.
,
Mackow
E. R.
,
Robinson
M. K.
,
Coulson
B. S.
.
2003
.
Integrin-using rotaviruses bind α2β1 integrin α2 I domain via VP4 DGE sequence and recognize αXβ2 and αVβ3 by using VP7 during cell entry.
J. Virol.
77
:
9969
9978
.
42
Hewish
M. J.
,
Takada
Y.
,
Coulson
B. S.
.
2000
.
Integrins α2β1 and α4β1 can mediate SA11 rotavirus attachment and entry into cells.
J. Virol.
74
:
228
236
.
43
Letourneau
J.
,
Levesque
C.
,
Berthiaume
F.
,
Jacques
M.
,
Mourez
M.
.
2011
.
In vitro assay of bacterial adhesion onto mammalian epithelial cells.
J. Vis. Exp.
(
51
):
2783
.
44
Bunthof
C. J.
,
Abee
T.
.
2002
.
Development of a flow cytometric method to analyze subpopulations of bacteria in probiotic products and dairy starters.
Appl. Environ. Microbiol.
68
:
2934
2942
.
45
Yuan
L.
,
Geyer
A.
,
Saif
L. J.
.
2001
.
Short-term immunoglobulin A B-cell memory resides in intestinal lymphoid tissues but not in bone marrow of gnotobiotic pigs inoculated with Wa human rotavirus.
Immunology
103
:
188
198
.
46
Iosef
C.
,
Van Nguyen
T.
,
Jeong
K.-i.
,
Bengtsson
K.
,
Morein
B.
,
Kim
Y.
,
Chang
K.-O.
,
Azevedo
M. S.
,
Yuan
L.
,
Nielsen
P.
,
Saif
L. J.
.
2002
.
Systemic and intestinal antibody secreting cell responses and protection in gnotobiotic pigs immunized orally with attenuated Wa human rotavirus and Wa 2/6-rotavirus-like-particles associated with immunostimulating complexes.
Vaccine
20
:
1741
1753
.
47
Trask
S. D.
,
McDonald
S. M.
,
Patton
J. T.
.
2012
.
Structural insights into the coupling of virion assembly and rotavirus replication.
Nat. Rev. Microbiol.
10
:
165
177
.
48
Robinson
C. M.
,
Jesudhasan
P. R.
,
Pfeiffer
J. K.
.
2014
.
Bacterial lipopolysaccharide binding enhances virion stability and promotes environmental fitness of an enteric virus.
Cell Host Microbe
15
:
36
46
.
49
Ludert
J. E.
,
Feng
N.
,
Yu
J. H.
,
Broome
R. L.
,
Hoshino
Y.
,
Greenberg
H. B.
.
1996
.
Genetic mapping indicates that VP4 is the rotavirus cell attachment protein in vitro and in vivo.
J. Virol.
70
:
487
493
.
50
Zhang
B.
,
Chassaing
B.
,
Shi
Z.
,
Uchiyama
R.
,
Zhang
Z.
,
Denning
T. L.
,
Crawford
S. E.
,
Pruijssers
A. J.
,
Iskarpatyoti
J. A.
,
Estes
M. K.
, et al
.
2014
.
Viral infection: prevention and cure of rotavirus infection via TLR5/NLRC4-mediated production of IL-22 and IL-18.
Science
346
:
861
865
.
51
Grabig
A.
,
Paclik
D.
,
Guzy
C.
,
Dankof
A.
,
Baumgart
D. C.
,
Erckenbrecht
J.
,
Raupach
B.
,
Sonnenborn
U.
,
Eckert
J.
,
Schumann
R. R.
, et al
.
2006
.
Escherichia coli strain Nissle 1917 ameliorates experimental colitis via Toll-like receptor 2‑ and Toll-like receptor 4‑dependent pathways.
Infect. Immun.
74
:
4075
4082
.
52
Henker
J.
,
Laass
M.
,
Blokhin
B. M.
,
Bolbot
Y. K.
,
Maydannik
V. G.
,
Elze
M.
,
Wolff
C.
,
Schulze
J.
.
2007
.
The probiotic Escherichia coli strain Nissle 1917 (EcN) stops acute diarrhoea in infants and toddlers.
Eur. J. Pediatr.
166
:
311
318
.
53
Henker
J.
,
Laass
M. W.
,
Blokhin
B. M.
,
Maydannik
V. G.
,
Bolbot
Y. K.
,
Elze
M.
,
Wolff
C.
,
Schreiner
A.
,
Schulze
J.
.
2008
.
Probiotic Escherichia coli Nissle 1917 versus placebo for treating diarrhea of greater than 4 days duration in infants and toddlers.
Pediatr. Infect. Dis. J.
27
:
494
499
.
54
Butler
J. E.
,
Weber
P.
,
Sinkora
M.
,
Baker
D.
,
Schoenherr
A.
,
Mayer
B.
,
Francis
D.
.
2002
.
Antibody repertoire development in fetal and neonatal piglets. VIII. Colonization is required for newborn piglets to make serum antibodies to T-dependent and type 2 T-independent antigens.
J. Immunol.
169
:
6822
6830
.
55
Haverson
K.
,
Rehakova
Z.
,
Sinkora
J.
,
Sver
L.
,
Bailey
M.
.
2007
.
Immune development in jejunal mucosa after colonization with selected commensal gut bacteria: a study in germ-free pigs.
Vet. Immunol. Immunopathol.
119
:
243
253
.
56
Wechsler-Reya
R. J.
,
Monroe
J. G.
.
1996
.
Lipopolysaccharide prevents apoptosis and induces responsiveness to antigen receptor cross-linking in immature B cells.
Immunology
89
:
356
362
.
57
Hayashi
E. A.
,
Akira
S.
,
Nobrega
A.
.
2005
.
Role of TLR in B cell development: signaling through TLR4 promotes B cell maturation and is inhibited by TLR2.
J. Immunol.
174
:
6639
6647
.
58
Pone
E. J.
,
Lou
Z.
,
Lam
T.
,
Greenberg
M. L.
,
Wang
R.
,
Xu
Z.
,
Casali
P.
.
2015
.
B cell TLR1/2, TLR4, TLR7 and TLR9 interact in induction of class switch DNA recombination: modulation by BCR and CD40, and relevance to T-independent antibody responses.
Autoimmunity
48
:
1
12
.
59
Burdin
N.
,
Van Kooten
C.
,
Galibert
L.
,
Abrams
J. S.
,
Wijdenes
J.
,
Banchereau
J.
,
Rousset
F.
.
1995
.
Endogenous IL-6 and IL-10 contribute to the differentiation of CD40-activated human B lymphocytes.
J. Immunol.
154
:
2533
2544
.
60
Rousset
F.
,
Garcia
E.
,
Defrance
T.
,
Péronne
C.
,
Vezzio
N.
,
Hsu
D. H.
,
Kastelein
R.
,
Moore
K. W.
,
Banchereau
J.
.
1992
.
Interleukin 10 is a potent growth and differentiation factor for activated human B lymphocytes.
Proc. Natl. Acad. Sci. USA
89
:
1890
1893
.
61
Ramsay
A. J.
,
Husband
A. J.
,
Ramshaw
I. A.
,
Bao
S.
,
Matthaei
K. I.
,
Koehler
G.
,
Kopf
M.
.
1994
.
The role of interleukin-6 in mucosal IgA antibody responses in vivo.
Science
264
:
561
563
.
62
Karlsson
H.
,
Larsson
P.
,
Wold
A. E.
,
Rudin
A.
.
2004
.
Pattern of cytokine responses to Gram-positive and Gram-negative commensal bacteria is profoundly changed when monocytes differentiate into dendritic cells.
Infect. Immun.
72
:
2671
2678
.
63
Hessle
C.
,
Andersson
B.
,
Wold
A. E.
.
2000
.
Gram-positive bacteria are potent inducers of monocytic interleukin-12 (IL-12) while Gram-negative bacteria preferentially stimulate IL-10 production.
Infect. Immun.
68
:
3581
3586
.
64
Azevedo
M. S.
,
Zhang
W.
,
Wen
K.
,
Gonzalez
A. M.
,
Saif
L. J.
,
Yousef
A. E.
,
Yuan
L.
.
2012
.
Lactobacillus acidophilus and Lactobacillus reuteri modulate cytokine responses in gnotobiotic pigs infected with human rotavirus.
Benef. Microbes
3
:
33
42
.
65
Zhang
H.
,
Wang
H.
,
Shepherd
M.
,
Wen
K.
,
Li
G.
,
Yang
X.
,
Kocher
J.
,
Giri-Rachman
E.
,
Dickerman
A.
,
Settlage
R.
,
Yuan
L.
.
2014
.
Probiotics and virulent human rotavirus modulate the transplanted human gut microbiota in gnotobiotic pigs.
Gut Pathog.
6
:
39
.
66
Wen
K.
,
Tin
C.
,
Wang
H.
,
Yang
X.
,
Li
G.
,
Giri-Rachman
E.
,
Kocher
J.
,
Bui
T.
,
Clark-Deener
S.
,
Yuan
L.
.
2014
.
Probiotic Lactobacillus rhamnosus GG enhanced Th1 cellular immunity but did not affect antibody responses in a human gut microbiota transplanted neonatal gnotobiotic pig model.
PLoS One
9
:
e94504
.
67
Eloe-Fadrosh
E. A.
,
Brady
A.
,
Crabtree
J.
,
Drabek
E. F.
,
Ma
B.
,
Mahurkar
A.
,
Ravel
J.
,
Haverkamp
M.
,
Fiorino
A. M.
,
Botelho
C.
, et al
.
2015
.
Functional dynamics of the gut microbiome in elderly people during probiotic consumption.
MBio
6
:
6
.
68
Denou
E.
,
Rezzonico
E.
,
Panoff
J. M.
,
Arigoni
F.
,
Brüssow
H.
.
2009
.
A Mesocosm of Lactobacillus johnsonii, Bifidobacterium longum, and Escherichia coli in the mouse gut.
DNA Cell Biol.
28
:
413
422
.
69
Patzer
S. I.
,
Baquero
M. R.
,
Bravo
D.
,
Moreno
F.
,
Hantke
K.
.
2003
.
The colicin G, H and X determinants encode microcins M and H47, which might utilize the catecholate siderophore receptors FepA, Cir, Fiu and IroN.
Microbiology
149
:
2557
2570
.

The authors have no financial conflicts of interest.

Supplementary data