ABSTRACT
Human rotavirus (HRV) is a major cause of childhood diarrhea in developing countries where widespread malnutrition contributes to the decreased oral vaccine efficacy and increased prevalence of other enteric infections, which are major concerns for global health. Neonatal gnotobiotic (Gn) piglets closely resemble human infants in their anatomy, physiology, and outbred status, providing a unique model to investigate malnutrition, supplementations, and HRV infection. To understand the molecular signatures associated with immune enhancement and reduced diarrheal severity by Escherichia coli Nissle 1917 (EcN) and tryptophan (TRP), immunological responses and global nontargeted metabolomics and lipidomics approaches were investigated on the plasma and fecal contents of malnourished pigs transplanted with human infant fecal microbiota and infected with virulent (Vir) HRV. Overall, EcN + TRP combined (rather than individual supplement action) promoted greater and balanced immunoregulatory/immunostimulatory responses associated with greater protection against HRV infection and disease in malnourished humanized piglets. Moreover, EcN + TRP treatment upregulated the production of several metabolites with immunoregulatory/immunostimulatory properties: amino acids (N-acetylserotonin, methylacetoacetyl-CoA), lipids (gamma-butyrobetaine, eicosanoids, cholesterol-sulfate, sphinganine/phytosphingosine, leukotriene), organic compound (biliverdin), benzenoids (gentisic acid, aminobenzoic acid), and nucleotides (hypoxathine/inosine/xanthine, cytidine-5′-monophosphate). Additionally, the levels of several proinflammatory metabolites of organic compounds (adenosylhomocysteine, phenylacetylglycine, urobilinogen/coproporphyrinogen) and amino acid (phenylalanine) were reduced following EcN + TRP treatment. These results suggest that the EcN + TRP effects on reducing HRV diarrhea in neonatal Gn pigs were at least in part due to altered metabolites, those involved in lipid, amino acid, benzenoids, organic compounds, and nucleotide metabolism. Identification of these important mechanisms of EcN/TRP prevention of HRV diarrhea provides novel targets for therapeutics development.
IMPORTANCE Human rotavirus (HRV) is the most common cause of viral gastroenteritis in children, especially in developing countries, where the efficacy of oral HRV vaccines is reduced. Escherichia coli Nissle 1917 (EcN) is used to treat enteric infections and ulcerative colitis while tryptophan (TRP) is a biomarker of malnutrition, and its supplementation can alleviate intestinal inflammation and normalize intestinal microbiota in malnourished hosts. Supplementation of EcN + TRP to malnourished humanized gnotobiotic piglets enhanced immune responses and resulted in greater protection against HRV infection and diarrhea. Moreover, EcN + TRP supplementation increased the levels of immunoregulatory/immunostimulatory metabolites while decreasing the production of proinflammatory metabolites in plasma and fecal samples. Profiling of immunoregulatory and proinflammatory biomarkers associated with HRV perturbations will aid in the identification of treatments against HRV and other enteric diseases in malnourished children.
KEYWORDS: Escherichia coli Nissle 1917, tryptophan, human rotavirus infection, metabolomics, lipidomics, neonatal gnotobiotic pigs, malnutrition
INTRODUCTION
Human rotavirus (HRV) is a leading cause of diarrhea in children. It is associated with significant morbidity and mortality, especially in developing countries (1). Coincidently, the efficacy of RV vaccine is low in Low and Middle Income Countires (LMICs) (2). Malnutrition, micronutrient deficiencies, and interference by maternal milk antibodies are implicated in the lower efficacy of oral RV vaccines (3–5). Probiotics are increasingly used to enhance oral vaccine responses and treat enteric infections (6) and ulcerative colitis in children (7). The probiotic Escherichia coli Nissle 1917 (EcN) has been widely used in the treatment of ulcerative colitis in humans (8). EcN lacks virulence factors and possesses unique health-promoting properties (9). Our research group has previously shown that EcN protects gnotobiotic (Gn) pigs against HRV infection and decreases the severity of diarrhea by modulating innate and adaptive immunity and protecting the intestinal epithelium by binding HRV via histo-blood group antigen-like bacterial glycans (10–12).
Tryptophan (TRP) is one of the essential amino acids of protein metabolism, and the kynurenine-to-tryptophan ratio is negatively associated with the linear growth of children (13, 14). TRP also plays an important role in immune regulation. Altered TRP homeostasis coinciding with decreased serum angiotensin I-converting enzyme 2 levels has been observed previously in association with virulent (Vir) HRV challenge of Gn pigs fed a protein-calorie-deficient diet (15). TRP supplementation alleviated intestinal inflammation and normalized intestinal microbiota in malnourished mice (16). Moreover, it is catabolized via the indoleamine 2,3-dioxygenase pathway that has immunoregulatory effects, and its depletion has been well studied in the context of chronic infections (17). TRP is the precursor to serotonin, and EcN enhances the bioavailability of serotonin in gut tissues through modulation of its synthesis and clearance (18). Indole produced by EcN via tryptophanase also improves intestinal barrier function (19). Thus, EcN may influence and be affected by host nutritional status, and its coadministration with TRP may provide additional benefits to counteract malnutrition impacts efficiently and alleviate HRV disease. In addition, the interactions between EcN and TRP and the combined impact of these interactions on immune responses/metabolic activities to HRV disease in the context of host malnutrition have not been investigated previously.
Neonatal Gn pigs resemble infants in their physiology, anatomy, mucosal immune system, and outbred status (20–22). Gn pigs are immunocompetent at birth but immunologically immature (23). HRV-infected Gn pigs exhibit diarrhea, transient viremia, and intestinal lesions mimicking those in children (22). Gn pigs are caesarian derived and housed in sterile isolators to ensure their germfree status, permitting studies of gut colonization with single probiotic bacteria or complete fecal microbiota. Thus, Gn pigs are a unique animal model to study the impact of specific probiotics on host metabolism, the neonatal immune system, enteric viral infections, or oral vaccines without other confounding microbiota (24, 25). Importantly, transplantation of human infant fecal microbiota (HIFM) into Gn piglets recapitulates the infant microbial community (26, 27). The resulting microbiota-humanized Gn pigs allow manipulation of multiple variables and sampling of gut responses that are not possible in infants or conventional animal models.
Metabolomics is the analysis of the metabolites present in mammalian specimens in various states of health and disease and can identify disease biomarkers. All the biological samples can be investigated, but plasma, serum, urine, and feces are used commonly because the fecal metabolome reflects the host-microbiota interactions, in contrast to plasma/serum, which more closely captures the host’s metabolism. Depending on the type of biological material tested, various metabolites can be discovered (28). Recent findings confirmed alterations in the numbers of metabolites, mainly related to lipids and amino acids, in the serum of patients with inflammatory bowel disease (29–31).
Our study aimed to determine the usefulness of global nontargeted metabolomics and lipidomics of plasma and fecal metabolites in a humanized malnourished Gn pig model supplemented with EcN and TRP. Further, we investigated the potential immunoregulatory and proinflammatory biomarkers associated with reduced HRV severity in children. Additionally we evaluated the association between metabolites, immune responses, and HRV disease severity.
We hypothesized that combined EcN and TRP will enhance host immune responses and alter host metabolites in the humanized HIFM Gn pigs, which may be associated with moderating HRV infection. We further hypothesized that the identification of HRV-associated metabolic biomarkers will delineate host pathways and molecular targets, which can be used to develop HRV therapeutics targeting pathways perturbed by HRV. Our preliminary studies have indicated that treatment of malnourished humanized Gn pigs with combined EcN + TRP ameliorated the HRV diarrhea rather than single (EcN or TRP) supplementation.
RESULTS
EcN and TRP combination increased survival rate and body weight, reduced viral shedding and diarrheal occurrence, and enhanced immunological responses.
EcN, TRP, and EcN + TRP supplementations increased the survival rates to 74%, 85%, and 86%, respectively, compared with the untreated (Def) pigs’ survival rate of 55% (Fig. 1A). During the first week of age, all the piglets had gained weight only marginally. At 2 weeks of age, a significant weight gain was evident in EcN + TRP pigs followed by EcN- and TRP-treated pigs compared with control untreated Def pigs (Fig. 1B). Analysis revealed that EcN + TRP-treated pigs had decreased peak virus shedding titers at PCD3 to PCD5 compared with other groups (Fig. 1C). Whereas single EcN or TRP treatment pigs had decreased peak virus shedding titers later at PCD5 to PCD6. Our results showed decreased diarrheal occurrence in EcN + TRP (28%) group compared with EcN (33%), TRP (42%), and Def group (75%). The TRP treatment group had the lowest diarrheal score (3.7) followed by EcN + TRP (4.1), EcN (5.1) treatments and control Def (8.0) groups (Table 1). Moreover, delayed onset of diarrhea was observed in EcN + TRP (4.0), EcN (3.0), and TRP (4.3) compared with Def (2.6) groups. Additionally, the shortened duration of diarrhea was also observed in EcN + TRP (0.3)-treated followed by EcN (1.0)- and TRP (0.4)-treated groups compared with Def (4.0) group pigs. Furthermore, diarrheal scores were correlated with viral shedding titers (R = 0.44, P = 0.02).
FIG 1.
EcN + TRP treatment enhanced B cell immune responses post-VirHRV challenge. Survival rate (A) and normalized weight gain of pigs (B) after HIFM transplantation, supplementation, and VirHRV challenge were monitored for 2 weeks. (C) Virus shedding was determined by cell culture immunofluorescence assay and expressed as log10 FFU/ml. (D) Principal-component analysis (PCA) of immunological parameters. (E) Mean HRV-specific IgA antibody-secreting cells (ASCs) in splenic and duodenal cells. (F to H) Geometric mean titers (log10) HRV-specific IgA antibody in serum, small intestinal contents (SIC), and large intestinal content (LIC). (I) Mean frequencies of CD79β+IgA+ B cells in splenic and duodenal cells. (J) Mean frequencies of activated antibody-forming (CD79β+CD21+CD2−) B cells in systemic and ileal cells. Data are shown as means ± SEM, and data are compared with untreated group pigs. Gnotobiotic (Gn) pigs were cesarian derived and transplanted with HIFM at 4 days of age, post-HIFM transplantation day (PBTD) 0. Pigs were fed a deficient (Def) and/or sufficient (Suf) diet, supplemented orally with Escherichia Coli Nissle 1917 (EcN) and/or tryptophan (TRP) at PBTD7, subsequently challenged with virulent (Vir) HRV at PBTD9-10/postchallenge day (PCD) 0, and euthanized on PBTD24/PCD14.
TABLE 1.
Summary of diarrhea following VirHRV challenge (PCD1–PCD6)
| Groupsa | n | Diarrheab (%) | Mean cumulative fecal scorec | Mean days to onset of diarrhea | Mean duration of diarrhead |
|---|---|---|---|---|---|
| Def + EcN + HIFM + VirHRV | 6 | 33 | 5.1*e | 3.0 | 1.0 |
| Def + TRP + HIFM + VirHRV | 7 | 42 | 3.7* | 4.3 | 0.4 |
| Def + EcN + TRP + HIFM + VirHRV | 7 | 28 | 4.1* | 4.0 | 0.3 |
| Def + HIFM + VirHRV | 6 | 75 | 8.0**f | 2.6 | 4.0 |
| Suf + HIFM + VirHRV | 7 | 57 | 6.7***g | 3.0 | 1.0 |
Gnotobiotic (Gn) pigs were transplanted with human infant fecal microbiota (HIFM) at 4 days of age, post-HIFM transplantation day (PBTD) 0. Pigs were fed either deficient (Def) or sufficient (Suf) diet. Pigs were supplemented orally with E. coli Nissle (EcN) 1917 and/or tryptophan (TRP) at PBTD7, subsequently challenged with virulent human rotavirus (VirHRV) at PBTD9-10/postchallenge day (PCD) 0 and pigs were euthanized on PBTD24/PCD14.
Pigs with fecal score > 1 were considered diarrheic. Fecal consistency was scored as follows: 0, normal; 1, pasty; 2, semiliquid; and 3, liquid.
Mean of total fecal score from PCD1 to PCD6. Means in the same column with different asterisks differ significantly (determined by two-way ANOVA followed by Bonferroni posttest, P < 0.05).
Mean of the total days with fecal score > 1.
*, P < 0.05.
**, P < 0.01.
***, P < 0.001.
Principal-component analysis (PCA) identified immunological similarities/dissimilarities among the treatment groups (Fig. 1D). Clear immunological segregation was observed between Suf and Def diet pig groups expanding over 41% variance. Malnourished piglets supplemented with EcN + TRP and EcN had the highest immunological resemblance followed by TRP supplementation compared with the Suf pig group at component 1.
Analysis of B cells revealed that EcN + TRP, EcN, and TRP treatments had increased numbers of HRV-specific IgA antibody-secreting cells (ASCs) in splenic (negatively correlated with viral shedding titers, R = −0.5, P = 0.001) and duodenal cells (Fig. 1E). Coincident with decreased diarrheal score and increased HRV-specific ASCs, EcN + TRP, EcN, and TRP treatments increased HRV-specific IgA antibody titers in serum, small intestinal content (SIC), and large intestinal contents (LIC) (Fig. 1F to H). IgA antibody is a major functional component of the humoral adaptive immune system at mucosal sites (32) and the levels of HRV-specific IgA antibodies in pigs strongly correlate with protection against HRV infection (33–35). Similar to ASCs and IgA antibody titers, EcN + TRP, EcN, and TRP treatments also increased frequencies of IgA+ B lymphocytes among splenic and duodenal mononuclear cells (MNCs) (Fig. 1I). This is in agreement with previous studies where oral administration of probiotics increased the number of IgA+ B cells in the lamina propria (36, 37). EcN + TRP, EcN, and TRP treatments numerically increased frequencies of activated/primed (CD21+CD2−CD79β+) B cells among systemic and ileal MNCs (Fig. 1J). Taken together these results indicate that concurrent EcN + TRP treatment enhanced B cells in circulation and the gut of malnourished VirHRV-challenged HIFM-transplanted Gn pig model.
Analysis of MNCs in different tissues revealed that EcN + TRP (significantly), EcN, and TRP treatments increased the total numbers of MNCs among ileal cells (Fig. 2A) and were negatively correlated with viral shedding titers (R = −0.5, P = 0.004). Similarly, EcN + TRP, EcN, and TRP treatments increased the frequencies of lymphocytes among total MNCs in systemic tissues (Fig. 2B). Furthermore, EcN + TRP, EcN, and TRP treatments significantly increased frequencies of T-helper (CD3+CD4+) cells among blood and ileal MNCs (Fig. 2C), and they were positively correlated with each other (R = 0.88, P = 0.0004). T-helper cell frequencies were positively correlated with total ileal MNC counts (R = 0.65, P = 0.007) and negatively correlated with viral shedding titers (R = −0.5, P = 0.01). Similarly, EcN + TRP (significantly), EcN, and TRP treatments increased the frequencies of cytotoxic (CD3+CD8+) T cells in blood (Fig. 2D); frequencies of activated (CD4+CD25+FoxP3−) Treg cells in blood (significantly, Fig. 2E) that correlated with HRV-specific IgA ASCs in duodenum (R = 0.88, P = 0.0004); and frequencies of natural cytotoxic (CD8+CD25+FoxP3+) Treg cells among duodenal MNCs (Fig. 2F) that correlated with HRV-specific IgA ASCs in duodenal cells (R = 0.67, P = 0.006). Moreover, EcN + TRP, EcN, and TRP treatments increased the frequencies of CD4/CD8+ TGF-β + T cells among splenic MNCs (Fig. 2G and H). These results suggest that combined EcN + TRP treatment modulated the immunoregulatory environment and served as a potent inducer of intestinal immunity, restored gut immune homeostasis, and thus moderated HRV infection post-VirHRV challenge.
FIG 2.
EcN + TRP treatment enhanced T cell immune responses post-VirHRV challenge. (A) Total numbers of mononuclear cells (MNCs) in the ileum. (B) Mean frequencies of lymphocytes among total MNCs in systemic tissues. (C) Mean frequencies of T-helper (CD3+CD4+) cells in the blood and ileal tissues. (D and E) Mean frequency of cytotoxic (CD3+CD8+) T and activated (CD4+CD25+FOXP3−) T-regulatory cells in blood cells. (F) Mean frequency of natural (CD8+CD25+FOXP3+) T-regulatory cells in duodenal tissues. (G and H) Mean frequency of CD4/CD8+TGF-β + T cells in splenic cells. (I) Cytokines profile was determined in serum samples collected at the terminal time point (PCD7). Data are shown as means ± SEM. Gnotobiotic (Gn) pigs were cesarian derived and transplanted with human infant fecal microbiota (HIFM) at 4 days of age, post-HIFM transplantation day (PBTD) 0. Pigs were fed a deficient (Def) and/or sufficient (Suf) diet, supplemented orally with Escherichia Coli Nissle 1917 (EcN) and/or tryptophan (TRP) at PBTD7, subsequently challenged with virulent (Vir) HRV at PBTD9-10/postchallenge day (PCD) 0, and euthanized on PBTD24/PCD14. *, P < 0.05; **, P < 0.01, and ***, P < 0.001.
Additionally, proinflammatory and immunoregulatory cytokine responses post-VirHRV challenge were assessed by measuring levels of serum cytokines at terminal time points. Coincident with reduced diarrheal scores, EcN + TRP, EcN, and TRP treatments significantly reduced proinflammatory cytokine (interleukin-1β [IL-1β], IL-6, tumor necrosis factor-α [TNF-α], interferon-α [IFN-α], and IFN-γ) levels in serum, with combined EcN + TRP treatment showing remarkable reduction in IL-1β and TNF-α levels compared to single EcN/TRP treatments (Fig. 2I). Moreover, IFN-γ levels were correlated with diarrheal scores (R = 0.5, P = 0.003). These data suggest that concurrent EcN and TRP treatment reduced local (gut) inflammation caused by HRV infection. In contrast, immunoregulatory cytokine (IL-10 and TGF-β) levels were significantly increased in serum of EcN + TRP-, EcN-, and TRP-treated pigs (Fig. 2I), suggesting that EcN + TRP treatment induced an anti-inflammatory environment post-VirHRV challenge, thereby inhibiting proinflammatory cytokine responses.
Analysis of innate immune responses demonstrated that EcN + TRP (significantly), EcN, and TRP treatments increased frequencies of CD103+ pDCs in ileal MNCs compared to the control Def group (Fig. 3A). Coincident with decreased diarrhea severity, the frequencies of Toll-like receptor 4 (TLR4) (associated with proinflammatory signaling) expressing MNCs were decreased (numerically) in systemic and intestinal MNCs of EcN + TRP-treated pigs compared with untreated Def pigs (Fig. 3B), which is consistent with previous observations (10, 38). The frequencies of TLR3 (associated with anti-RV protection) and TLR9 (associated with anti-inflammatory signaling) expressing MNCs were increased among intestinal, blood (significantly), and splenic MNCs, respectively, of EcN + TRP-, EcN-, and TRP-treated pigs compared with untreated Def pigs (Fig. 3C and D). Moreover, the numbers of TLR9 expressing MNCs in the spleen and blood were positively correlated with each other (R = 0.66 P = 0.007). Furthermore, frequencies of TLR9 expressing MNCs in blood were positively correlated with CD103+ pDCs in ileal MNCs (R = 0.67 P = 0.006) and were negatively correlated with diarrheal scores (R = −0.54, P = 0.021). These results indicate that enhanced TLR3/TLR9 expression associated with EcN + TRP treatment facilitated more efficient RV clearance. Similarly, EcN + TRP, EcN, and TRP treatments numerically increased the frequency of NK cells among systemic MNCs (Fig. 3E) and NK cell function in the blood MNCs (Fig. 3F). Moreover, compared with untreated Def pigs, EcN + TRP, EcN, and TRP treatments decreased the frequencies of apoptotic MNCs among blood, splenic and duodenal MNCs (Fig. 3G). Apoptotic MNCs in the blood was negatively correlated with CD103+ pDCs in ileum (R = −0.67, P = 0.006); lymphocytes in the blood (R = −0.71, P = 0.002); and T-helper cells in the blood (R = −0.66, P = 0.007). Additionally, apoptotic MNCs in blood and spleen were negatively correlated with viral shedding titers (R = −0.5 P = 0.02, R = −0.5 P = 0.04, respectively). These data suggest that EcN and TRP treatment either singly or in combination enhanced innate immune responses and improved protection against HRV infection associated with malnourishment and VirHRV in Gn pig model, with combined treatment (EcN + TRP) generally showing enhanced effects.
FIG 3.
EcN + TRP treatment enhanced innate immune responses post-VirHRV challenge. (A) Mean frequency of CD103+ pDCs among total mononuclear cells (MNCs) in the ileum. (B) Mean frequencies of MNCs expressing TLR4 in systemic and intestinal tissues, (C) TLR3 in splenic and intestinal tissues, and (D) TLR9 in systemic tissues. (E) Mean frequency of NK (SWC3a+CD16+) and (F) NK cell function in systemic and blood MNCs. Blood MNCs and carboxyfluorescein diacetate succinimidyl ester (CFSE) stained K562 tumor cells were used as effector and target cells, respectively, and cocultured at set ratios to assess the NK cytotoxic function. For the effector, target cell cocultures were stained with 7-aminoactinomycin D (7AAD) after 12 h of incubation at 37°C, and the frequencies of CFSE-7AAD double-positive cells (lysed K562 target cells) were assessed by flow cytometry. (G) Mean frequencies of apoptotic MNCs (PI-/Annexin-APC-) among total blood MNCs. Data are shown as means ± SEM. Gnotobiotic (Gn) pigs were cesarian derived and transplanted with human infant fecal microbiota (HIFM) at 4 days of age, post-HIFM transplantation day (PBTD) 0. Pigs were fed a deficient (Def) and/or sufficient (Suf) diet, supplemented orally with Escherichia coli Nissle 1917 (EcN) and/or tryptophan (TRP) at PBTD7, subsequently challenged with VirHRV at PBTD9-10/postchallenge day (PCD) 0, and euthanized on PBTD24/PCD14. *, P < 0.05.
Untargeted plasma and fecal metabolomics revealed differentially altered metabolites among EcN, TRP, and EcN + TRP treatment groups.
A total of 14,380 putative metabolites were detected in plasma metabolomics (PM); however, only 21 metabolites were found significant (P < 0.005) (Table 2 and 3 and Table S1 in the supplemental material). Multivariate analysis by principal-component analysis (PCA) and a heatmap revealed segregation of the treatment groups compared with the untreated (Def and Suf) groups (Fig. 4 and 5). The PCA plot identified and segregated metabolites among treated/untreated groups in PM samples. A close resemblance of metabolites was observed between EcN + TRP and EcN groups followed by the TRP group compared to untreated Suf diet group. Clear segregation of metabolites from the Def diet group of pigs revealed over 10% variance at component 2.
TABLE 2.
Altered metabolites in different treatment groups
| Treatment groupsa | Immunoregulatory metabolites (↑)b | Proinflammatory metabolites (↓)c |
|---|---|---|
| Def + EcN + TRP + HIFM + VirHRV | Acetylserotonin, aminobenzoic acid, arginine, γ-butyrobetaine, biliverdin, cytidine-5′-monophosphate, cytidine, deoxythymidine, eicosanoids glycerophosethanolamine, hypoxanthine, inosine, leukotriene, phytosphingosine, presqualene diphosphate, retinoyl glucuronide, sphinganine, tetrahydropteroyldiglutamate (folic acid), trienoic acid, xanthine, | Acetylputrescine, adenosylhomocysteine, coproporphyrinogen, phenylalanine, phenylacetylglycine, urobilinogen |
| Def + EcN + HIFM + VirHRV | Acetylserotonin, acetyl glucosamine, gentisic acid/glycocholate/glycocholic acid, heme, methylacetoacetyl-CoA | Coproporphyrinogen, phenylalanine, urobilinogen |
| Def + TRP + HIFM + VirHRV | Nicotinamide adenine dinucleotide, palmitoylcarnitine, retinoyl glucuronide | Coproporphyrinogen, phenylalanine, urobilinogen |
Gnotobiotic (Gn) pigs were transplanted with human infant fecal microbiota (HIFM) at 4 days of age, post-HIFM transplantation day (PBTD) 0. Pigs were fed a deficient (Def) and/or sufficient (Suf) diet. Pigs were supplemented orally with E. coli Nissle (EcN) 1917 and/or tryptophan (TRP) at PBTD7, subsequently challenged with virulent human rotavirus (VirHRV) at PBTD9-10/postchallenge day (PCD) 0, and euthanized on PBTD24/PCD14.
↑ Indicates upregulated.
↓ Indicates downregulated.
TABLE 3.
Dysregulated metabolites associated with different treatments in plasma metabolomicsa
| Metabolite | Pathway | Classification | Def + TRP + HIFM + HRV | Def + EcN + HIFM + HRV | Def + EcN + TRP + HIFM + HRV | Suf + HIFM + HRV |
|---|---|---|---|---|---|---|
| d-glucose | Glycolysis, gluconeogenesis, | Carbohydrates | −0.9 | −0.4 | −0.8 | 5.5 |
| Deoxyribose | Pentose phosphate pathway | Carbohydrate | −0.5 | −0.9 | −0.7 | 1.7 |
| S-adenosylhomocysteine; S-adenosyl-l-homocysteine | Cysteine and methionine metabolism, biosynthesis of amino acids | Organic compound | 1.2 | 1.8 | −0.5 | 2.9 |
| Heme; protoheme; heme B; protoheme IX | Biosynthesis of secondary metabolites, ABC transporters, ferroptosis, mineral absorption | Organic compound | 8.7 | −06 | 1.3 | −0.1 |
| (GalNAc)2 (GlcA)1 (LIdoA)1 (S)3 | Glycosaminoglycan degradation | Organic compound | 1.4 | −0.8 | 1.0 | −0.6 |
| Cytidine-5′-monophosphate; CMP; cytidylic acid | Pyrimidine metabolism | Nucleotide | 1.3 | 3.4 | 2.9 | 4.1 |
| Nicotinic acid adenine dinucleotide | Nicotinate and nicotinamide metabolism | Nucleotide | 87 | 1,596 | 690 | 0 |
| Inosine | Purine metabolism | Nucleotide | 2.8 | 15 | 81.5 | 6.8 |
| Deoxythymidine 5′-diphosphate | Pyrimidine metabolism | Nucleotide | 1.4 | 1.0 | 1.3 | 1.0 |
| Xanthine | Purine metabolism | Nucleotide | 1.5 | 1.2 | 1.6 | 1.4 |
| Gentisic acid | Benzoate degradation | Benzenoids | 2.7 | 1.2 | 2.5 | 2.0 |
| Benzo[a]pyrene-7,8-diol | Metabolism of xenobiotics by cytochrome P450 | Benzenoids | 1.4 | 1.1 | 1.3 | 1.0 |
| 2-Methylacetoacetyl-CoA | Valine, leucine, and isoleucine degradation | Amino acid | 445 | 8,802 | 2,156 | 13 |
| N-acetylserotonin; N-acetyl-5-hydroxytryptamine | Tryptophan metabolism | Amino acid | 1.5 | 3.1 | 5.2 | 3.9 |
| Sphinganine | Sphingolipid metabolism | Lipid | −0.5 | −0.4 | 2.0 | −0.1 |
| Glycerophosphoethanolamine; sn-glycero-3-phosphoethanolamine | Glycerophospholipid metabolism, ether lipid metabolism | Lipid | −0.7 | 1.1 | −0.9 | 1.2 |
| Leukotriene C4 | Arachidonic acid metabolism, asthma | Lipid | 1,644 | 6,654 | 1,517 | 18 |
| Eicosanoic acid; icosanoic acid; arachidic acid | Biosynthesis of unsaturated fatty acids | Lipid | 2.4 | 1.0 | 1.9 | -0.4 |
| Retinoyl glucuronide;all-trans-retinoyl-beta-glucuronide | Retinol metabolism | Lipid | 2.0 | 1.3 | 2.0 | 6.3 |
| Phytosphingosine; 4-d-Hydroxysphinganine | Sphingolipid metabolism | Lipid | 549 | 3,931 | 1,020 | 645 |
| Cholesterol sulfate; Cholest-5-en-3beta-ol sulfate | Steroid hormone biosynthesis | Lipid | 1.2 | 1.8 | 2.1 | 1.0 |
Gnotobiotic (Gn) pigs were transplanted with human infant fecal microbiota (HIFM) at 4 days of age, post-HIFM transplantation day (PBTD) 0. Pigs were fed deficient (Def) and/or sufficient (Suf) diets. Pigs were supplemented orally with E. coli Nissle (EcN) 1917 and/or tryptophan (TRP) at PBTD7, subsequently challenged with virulent human rotavirus (VirHRV) at PBTD9-10/postchallenge day (PCD) 0, and euthanized on PBTD24/PCD14. Treatment groups were compared with untreated (Def) control group pigs and presented as fold change. A negative sign represents decreased relative intensity. Metabolites highlighted in bold are correlated with immunological assays, determined by Spearman’s nonparametric correlation method.
FIG 4.
Principal-component analysis (PCA) of plasma/fecal metabolomics and lipidomics. Gnotobiotic (Gn) pigs were cesarian-derived and transplanted with human infant fecal microbiota (HIFM) at 4 days of age, post-HIFM transplantation day (PBTD) 0. Pigs were fed a deficient (Def) and/or sufficient (Suf) diet, supplemented orally with Escherichia coli Nissle 1917 (EcN) and/or tryptophan (TRP) at PBTD7, subsequently challenged with VirHRV at PBTD9-10/postchallenge day (PCD) 0, and euthanized on PBTD24/PCD14. PM, plasma metabolomics; PL, plasma lipidomics; FM, fecal metabolomics; FL, fecal lipidomics.
FIG 5.
Heatmap of significant metabolites of plasma/fecal metabolomics and lipidomics. Heatmap visualizes the intensity/view of metabolites. More intense color represents the higher magnitude while the less intense color represents the lower magnitude of metabolite. In addition, the magnitude of metabolite is arranged to show their relatedness in the cluster heatmap. Gnotobiotic (Gn) pigs were cesarian derived and transplanted with human infant fecal microbiota (HIFM) at 4 days of age, post-HIFM transplantation day (PBTD) 0. Pigs were fed a deficient (Def) and/or sufficient (Suf) diet, supplemented orally with Escherichia coli Nissle 1917 (EcN) and/or tryptophan (TRP) at PBTD7, and, subsequently challenged with VirHRV at PBTD9-10/postchallenge day (PCD) 0, and euthanized on PBTD24/PCD14. PM, plasma metabolomics; PL, plasma lipidomics; FM, fecal metabolomics; FL, fecal lipidomics.
Excel file, relative intensities of dysregulated metabolites. Download Table S1, XLSX file, 0.02 MB (21KB, xlsx) .
Copyright © 2022 Michael et al.
This content is distributed under the terms of the Creative Commons Attribution 4.0 International license.
A decreased concentration (based on relative intensity) of d-glucose and deoxyribose was observed among all treatment groups compared with untreated Def group (Table 3). Single EcN treatment increased the intensity of the following metabolites: S-adenosyl-l-homocysteine, cytidine-5′-monophosphate, nicotinic acid adenine dinucleotide, glycerophosphoethanolamine, leukotriene C4, 2-Methylacetoacetyl-CoA, and phytosphingosine. While single TRP treatment increased the relative intensity of heme, deoxythymidine 5′-diphosphate, gentisic acid, eicosanoic acid, benzo[a]pyrene-7,8-diol/benzo[a]pyrene-7,8-dihydrodiol, and (GalNAc)2 (GlcA)1 (LIdoA)1 (S).
The combined EcN + TRP treatment increased the relative intensity of inosine, xanthine, sphinganine, N-acetylserotonin/N-acetyl-5-hydroxytryptamine, and cholesterol sulfate. Interestingly, TRP and EcN + TRP treatment equally increased the relative intensity of retinoyl glycuronide.
A total of 4,765 putative metabolites were detected in fecal metabolomics (FM) analysis; however, only 17 metabolites were found significant (P < 0.005) (Table 4). Multivariate analysis by PCA plot and a heatmap revealed segregation of the treatment groups compared with the untreated (Def) group in FM analysis (Fig. 4 and 5 and Table S1). PCA identified metabolites among treatments/untreated groups in FM samples. Clear discrimination of metabolites was observed in Def pigs revealing over 10% variance at component 2. Malnourished piglets (Def group) supplemented with EcN + TRP, EcN, and TRP had the highest resemblance of metabolites with untreated Suf diet group pigs with over 8% variance at component 3.
TABLE 4.
Dysregulated metabolites in fecal metabolomicsa
| Metabolite | Pathway | Classification | Def + TRP + HIFM + VirHRV | Def + EcN + HIFM + VirHRV | Def + EcN + TRP + HIFM + VirHRV | Suf + HIFM + VirHRV |
|---|---|---|---|---|---|---|
| N-acetylputrescine | Arginine and proline metabolism | Organic compound | −0.2 | −0.3 | −0.5 | −0.9 |
| Coproporphyrinogen III | Porphyrin and chlorophyll metabolism | Organic compound | −0.2 | −0.5 | −0.1 | −0.1 |
| d-urobilinogen | Biosynthesis of secondary metabolites | Organic compound | −0.1 | −0.6 | −0.2 | −0.1 |
| Phenylacetylglycine | Phenylalanine metabolism | Organic compound | −0.6 | −0.8 | 1.4 | −0.6 |
| Cytidine | Pyrimidine metabolism | Nucleotide | −0.7 | 1.5 | 2.3 | 1.1 |
| Hypoxanthine; urine-6-ol | Purine metabolism | Nucleotide | −0.1 | 1.0 | 1.0 | 1.2 |
| l-phenylalanine | Phenylalanine | Amino acid | −0.4 | −0.3 | −0.5 | −0.2 |
| l-arginine; (S)-2-amino-5-guanidinovaleric acid | Arginine biosynthesis, biosynthesis of amino acids | Amino acid | 3.6 | 5.0 | 27 | 7.2 |
| N-acetylserotonin; N-acetyl-5-hydroxytryptamine | Tryptophan metabolism | Amino acid | −0.5 | −0.6 | −0.1 | −0.5 |
| Phytosphingosine; 4-d-hydroxysphinganine | Sphingolipid metabolism | Lipid | 2.2 | 1.6 | 3.5 | 11 |
| 5,6-DHET; (8Z,11Z,14Z)-5,6-dihydroxyeicosa-8,11,14-trienoic acid; (8Z,11Z,14Z)-5,6- | Arachidonic acid metabolism | Lipid | 1.6 | 1.4 | 2.6 | 1.8 |
| l-palmitoylcarnitine | Fatty acid degradation and metabolism | Lipid | −0.9 | 1.3 | −0.4 | 4.6 |
| Glycocholate; glycocholic acid; | Primary and secondary bile acid biosynthesis | Lipid | 30 | 1.5 | −0.4 | 5.2 |
| 2-Methoxy-estradiol-17beta 3-glucuronide | Steroid hormone biosynthesis | Lipid | −0.1 | −0.4 | −0.1 | −0.1 |
| Glycerophosphoethanolamine | Glycerophospholipid metabolism, lipid metabolism | Lipid | −0.5 | 1.0 | 2.0 | −0.9 |
| Gamma-butyrobetaine | Lysine degradation | Lipid | −0.3 | −0.4 | 2.1 | −0.3 |
| Presqualene diphosphate | Sesquiterpenoid and triterpenoid biosynthesis |
Lipid | −0.6 | −0.6 | 1.1 | 1.0 |
Gnotobiotic (Gn) pigs were transplanted with human infant fecal microbiota (HIFM) at 4 days of age, post-HIFM transplantation day (PBTD) 0. Pigs were fed deficient (Def) and/or sufficient (Suf) diet. Pigs were supplemented orally with E. coli Nissle (EcN) 1917 and/or tryptophan (TRP) at PBTD7, subsequently challenged with virulent human rotavirus (VirHRV) at PBTD9-10/postchallenge day (PCD) 0, and euthanized on PBTD24/PCD14. Treatment groups were compared with untreated (Def) group pigs and presented as fold change. A negative sign represents decreased relative intensity. Metabolites highlighted in bold are correlated with immunological assays, determined by Spearman’s nonparametric correlation method.
A decreased relative intensity of metabolites was observed among all treated groups compared with the untreated Def pigs, including N-acetylputrescine, l-phenylalanine, N-acetylserotonin/N-acetyl-5-hydroxytryptamine, 2-methoxy-estradiol-17beta 3-glucuronide, urobilinogen, and coproporphyrinogen III. However, combined EcN + TRP treatment also increased the relative intensity of the following metabolites glycerophosphoethanolamine; phenylacetylglycine; l-arginine; phytosphingosine; gamma-butyrobetaine; (8Z,11Z,14Z)-5,6-dihydroxyeicosa-8,11,14-trienoic acid; and presqualene diphosphate. Similarly, a single treatment of EcN increased the relative intensity of l-palmitoylcarnitine. Interestingly, EcN and EcN +TRP treatment equally increased the relative intensity of hypoxanthine/purine-6-ol.
Individual treatment with either EcN or TRP increased the relative intensity of phytosphingosine while combined EcN + TRP treatment profoundly increased the relative intensity of the same metabolite. A single treatment with EcN and TRP increased and decreased, respectively, while combined EcN + TRP treatment profoundly increased the relative intensity of cytidine. A single treatment with TRP profoundly increased the relative intensity of glycocholate/glycocholic acid. N-acetylserotonin, phytosphingosine, and glycerophosphoethanolamine metabolites were identified commonly in PM and FM samples.
Untargeted plasma and fecal lipidomics revealed differentially altered metabolites among EcN, TRP, and EcN + TRP treatment groups.
A total of 1,084 putative metabolites were detected in plasma lipidomics (PL) analysis. Among those, only three metabolites were found to differ significantly (P < 0.005) (Table 5 and Table S1). Multivariate analysis by PCA plot and a heatmap revealed segregation of treatment groups from the untreated group in PL analysis (Fig. 4 and 5). PCA plot categorized metabolites among treatments/untreated groups in PL samples. Clear segregation of metabolites was observed for Def diet group pigs expanding over 13% variance at component 2. Malnourished piglets supplemented with EcN + TRP, EcN, and TRP groups had closely resembled metabolites with untreated Suf group pigs over 16% variance at component 1.
TABLE 5.
Dysregulated metabolites in plasma lipidomicsa
| Metabolite | Pathway | Classification | Def + TRP + HIFM + VirHRV | Def + EcN + HIFM + VirHRV | Def + EcN + TRP + HIFM + VirHRV | Suf + HIFM + VirHRV |
|---|---|---|---|---|---|---|
| N-acetylserotonin; N-acetyl-5-hydroxytryptamine | Tryptophan metabolism | Amino acid | −0.9 | 1.7 | 3.6 | 1.9 |
| Retinoyl glucuronide; all-trans-retinoyl-beta-glucuronide | All-trans-retinoyl-beta-glucuronide | Lipid | −0.9 | −0.8 | −0.8 | −0.8 |
| Cholesterol sulfate; cholest-5-en-3beta-ol sulfate | Steroid hormone biosynthesis | Lipid | 1.1 | −0.9 | −0.7 | −0.9 |
Gnotobiotic (Gn) pigs were transplanted with human infant fecal microbiota (HIFM) at 4 days of age, post-HIFM transplantation day (PBTD) 0. Pigs were fed deficient (Def) and/or sufficient (Suf) diets. Pigs were supplemented orally with E. coli Nissle (EcN) 1917 and/or tryptophan (TRP) at PBTD7, subsequently challenged with virulent human rotavirus (VirHRV) at PBTD9-10/postchallenge day (PCD) 0, and euthanized on PBTD24/PCD14. Treatment groups were compared with untreated (Def) group pigs and presented as fold change. A negative sign represents the decreased relative intensity. Metabolites highlighted in bold are correlated with immunological assays, determined by Spearman’s nonparametric correlation method.
Analysis revealed that single TRP treatment increased the relative intensity of cholesterol sulfate compared to untreated (Def) group pigs. Moreover, EcN and EcN + TRP treatment increased the relative intensity of N-acetylserotonin/N-acetyl-5-hydroxytryptamine. Lastly, retinoyl glucuronide relative intensity was decreased among all treatment groups compared with the untreated group.
A total of 3,517 putative metabolites were detected in fecal lipidomics (FL) analysis. Among those, nine metabolites differed significantly (P < 0.005) (Table 6 and Table S1). Multivariate analysis by PCA plot and a heatmap revealed segregation of treatment groups compared with the untreated group in FL analysis (Fig. 4 and 5). PCA plot revealed that TRP-treated pig and Suf group metabolites were similar compared to the other treatment groups in FL samples. Untreated (Def) and other treatment group metabolites overlapped with each other.
TABLE 6.
Dysregulated metabolites in fecal lipidomicsa
| Metabolite | Pathway | Classification | Def + TRP + HIFM + VirHRV | Def + EcN + HIFM + VirHRV | Def + EcN + TRP + HIFM + VirHRV | Suf + HIFM + VirHRV |
|---|---|---|---|---|---|---|
| THF-l-glutamate; tetrahydropteroyldiglutamate; (6S)-H4PteGlu2 | Folate biosynthesis | Organic compound | 3.8 | 36.4 | 12.7 | 5.5 |
| Biliverdin; biliverdin IX alpha | Biosynthesis of secondary metabolites | Organic compound | 79,714 | 222,806 | 263,611 | 71,308 |
| Anthranilate; anthranilic acid; o-aminobenzoic acid | Aminobenzoate degradation | Benzenoids | 1.5 | 1.9 | 1.7 | 0.8 |
| N-acetylserotonin; N-acetyl-5-hydroxytryptamine | Tryptophan metabolism | Amino acid | 3.0 | 9.4 | 3.8 | 3.9 |
| Corticosterone; 11beta,21-dihydroxy-4-pregnene-3,20-dione | Steroid hormone biosynthesis | Lipid | 1.3 | 1.7 | 1.4 | 2.5 |
| Phytosphingosine; 4-d-hydroxysphinganine | Sphingolipid metabolism | Lipid | 1.1 | 1.2 | 1.4 | 1.1 |
| Cholesterol sulfate; cholest-5-en-3beta-ol sulfate | Steroid hormone biosynthesis | Lipid | 1.5 | 1.9 | 3.7 | 1.6 |
| l-palmitoylcarnitine | Fatty acid degradation | Lipid | 1,997 | 14,425 | 4,426 | 6,100 |
| Glycocholate; glycocholic acid; 3alpha,7alpha,12alpha-trihydroxy-5beta-cholan-24-oylglycine | Primary and secondary bile acid biosynthesis, cholesterol metabolism, Bile secretion | Lipid | 2.1 | 1.9 | 1.9 | 1.7 |
Gnotobiotic (Gn) pigs were transplanted with human infant fecal microbiota (HIFM) at 4 days of age, post-HIFM transplantation day (PBTD) 0. Pigs were fed deficient (Def) and/or sufficient (Suf) diets. Pigs were supplemented orally with E. coli Nissle (EcN) 1917 and/or tryptophan (TRP) at PBTD7, subsequently challenged with virulent human rotavirus (VirHRV) at PBTD9-10/postchallenge day (PCD) 0, and euthanized on PBTD24/PCD14. Treatment groups were compared with untreated (Def) group pigs and presented as fold change. A negative sign represents the decreased relative intensity. Metabolites highlighted in bold are correlated with immunological assays, determined by Spearman’s nonparametric correlation method.
Analysis revealed that single EcN treatment increased the relative intensity of N-acetylserotonin/N-acetyl-5-hydroxytryptamine, corticosterone, tetrahydropteroyldiglutamate, l-palmitoylcarnitine, and aminobenzoic acid. Moreover, the analysis revealed that a single TRP treatment increased glycocholate/glycocholic acid. Finally, EcN + TRP treatment increased the relative intensity of phytosphingosine, cholesterol sulfate, and biliverdin.
Interestingly, in addition to the metabolites associated with lipids; the metabolites associated with amino acids, organic compounds, and benzenoids were also detected in the lipidomics analysis (Tables 5 and 6). N-acetylserotonin and cholesterol sulfate metabolites were identified in both PL and FL samples.
DISCUSSION
The multifactorial pathobiology of malnutrition is associated with perturbed intestinal homeostasis, epithelial dysfunction, altered metabolism, impaired immunity, intestinal inflammation, and malabsorption (39, 40). Impaired immune defense compromises gut integrity and causes a microbial imbalance that can predispose malnourished hosts to intestinal infections (41). These concerns require further research to improve the condition and treatment options of malnourished hosts.
EcN ± TRP supplementation enhanced immune responses.
Using a malnourished HIFM-transplanted Gn pig model of VirHRV infection, we evaluated the effects of treatment with EcN and/or TRP, and EcN + TRP on multiple aspects of the immune response and clinical parameters of the enteric disease. Our results suggested that supplementation have resulted in reduced diarrhea in HIFM piglets. Malnutrition-associated enteric dysfunction contributes to persistent systemic and intestinal inflammation that further exacerbates physiological and intestinal abnormalities, thus negatively affecting growth (39, 42, 43). In this study, a significant weight gain in EcN + TRP-treated pigs was observed compared to untreated Def pigs suggesting the restoration of intestinal homeostasis. In addition, EcN/TRP supplementation may have contributed to the energy absorption that led to increased survival rates. Concurrent EcN + TRP treatment increased systemic activated T regs that are associated with higher serum immunoregulatory cytokine levels. This could have contributed to reduced proinflammatory serum cytokine levels indicative of an anti-inflammatory microenvironment, thus reducing the severity of HRV disease (44–46). Moreover, enhanced immunoregulatory cytokines could have also contributed to increased HRV-specific IgA responses observed in EcN + TRP-treated pigs through TLR9 signaling (47–53). Interestingly, the relative intensity of deoxyribose was reduced (although not significantly) in treated Def pigs compared with Suf pigs in PM samples, as deoxyribose act as an agonist and activates TLR9 (54). In addition, increased serum levels of IFN-γ and TNF-α (an indicator of inflammation) in untreated malnourished pigs and reduced levels of these cytokines in EcN + TRP-, EcN-, and TRP-treated pigs are suggestive of moderation of inflammation (11, 12, 26, 55–57). We previously demonstrated that protein malnutrition altered innate immunity in HRV-infected neonatal Gn pigs (26). This study suggests that EcN + TRP supplementation (i) can inhibit the proapoptotic effects of HRV infection by inhibiting TLR-mediated proapoptotic signaling or by activating antiapoptotic pathways (10), and (ii) support the adequate adaptive immune function (38, 58) and programmed cell death by enhancing innate immune functions (53, 59–64).
Untargeted global metabolomics revealed alterations in both plasma and fecal metabolites and metabolic pathways that may have contributed to prophylactic diarrheal protection. Our results further highlight those metabolites/compounds that may be associated with protection against HRV infection related to increased production of gut-protective immunomodulatory, and anti-HRV metabolites/compounds. Moreover, several metabolites were correlated with immune responses (Table S2).
Excel file, correlations of immune responses with significantly altered metabolites in plasma and fecal metabolomic and lipidomic analysis. Download Table S2, XLSX file, 0.01 MB (14.3KB, xlsx) .
Copyright © 2022 Michael et al.
This content is distributed under the terms of the Creative Commons Attribution 4.0 International license.
EcN ± TRP supplementation altered amino acid-associated metabolites.
TRP is an essential amino acid in protein metabolism and the precursor of serotonin. TRP homeostasis and catabolism are disrupted by a protein deficiency that affects adaptive immune responses in Gn pigs while TRP supplementation enhances the frequencies of T-regulatory cells in pigs (15). Moreover, it was demonstrated that malnourished hosts were unable to catabolize TRP indicating protein turnover and the TRP degradation pathway was suppressed (65). In our current study, N-acetylserotonin/N-acetyl-5-hydroxytryptamine (involved in TRP metabolism) intensity was increased in EcN (FL and EcN) + TRP (PM and PL)-treated malnourished pigs. These metabolites are involved in adaptive immune responses suggesting that EcN and TRP supplementation may have improved or restored amino acid and protein homeostasis, which coincided with enhanced immunological parameters. Methylacetoacetyl-CoA is an intermediate in the degradation of isoleucine. Dietary isoleucine improved the growth performance and immune function via activation of pattern recognition receptor signaling pathways in RV-infected piglets (66). In our study, methylacetoacetyl-CoA intensity was substantially increased in EcN (PM)-treated pigs, indicating the role of EcN in enhancing immune responses, and it may also lead to the weight gain of malnourished piglets. Phenylalanine is an essential amino acid and is implicated in several body functions. However, during infections or inflammatory conditions, phenylalanine is significantly increased in serum (67). In addition, phenylalanine is excreted in greater amounts by stunted children suggesting a functional modulation of the gut immune/microbiome due to increased proteolytic activity (68). Our data demonstrated that phenylalanine intensity was decreased among all treated pigs compared with untreated Def pigs in the FM analysis. This is indicative of immune modulation in the host gastrointestinal system as it correlates with enhanced immunological responses.
EcN ± TRP supplementation altered benzenoid-associated metabolites.
Aminobenzoic acid belongs to the family of benzoic acid, and it is known to improve gut functions via regulating enzyme activity, redox status, immunity, and microbiota (69). In this study, EcN treatment increased aminobenzoic acid levels in FL. This suggests that EcN supplementation enhanced nutrient digestion, absorption, and barrier function of the malnourished piglets (11). Gentisic acid, a metabolite of aspirin, inhibits prostaglandin (involved in inflammatory responses) formation in response to LPS (70). Our data revealed that TRP treatment increased gentisic acid intensity in plasma. This suggests that TRP supplementation may have anti-inflammatory components, and thus, it modulates adaptive and innate immune responses and protects against HRV diarrhea in malnourished hosts.
EcN ± TRP supplementation altered organic compound-associated metabolites.
The elevated intensity of homocysteine has been implicated in intestinal inflammation (71). Moreover, increased serum homocysteine levels in inflammatory bowel syndrome in rats led to intestinal epithelial barrier dysfunction (72). Adenosylhomocysteine is the biosynthetic precursor to homocysteine, a metabolic indicators of cellular methylation and a biomarker of cardiovascular disease. It is decreased in the EcN + TRP-treated pigs in PM indicating that the supplementation may have reduced oxidative stress, decreased inflammation, lowered permeability, and modulated the mucosal immune system in the intestinal tract of neonatal pigs (73, 74). EcN + TRP treatment modulates adenosylhomocysteine that may have favorably shaped the mucosal immune response and enteric system to protect enterocytes and gut microbiota from oxidative damage and maintain normal gut motility during HRV infection and diarrhea (11, 73–75). Moreover, reducing inflammation in the gastrointestinal tract may provide the host with improved barrier function (76, 77). Biliverdin, a product of heme catabolism, can act as a cytoprotective and anti-inflammatory molecule. Recent data have shown that biliverdin is involved in innate immune responses resulting from acute insult and injury (78). Malnutrition can cause a decreased intensity of heme oxygenase activity leading to decreased biliverdin intensity as observed in FL samples of untreated pigs. However, EcN + TRP treatment increased the levels of biliverdin in FL samples, indicating an improved innate immune response and nutritional status that may have led to an increased survival rate and weight gain in the malnourished pigs. Coproporphyrinogen III is an intermediate metabolite in the normal biosynthesis of heme. This compound is not normally produced by the human body in a normal state; however, it is produced during protein-calorie malnutrition (79). Our current results show that coproporphyrinogen levels were decreased in all treatment groups in FM analysis. We hypothesized that EcN ± TRP treatment increased the ability of the liver to convert coproporphyrin to protoporphyrin (which acts as a precursor to heme/hemoglobin) by enhancing the enzyme coproporphyrinogen oxidase. This in turn improved adaptive and innate immune responses and nutritional status of the malnourished piglets and thus decreased diarrhea and virus shedding. Urobilinogen is a colorless by-product of bilirubin reduction, and increased levels are implicated in several liver diseases. Previous studies have shown that malnutrition induces gut atrophy and impaired small intestinal epithelial barrier in malnourished pigs, which may have resulted in the elevation of bilirubin (26, 80). Our current data demonstrate that all treatment groups had decreased urobilinogen levels in FM relative to the untreated Def group, indicating that the treatments may have protected the intestinal epithelium maintaining the absorptive function and thus decreasing the severity of diarrhea in HRV infection (11). These findings are in agreement with previous studies that EcN/TRP supplementation enhanced the immune responses in malnourished pigs (15, 48). Phenylacetylglycine is a putative biomarker of phospholipidosis, and its metabolic activity is disrupted by malnutrition (68), while EcN + TRP enhanced its levels in FM, indicative of enhanced adaptive and innate immune function.
EcN ± TRP supplementation altered lipid-associated metabolites.
Sphingolipids show the potential to impact chronic inflammatory diseases through inhibiting intestinal lipid absorption, altering the gut microbiota, activation of anti-inflammatory nuclear receptors, and LPS neutralization (81). Our current data show that EcN + TRP treatment increased the sphinganine levels in PM analysis, thus indicating improved adaptive and innate immune responses. Phytosphingosine exhibits anti-inflammation and antimicrobial activities; in addition, it stimulated epidermal differentiation through PPAR activation (82). Similarly, like other sphingolipids, EcN + TRP treatment increased phytosphingosine intensity in FM and FL while EcN treatment increased it in PM analysis. This suggests EcN + TRP and/or TRP enhanced lipid metabolism, thus ameliorating HRV infection. Arachidonic acid is a precursor of inflammatory mediators and dysregulated during protein-calorie malnutrition due to perturbation of essential fatty acid metabolism (83). In this study, EcN treatment increased leukotriene (a metabolite of arachidonic acid) intensity relative to untreated Def pig (indicating an impaired immune response) in PM analysis suggesting that such treatment reduced HRV disease and the inflammatory environment in malnourished hosts. Moreover, arachidonic acid is involved in the immune function of several organs and systems either directly or via conversion to eicosanoids (84, 85). Eiconoids exhibit a role in inflammation, protection of mucosal integrity of the gastrointestinal tract, and regulation of aggregation of platelets. In this study, TRP treatment increased levels of eicosanoids in PM samples indicating an enhancement of the immune function, inflammation, and maintenance of gut integrity. Cholesterol serves as the substrate for steroid hormone production and lower levels of cholesterol and is an indicator of protein-calorie malnutrition (86). In our current study, TRP (PL) and EcN + TRP (PM and FL) treatment groups had increased levels of cholesterol sulfate. This indicates that supplementation of EcN ± TRP has improved the nutritional status of malnourished pigs (15, 48). Gamma-butyrobetaine is an intermediate precursor of carnitine, and it promotes the development of functionally distinct microbial communities to optimize the metabolism of carnitine (87). This study shows that EcN + TRP treatment increased the intensities of gamma-butyrobetaine in FM analysis. Carnitine is an essential nutrient that plays a role in energy production and transport of activated fatty acids across the mitochondrial membrane during mitochondrial beta-oxidation, and its deficiency occurs in malnutrition (88). In this study, EcN treatment increased the levels of palmitoylcarnitine in FM and FL analysis. This indicates that EcN supplementation may have ameliorated malnutrition effects or total energy expenditure leading to increased survival rates and weight gain in malnourished piglets. Collectively, it suggests that there could be an important mechanism both locally and systemically for promoting diarrheal protection in EcN + TRP-treated pigs compared with untreated Def pigs.
EcN ± TRP supplementation altered nucleotide-associated metabolites.
Cellular nucleotides, represented by purines and pyrimidines, play an important role in the synthesis of the nucleic acids RNA and DNA. Nucleotides play a vital role in host cell metabolism and are essential for viral replication dependent on the host supply of nucleotide biosynthesis. Many inhibitors targeting de novo nucleotide biosynthesis have been studied extensively in dengue virus, hepatitis B virus, hepatitis E virus, and other virus infection models (89–96). Purine metabolism plays a principal role in cellular redox regulation and accelerated tissue ATP degradation, and it is significantly reduced with malnutrition (97). EcN ± TRP treatments increased hypoxanthine levels in FM while EcN + TRP treatment also increased inosine and xanthine levels in PM. This suggests that the supplementations upregulated purine metabolism that coordinated the actions of salvage and de novo biosynthetic pathways improving the immune and growth functions. Cytidine-5′-monophosphate is a pyrimidine base found in RNA that helps to promote new cell growth. EcN treatment increased cytidine-5′-monophosphate levels in plasma indicating that the EcN-mediated increase in cytidine-5′-monophosphate synthesis may in part explain improved weight gain in the treated group relative to untreated Def pigs. NAD is an essential pyridine nucleotide that serves as a cofactor and substrate for several critical cellular processes involved in oxidative phosphorylation and ATP production, DNA repair, epigenetically modulated gene expression, intracellular calcium signaling, and immunological functions (98). NAD depletion may occur due to excessive DNA damage due to free radicals, energy crisis and reduced ATP production, chronic increase in immune activation, and inflammatory cytokines production (99). We observed that EcN supplementation to Def pigs substantially increased NAD levels in plasma compared to other treatments. This suggests that EcN may act via tryptophanase that can lead to de novo NAD synthesis in mammalian cells (100), which may have led to improved immune activation, barrier function, and anti-inflammatory response.
Low vaccine efficacy and environmental enteric dysfunction exacerbate HRV prevalence and severity in developing countries necessitating optimization of the existing and/or development of alternative preventive approaches. The combination of EcN + TRP or individual (EcN or TRP) treatments exhibits the potential to serve as a safe, natural, prophylactic treatment aimed at reducing HRV diarrhea in malnourished hosts, and it is accomplished at least in part via modification of the host lipid, bile acid, steroid hormone, and amino acid/peptide metabolism. In addition, EcN and TRP metabolism may have exerted direct antiviral activity against HRV by interfering with HRV infection, replication, and pathogenesis, improving gut barrier function and modulating mucosal immune responses, especially inflammatory processes that influence the development of HRV diarrhea. Our data on direct modulation of the host metabolism and immunity indicate that while EcN and TRP may provide effective therapeutics against HRV, this combination likely provides broadened and enhanced protection possibly against other enteric pathogens.
In summary, our results suggest that the EcN probiotic and TRP combination conferred greater protection in most cases (rather than individual EcN and TRP treatments). Moreover, EcN + TRP treatment (compared with other treatment groups) mostly upregulated immunoregulatory while downregulating proinflammatory cytokines following VirHRV infection in malnourished humanized Gn piglets. Our results suggest that low-cost dietary supplementation of combined EcN and TRP can potently ameliorate HRV-associated diarrhea and possibly other enteric infections.
MATERIALS AND METHODS
Human infant fecal microbiota.
The collection and use of HIFM were approved by The Ohio State University (OSU) Institutional Review Board. With parental consent, sequential fecal samples were collected from a healthy, 2-month-old, exclusively breastfed, vaginally delivered infant. Samples were pooled and diluted to 1:20 (wt/vol) in PBS solution containing 0.05% (vol/vol) cysteine and 30% glycerol and stored at −80°C as described previously (15, 26).
Virus.
The Gn pig passaged virulent human rotavirus (VirHRV) Wa strain at pig passages 25 to 26 was used as a challenge virus at a dose of 1 × 106 focus-forming unit (FFU) per pig as described previously (15, 26).
Animal experimentation.
All animal experiments were approved by the Institutional Animal Care and Use Committee at the OSU. Near-term pregnant sows (Landrace × Yorkshire × Duroc cross-bred) from the OSU swine center facility were used to derive the Gn pigs by cesarean section, and the Gn pigs were maintained in sterile isolators (101). Neonatal pigs obtained from five litters (5 to 15 pigs/litter) were randomly assigned to the following groups: (i) deficient (Def) diet (n = 6), (ii) deficient (Def + EcN) diet with EcN 1 × 105 CFU/dose supplementation (n = 6), (iii) deficient (Def + TRP) diet with 0.4-g/day l-tryptophan (TRP) supplementation (n = 7), (iv) deficient (Def + EcN + TRP) diet with EcN and TRP combined supplementation (n = 7), and (v) sufficient (Suf) diet (n = 7). EcN and/or TRP supplementations were administered orally at 7 day following HIFM transplantation (Fig. 6). Deficient pigs were fed a diet of 50% Parmalat and 50% sterile water, which fell within NRC standards for calories, fat, and carbohydrates, while protein levels were half the recommended levels (7.5% versus 15% of diet). To confirm sterility from bacterial or fungal contamination before HIFM transplant, aerobic and anaerobic cultures of fecal swabs were performed. Additionally, all piglets were confirmed negative for the enteric viruses rotavirus, transmissible gastroenteritis virus, porcine epidemic diarrhea virus, calicivirus/sapovirus, astrovirus, and kobuvirus before HIFM inoculation. Four days postderivation, Gn pigs were inoculated orally with 2 ml of diluted HIFM stock, postbacterial transplantation day 0 (PBTD 0). Nine to 10 days were allowed for the establishment and stabilization of the fecal microbiota transplant. At PBTD 9 to 10, pigs were challenged with VirHRV (1 × 106 FFU, postchallenge day/PCD 0). All pigs were humanly euthanized at PBTD 24/PCD 14. Blood, spleen, duodenum, and ileum were collected to isolate mononuclear cells (MNCs) for subsequent immunologic assays (33, 47, 102, 103). Serum, small intestinal contents (SIC), and large intestinal contents (LIC) were collected to determine the impact of EcN, TRP, and Def diet on VirHRV infection.
FIG 6.
Experimental design showing time points for hysterectomy, human infant fecal microbiota (HIFM) transplantation, E. coli Nissle (EcN) 1917 probiotic/tryptophan (TRP) treatment, virulent human rotavirus (VirHRV) challenge, and euthanasia. PBTD, postbacterial transplantation day; PCD, PBTD; postchallenge.
Assessment of clinical signs.
Piglets were weighed and fecal swabs were collected post-VirHRV challenge. Fecal consistency was scored as follows: 0, normal; 1, pasty/semiliquid; and 2, liquid, and pigs with fecal scores more than 1 were considered diarrheic (10–12, 48, 58, 104, 105). Fecal swabs were suspended in 2 ml of minimum essential medium (MEM) (Life Technologies, Waltham, MA, USA), clarified by centrifugation at 800 × g for 10 min at 4°C, and stored at −20°C until quantification of infectious HRV by a cell culture immunofluorescence assay, as previously described (10–12, 48, 58, 104, 105).
Isolation of mononuclear cells and immunological analyses.
Systemic (blood, spleen) and intestinal cells (duodenum, ileum) were collected to isolate MNCs as described previously (33, 47, 102, 103). The purified MNCs were suspended in E-RPMI 1640. The viability of MNCs preparation was determined by trypan blue exclusion (≥95%). The HRV-specific antibody-secreting cells (ASCs) and HRV-specific antibody titers in serum and intestinal contents were quantified as previously described (11, 15, 47, 58, 106, 107). Freshly isolated MNCs were stained for quantification of different T-cell subsets: T-helper cells (CD3+CD4+), cytotoxic T cells (CD3+CD8+), and T-regulatory cells (CD4/CD8+CD25+FOXP3+) (102, 105). Frequencies of IgA+ B lymphocytes were determined by identifying CD79β and IgA expression in MNCs as reported previously (58). MNCs were also stained to assess frequencies of plasmacytoid dendritic cells (pDCs) CD103+ (SWC3a+CD4+). Toll-like receptor (TLR) expression on MNCs was assessed using monoclonal antibodies to porcine and human cell surface markers as reported previously (10, 58, 59, 108). TLR4 (ligand bacterial lipopolysaccharide), TLR3 (ligand double-stranded RNAs), and TLR9 (ligand bacterial CpGs) were assessed in our experiments. Frequencies of natural killer (NK) cells (SWC3a+CD16+) were also assessed among systemic MNCs (105). Appropriate isotype-matched irrelevant antibodies were included as negative controls. Subsequently, 50,000 events were acquired per sample using BD Accuri C6 flow cytometer (BD Biosciences, San Jose, CA, USA). Data were analyzed using C6 flow sampler software. To analyze NK cytotoxicity, total blood MNCs and K562 cells were used as effector and target cells, respectively. For the effector, target cell ratios of 10:1, 5:1, 1:1, and 0.5:1 were used, and the assay was performed as described previously (105, 109). The Annexin V Apoptosis Detection kit APC (eBiosciences, San Diego, CA) and Propidium Iodide Staining Solution (eBiosciences) were used according to the manufacturer protocols to detect and discriminate apoptotic and necrotic MNCs. Within 4 h after the staining, acquisition of 50,000 events was performed using Accuri C6 flow cytometer, and analyses were performed using CFlow software (10, 38).
Serum cytokines.
Serum samples were collected at the terminal time point and analyzed for proinflammatory (IL-1β, IL-6, TNF-α, IFN-α, and IFN-γ) and T-regulatory (IL-10 and TGF-β) cytokines as described previously with some modifications by increasing incubation timings of antibodies and samples (44, 102, 105).
Metabolomics and lipidomics analyses.
Plasma and fecal samples were prepared using the cold methanol extraction method, clarified up with 0.2-μm ultracentrifugation spin filters, and further diluted with water containing 0.1% formic acid and subjected to nontargeted metabolic profiling. Briefly, samples were analyzed by ultrahigh performance liquid chromatography/tandem mass spectrometry (UHPLC/MS/MS) with Thermo LTQ Orbitrap XL (Thermofisher, CA). Metabolites were identified by automated comparison of the ion features in the experimental samples to a reference library of chemical standard entries that included retention time, the mass-to-charge ratio (m/z), preferred adducts, as well as associated MS spectra, and were quality controlled using pooled sample alignment using the Progenesis QI metabolomics package.
Statistical analysis.
All statistical analyses were performed using GraphPad Prism version 6 (GraphPad Software, Inc., La Jolla, CA). Log10-transformed isotype ELISA antibody titers were analyzed using one-way ANOVA followed by Duncan’s multiple range test. Data represent the mean numbers of HRV-specific antibody-secreting cells per 5 × 105 MNCs and are analyzed using a nonparametric t test (Mann-Whitney). The mean total fecal score was from PCD1 to PCD6. Mean fecal scores were analyzed by two-way ANOVA followed by Bonferroni posttest. Correlation analysis was performed using Spearman’s nonparametric correlation method. Significant differences were as follows: *, P < 0.05; **, P < 0.01; and ***, P < 0.001. Error bars indicated the standard error of the mean. Metabolomics data were analyzed with both the Progenesis software and XCMS Online from the Scripps Metlin Database. Data were normalized by sum and autoscaled. Heatmap visualization was performed based on the reorganization of metabolites to show the contrast between the groups. Principal-component analysis (PCA) was used for classification analyses. Treatment groups were compared with untreated pig groups and are presented as fold change.
ACKNOWLEDGMENTS
This work was supported by the Bill and Melinda Gates Foundation (OPP 1117467); NIAID, NIH (R01 A1099451); NIH NICHD Grant R01 HD095881; federal and state funds appropriated to the Ohio Agricultural Research and Development Center, Ohio State University; and the NIH Office of Dietary Supplements (ODS) supplemental grant funds. We thank Food for Health (FFH) at the Ohio State University for providing a fellowship to H. Michael; FFH Ohio State University Discovery Theme Seed grant 2018 to G. Rajashekara; and Campus Chemical Instrument Center voucher (2019-CoRR5220) to G. Rajashekara.
We also thank Stephanie Langel, Ayako Miyazaki, Francine C. Paim, David D. Fischer, Marcia Lee, and Rosario Candelero-Rueda for their technical assistance and Juliette Hanson, Ronna Wood, Jeffery Ogg, Megan Strother, and Sara Tallmadge for animal care assistance.
The authors declared no conflict of interest.
Conceived and designed the experiments: A.N.V., L.J.S., and G.R. Data collection: H.M., V.S., and L.D. Analyzed the data: H.M., V.S., J.C., L.D., J.O.A. Wrote the paper: H.M. Critically revised the paper: A.N.V., L.J.S., and G.R.
Contributor Information
Linda J. Saif, Email: saif.2@osu.edu.
Gireesh Rajashekara, Email: rajashekara.2@osu.edu.
Anastasia N. Vlasova, Email: vlasova.1@osu.edu.
Vincent B. Young, University of Michigan-Ann Arbor
REFERENCES
- 1.Tate JE, Burton AH, Boschi-Pinto C, Steele AD, Duque J, Parashar UD, 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. doi: 10.1016/S1473-3099(11)70253-5. [DOI] [PubMed] [Google Scholar]
- 2.Lopman BA, Pitzer VE, Sarkar R, Gladstone B, Patel M, Glasser J, Gambhir M, Atchison C, Grenfell BT, Edmunds WJ, Kang G, Parashar UD. 2012. Understanding reduced rotavirus vaccine efficacy in low socio-economic settings. PLoS One 7:e41720. doi: 10.1371/journal.pone.0041720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Ferreira RB, Antunes LC, Finlay BB. 2010. Should the human microbiome be considered when developing vaccines? PLoS Pathog 6:e1001190. doi: 10.1371/journal.ppat.1001190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Qadri F, Bhuiyan TR, Sack DA, Svennerholm AM. 2013. Immune responses and protection in children in developing countries induced by oral vaccines. Vaccine 31:452–460. doi: 10.1016/j.vaccine.2012.11.012. [DOI] [PubMed] [Google Scholar]
- 5.Valdez Y, Brown EM, Finlay BB. 2014. Influence of the microbiota on vaccine effectiveness. Trends Immunol 35:526–537. doi: 10.1016/j.it.2014.07.003. [DOI] [PubMed] [Google Scholar]
- 6.Szajewska H, Mrukowicz JZ. 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):S17–25. doi: 10.1097/00005176-200110002-00004. [DOI] [PubMed] [Google Scholar]
- 7.Sanders ME, Guarner F, Guerrant R, Holt PR, Quigley EM, Sartor RB, Sherman PM, Mayer EA. 2013. An update on the use and investigation of probiotics in health and disease. Gut 62:787–796. doi: 10.1136/gutjnl-2012-302504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Kruis W, Fric P, Pokrotnieks J, Lukas M, Fixa B, Kascak M, Kamm MA, Weismueller J, Beglinger C, Stolte M, Wolff C, Schulze J. 2004. Maintaining remission of ulcerative colitis with the probiotic Escherichia coli Nissle 1917 is as effective as with standard mesalazine. Gut 53:1617–1623. doi: 10.1136/gut.2003.037747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Schultz M. 2008. Clinical use of E. coli Nissle 1917 in inflammatory bowel disease clinical use of E. coli Nissle 1917 in inflammatory bowel disease. Inflamm Bowel Dis 14:1012–1018. doi: 10.1002/ibd.20377. [DOI] [PubMed] [Google Scholar]
- 10.Vlasova AN, Shao L, Kandasamy S, Fischer DD, Rauf A, Langel SN, Chattha KS, Kumar A, Huang HC, Rajashekara G, Saif LJ. 2016. Escherichia coli Nissle 1917 protects gnotobiotic pigs against human rotavirus by modulating pDC and NK-cell responses. Eur J Immunol 46:2426–2437. doi: 10.1002/eji.201646498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Paim FC, Langel SN, Fischer DD, Kandasamy S, Shao L, Alhamo MA, Huang HC, Kumar A, Rajashekara G, Saif LJ, Vlasova AN. 2016. Effects of Escherichia coli Nissle 1917 and Ciprofloxacin on small intestinal epithelial cell mRNA expression in the neonatal piglet model of human rotavirus infection. Gut Pathog 8:66. doi: 10.1186/s13099-016-0148-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Kandasamy S, Vlasova AN, Fischer D, Kumar A, Chattha KS, Rauf A, Shao L, Langel SN, Rajashekara G, Saif LJ. 2016. Differential effects of Escherichia coli Nissle and Lactobacillus rhamnosus strain GG on human rotavirus binding, infection, and B cell immunity. J Immunol 196:1780–1789. doi: 10.4049/jimmunol.1501705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Guerrant RL, Leite AM, Pinkerton R, Medeiros PH, Cavalcante PA, DeBoer M, Kosek M, Duggan C, Gewirtz A, Kagan JC, Gauthier AE, Swann J, Mayneris-Perxachs J, Bolick DT, Maier EA, Guedes MM, Moore SR, Petri WA, Havt A, Lima IF, Prata MM, Michaleckyj JC, Scharf RJ, Sturgeon C, Fasano A, Lima AA. 2016. Biomarkers of environmental enteropathy, inflammation, stunting, and impaired growth in children in Northeast Brazil. PLoS One 11:e0158772. doi: 10.1371/journal.pone.0158772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Semba RD, Shardell M, Sakr Ashour FA, Moaddel R, Trehan I, Maleta KM, Ordiz MI, Kraemer K, Khadeer MA, Ferrucci L, Manary MJ. 2016. Child stunting is associated with low circulating essential amino acids. EBioMedicine 6:246–252. doi: 10.1016/j.ebiom.2016.02.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Fischer DD, Kandasamy S, Paim FC, Langel SN, Alhamo MA, Shao L, Chepngeno J, Miyazaki A, Huang HC, Kumar A, Rajashekara G, Saif LJ, Vlasova AN. 2017. Protein malnutrition alters tryptophan and angiotensin-converting enzyme 2 homeostasis and adaptive immune responses in human rotavirus-infected gnotobiotic pigs with human infant fecal microbiota transplant. Clin Vaccine Immunol 24:e00172-17. doi: 10.1128/CVI.00172-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Hashimoto T, Perlot T, Rehman A, Trichereau J, Ishiguro H, Paolino M, Sigl V, Hanada T, Hanada R, Lipinski S, Wild B, Camargo SM, Singer D, Richter A, Kuba K, Fukamizu A, Schreiber S, Clevers H, Verrey F, Rosenstiel P, Penninger JM. 2012. ACE2 links amino acid malnutrition to microbial ecology and intestinal inflammation. Nature 487:477–481. doi: 10.1038/nature11228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Mehraj V, Routy JP. 2015. Tryptophan catabolism in chronic viral infections: handling uninvited guests. Int J Tryptophan Res 8:41–48. doi: 10.4137/IJTR.S26862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Nzakizwanayo J, Dedi C, Standen G, Macfarlane WM, Patel BA, Jones BV. 2015. Escherichia coli Nissle 1917 enhances bioavailability of serotonin in gut tissues through modulation of synthesis and clearance. Sci Rep 5:17324. doi: 10.1038/srep17324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bansal T, Alaniz RC, Wood TK, Jayaraman A. 2010. The bacterial signal indole increases epithelial-cell tight-junction resistance and attenuates indicators of inflammation. Proc Natl Acad Sci USA 107:228–233. doi: 10.1073/pnas.0906112107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Butler JE, Lager KM, Splicha l, Francis D, Kacskovics I, Sinkora M, Wertz N, Sun J, Zhao Y, Brown WR, DeWald R, Dierks S, Muyldermans S, Lunney JK, McCray PB, Rogers CS, Welsh MJ, Navarro P, Klobasa F, Habe F, Ramsoondar J. 2009. The piglet as a model for B cell and immune system development. Vet Immunol Immunopathol 128:147–170. doi: 10.1016/j.vetimm.2008.10.321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Yuan L, Saif LJ. 2002. Induction of mucosal immune responses and protection against enteric viruses: rotavirus infection of gnotobiotic pigs as a model. Vet Immunol Immunopathol 87:147–160. doi: 10.1016/S0165-2427(02)00046-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Saif LJ, Ward LA, Yuan L, Rosen BI, To TL. 1996. The gnotobiotic piglet as a model for studies of disease pathogenesis and immunity to human rotaviruses. Arch Virol Suppl 12:153–161. doi: 10.1007/978-3-7091-6553-9_17. [DOI] [PubMed] [Google Scholar]
- 23.Hammerberg C, Schurig GG, Ochs D. 1989. Immunodeficiency in young pigs. J Vet Res 50:868–874. [PubMed] [Google Scholar]
- 24.Wagstrom EA, Yoon KJ, Zimmerman JJ. 2000. Immune components in porcine mammary secretions. Viral Immunol 13:383–397. doi: 10.1089/08828240050144699. [DOI] [PubMed] [Google Scholar]
- 25.Zhang W, Wen K, Azevedo MSP, Gonzalez A, Saif LJ, Li G, Yousef AE, Yuan L. 2008. Lactic acid bacterial colonization and human rotavirus infection influence distribution and frequency of monocytes/macrophage s and dendritic cells in neonatal gnotobiotic pigs. Vet Immunol Immunopathol 121:222–231. doi: 10.1016/j.vetimm.2007.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Vlasova AN, Paim FC, Kandasamy S, Alhamo MA, Fischer DD, Langel SN, Deblais L, Kumar A, Chepngeno J, Shao L, Huang HC, Candelero-Rueda RA, Rajashekara G, Saif LJ. 2017. Protein malnutrition modifies innate immunity and gene expression by intestinal epithelial cells and human rotavirus infection in neonatal gnotobiotic pigs. mSphere 2:e00046-17. [ doi: 10.1128/mSphere.00046-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Kumar A, Vlasova AN, Deblais L, Huang HC, Wijeratne A, Kandasamy S, Fischer DD, Langel SN, Paim FC, Alhamo MA, Shao L, Saif LJ, Rajashekara G. 2018. Impact of nutrition and rotavirus infection on the infant gut microbiota in a humanized pig model. BMC Gastroenterol 18:93. doi: 10.1186/s12876-018-0810-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.De Preter V. 2015. Metabolomics in the clinical diagnosis of inflammatory bowel disease. Dig Dis 33(Suppl 1):2–10. doi: 10.1159/000437033. [DOI] [PubMed] [Google Scholar]
- 29.Kolho KL, Pessia A, Jaakkola T, de Vos WM, Velagapudi V. 2017. Faecal and serum metabolomics in paediatric inflammatory bowel disease. J Crohns Colitis 11:321–334. doi: 10.1093/ecco-jcc/jjw158. [DOI] [PubMed] [Google Scholar]
- 30.Williams HR, Willsmore JD, Cox IJ, Walker DG, Cobbold JF, Taylor-Robinson SD, Orchard TR. 2012. Serum metabolic profiling in inflammatory bowel disease. Dig Dis Sci 57:2157–2165. doi: 10.1007/s10620-012-2127-2. [DOI] [PubMed] [Google Scholar]
- 31.Dawiskiba T, Deja S, Mulak A, Zabek A, Jawien E, Pawelka D, Banasik M, Mastalerz-Migas A, Balcerzak W, Kaliszewski K, Skora J, Barc P, Korta K, Pormanczuk K, Szyber P, Litarski A, Mlynarz P. 2014. Serum and urine metabolomic fingerprinting in diagnostics of inflammatory bowel diseases. World J Gastroenterol 20:163–174. doi: 10.3748/wjg.v20.i1.163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Macpherson AJ, McCoy KD, Johansen FE, Brandtzaeg P. 2008. The immune geography of IgA induction and function. Mucosal Immunol 1:11–22. doi: 10.1038/mi.2007.6. [DOI] [PubMed] [Google Scholar]
- 33.Yuan L, Ward LA, Rosen BI, To TL, Saif LJ. 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. doi: 10.1128/jvi.70.5.3075-3083.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Azevedo MS, Yuan L, Iosef C, Chang KO, Kim Y, Nguyen TV, Saif LJ. 2004. Magnitude of serum and intestinal antibody responses induced by sequential replicating and nonreplicating rotavirus vaccines in gnotobiotic pigs and correlation with protection. Clin Vaccine Immunol 11:12–20. doi: 10.1128/CDLI.11.1.12-20.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Saif LJ, Yuan L, Ward LA, T√¥ TL. 1998. Serum and intestinal isotype antibody responses and correlates of protective immunity to human rotavirus in a gnotobiotic pig model of disease. J Gen Virol 79:2661–2672. doi: 10.1099/0022-1317-79-11-2661. [DOI] [PubMed] [Google Scholar]
- 36.Fernandez MF, Boris S, Barbes C. 2003. Probiotic properties of human lactobacilli strains to be used in the gastrointestinal tract. J Appl Microbiol 94:449–455. doi: 10.1046/j.1365-2672.2003.01850.x. [DOI] [PubMed] [Google Scholar]
- 37.Villena J, Medina M, Vintini E, Alvarez S. 2008. Stimulation of respiratory immunity by oral administration of Lactococcus lactis. Can J Microbiol 54:630–638. doi: 10.1139/W08-052. [DOI] [PubMed] [Google Scholar]
- 38.Vlasova AN, Chattha KS, Kandasamy S, Liu Z, Esseili M, Shao L, Rajashekara G, Saif LJ. 2013. Lactobacilli and bifidobacteria promote immune homeostasis by modulating innate immune responses to human rotavirus in neonatal gnotobiotic pigs. PLoS One 8:e76962. doi: 10.1371/journal.pone.0076962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Guerrant RL, Oria RB, Moore SR, Oria MO, Lima AA. 2008. Malnutrition as an enteric infectious disease with long-term effects on child development. Nutr Rev 66:487–505. doi: 10.1111/j.1753-4887.2008.00082.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Prendergast AJ, Kelly P. 2016. Interactions between intestinal pathogens, enteropathy and malnutrition in developing countries. Curr Opin Infect Dis 29:229–236. doi: 10.1097/QCO.0000000000000261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Ibrahim MK, Zambruni M, Melby CL, Melby PC. 2017. Impact of childhood malnutrition on host defense and infection. Clin Microbiol Rev 30:919–971. doi: 10.1128/CMR.00119-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Liu J, Bolick DT, Kolling GL, Fu Z, Guerrant RL. 2016. Protein malnutrition impairs intestinal epithelial cell turnover, a potential mechanism of increased cryptosporidiosis in a murine model. Infect Immun 84:3542–3549. doi: 10.1128/IAI.00705-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Prendergast AJ, Rukobo S, Chasekwa B, Mutasa K, Ntozini R, Mbuya MN, Jones A, Moulton LH, Stoltzfus RJ, Humphrey JH. 2014. Stunting is characterized by chronic inflammation in Zimbabwean infants. PLoS One 9:e86928. doi: 10.1371/journal.pone.0086928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Azevedo MS, Yuan L, Pouly S, Gonzales AM, Jeong KI, Nguyen TV, Saif LJ. 2006. Cytokine responses in gnotobiotic pigs after infection with virulent or attenuated human rotavirus. J Virol 80:372–382. doi: 10.1128/JVI.80.1.372-382.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Azim T, Ahmad SM, Sefat EK, Sarker MS, Unicomb LE, De S, Hamadani JD, Salam MA, Wahed MA, Albert MJ. 1999. Immune response of children who develop persistent diarrhea following rotavirus infection. Clin Diagn Lab Immunol 6:690–695. doi: 10.1128/CDLI.6.5.690-695.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Jiang B, Snipes-Magaldi L, Dennehy P, Keyserling H, Holman RC, Bresee J, Gentsch J, Glass RI. 2003. Cytokines as mediators for or effectors against rotavirus disease in children. Clin Vaccine Immunol 10:995–1001. doi: 10.1128/CDLI.10.6.995-1001.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Michael H, Langel SN, Miyazaki A, Paim FC, Chepngeno J, Alhamo MA, Fischer DD, Srivastava V, Kathayat D, Deblais L, Rajashekara G, Saif LJ, Vlasova AN. 2020. Malnutrition decreases antibody secreting cell numbers induced by an oral attenuated human rotavirus vaccine in a human infant fecal microbiota transplanted gnotobiotic pig model. Front Immunol 11:196. doi: 10.3389/fimmu.2020.00196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Michael H, Paim FC, Miyazaki A, Langel SN, Fischer DD, Chepngeno J, Goodman SD, Rajashekara G, Saif LJ, Vlasova AN. 2021. Escherichia coli Nissle 1917 administered as a dextranomar microsphere biofilm enhances immune responses against human rotavirus in a neonatal malnourished pig model colonized with human infant fecal microbiota. PLoS One 16:e0246193. doi: 10.1371/journal.pone.0246193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Ertesvag A, Naderi S, Blomhoff HK. 2009. Regulation of B cell proliferation and differentiation by retinoic acid. Semin Immunol 21:36–41. doi: 10.1016/j.smim.2008.06.005. [DOI] [PubMed] [Google Scholar]
- 50.Michael H, Paim FC, Langel SN, Miyazaki A, Fischer DD, Chepngeno J, Amimo J, Deblais L, Rajashekara G, Saif LJ, Vlasova AN. 2021. Escherichia coli Nissle 1917 enhances innate and adaptive immune responses in a ciprofloxacin-treated defined-microbiota piglet model of human rotavirus infection. mSphere 6:e00074-21.[ doi: 10.1128/mSphere.00074-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.O'Mahony C, Scully P, O'Mahony D, Murphy S, O'Brien F, Lyons A, Sherlock G, MacSharry J, Kiely B, Shanahan F, O'Mahony L. 2008. Commensal-induced regulatory T cells mediate protection against pathogen-stimulated NF-kappaB activation. PLoS Pathog 4:e1000112. doi: 10.1371/journal.ppat.1000112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Zhong Y, Cantwell A, Dube PH. 2010. Transforming growth factor beta and CD25 are important for controlling systemic dissemination following Yersinia enterocolitica infection of the gut. Infect Immun 78:3716–3725. doi: 10.1128/IAI.00203-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Roselli M, Finamore A, Nuccitelli S, Carnevali P, Brigidi P, Vitali B, Nobili F, Rami R, Garaguso I, Mengheri E. 2009. Prevention of TNBS-induced colitis by different Lactobacillus and Bifidobacterium strains is associated with an expansion of gammadeltaT and regulatory T cells of intestinal intraepithelial lymphocytes. Inflamm Bowel Dis 15:1526–1536. doi: 10.1002/ibd.20961. [DOI] [PubMed] [Google Scholar]
- 54.Haas T, Metzger J, Schmitz F, Heit A, Muller T, Latz E, Wagner H. 2008. The DNA sugar backbone 2' deoxyribose determines toll-like receptor 9 activation. Immunity 28:315–323. doi: 10.1016/j.immuni.2008.01.013. [DOI] [PubMed] [Google Scholar]
- 55.He F, Peng J, Deng XL, Yang LF, Camara AD, Omran A, Wang GL, Wu LW, Zhang CL, Yin F. 2012. Mechanisms of tumor necrosis factor-alpha-induced leaks in intestine epithelial barrier. Cytokine 59:264–272. doi: 10.1016/j.cyto.2012.04.008. [DOI] [PubMed] [Google Scholar]
- 56.Wang F, Schwarz BT, Graham WV, Wang Y, Su L, Clayburgh DR, Abraham C, Turner JR. 2006. IFN-gamma-induced TNFR2 expression is required for TNF-dependent intestinal epithelial barrier dysfunction. Gastroenterology 131:1153–1163. doi: 10.1053/j.gastro.2006.08.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Ramsay AJ, Husband AJ, Ramshaw IA, Bao S, Matthaei KI, Koehler G, Kopf M. 1994. The role of interleukin-6 in mucosal IgA antibody responses in vivo. Science 264:561–563. doi: 10.1126/science.8160012. [DOI] [PubMed] [Google Scholar]
- 58.Kandasamy S, Chattha KS, Vlasova AN, Rajashekara G, Saif LJ. 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. doi: 10.4161/19490976.2014.969972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Vlasova AN, Chattha KS, Kandasamy S, Siegismund CS, Saif LJ. 2013. Prenatally acquired vitamin A deficiency alters innate immune responses to human rotavirus in a gnotobiotic pig model. J Immunol 190:4742–4753. doi: 10.4049/jimmunol.1203575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Jin CJ, Hong CY, Takei M, Chung SY, Park JS, Pham TN, Choi SJ, Nam JH, Chung IJ, Kim HJ, Lee JJ. 2010. All-trans retinoic acid inhibits the differentiation, maturation, and function of human monocyte-derived dendritic cells. Leuk Res 34:513–520. doi: 10.1016/j.leukres.2009.10.006. [DOI] [PubMed] [Google Scholar]
- 61.Krieg AM, Yi AK, Matson S, Waldschmidt TJ, Bishop GA, Teasdale R, Koretzky GA, Klinman DM. 1995. CpG motifs in bacterial DNA trigger direct B-cell activation. Nature 374:546–549. doi: 10.1038/374546a0. [DOI] [PubMed] [Google Scholar]
- 62.Belkaid Y, Oldenhove G. 2008. Tuning microenvironments: induction of regulatory T cells by dendritic cells. Immunity 29:362–371. doi: 10.1016/j.immuni.2008.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Tezuka H, Abe Y, Asano J, Sato T, Liu J, Iwata M, Ohteki T. 2011. Prominent role for plasmacytoid dendritic cells in mucosal T cell-independent IgA induction. Immunity 34:247–257. doi: 10.1016/j.immuni.2011.02.002. [DOI] [PubMed] [Google Scholar]
- 64.Deal EM, Lahl K, Narvaez CF, Butcher EC, Greenberg HB. 2013. Plasmacytoid dendritic cells promote rotavirus-induced human and murine B cell responses. J Clin Invest 123:2464–2474. doi: 10.1172/JCI60945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Lecker SH, Solomon V, Mitch WE, Goldberg AL. 1999. Muscle protein breakdown and the critical role of the ubiquitin-proteasome pathway in normal and disease states. J Nutr 129:227S–237S. doi: 10.1093/jn/129.1.227S. [DOI] [PubMed] [Google Scholar]
- 66.Mao X, Gu C, Ren M, Chen D, Yu B, He J, Yu J, Zheng P, Luo J, Luo Y, Wang J, Tian G, Yang Q. 2018. l-Isoleucine administration alleviates rotavirus infection and immune response in the weaned piglet model. Front Immunol 9:1654. doi: 10.3389/fimmu.2018.01654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Wannemacher RW, Jr, Klainer AS, Dinterman RE, Beisel WR. 1976. The significance and mechanism of an increased serum phenylalanine-tyrosine ratio during infection. Am J Clin Nutr 29:997–1006. doi: 10.1093/ajcn/29.9.997. [DOI] [PubMed] [Google Scholar]
- 68.Mayneris-Perxachs J, Swann JR. 2019. Metabolic phenotyping of malnutrition during the first 1000 days of life. Eur J Nutr 58:909–930. doi: 10.1007/s00394-018-1679-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Mao X, Yang Q, Chen D, Yu B, He J. 2019. Benzoic acid used as food and feed additives can regulate gut functions. Biomed Res Int 2019:5721585. doi: 10.1155/2019/5721585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Hinz B, Kraus V, Pahl A, Brune K. 2000. Salicylate metabolites inhibit cyclooxygenase-2-dependent prostaglandin E(2) synthesis in murine macrophages. Biochem Biophys Res Commun 274:197–202. doi: 10.1006/bbrc.2000.3123. [DOI] [PubMed] [Google Scholar]
- 71.Bauchart-Thevret C, Stoll B, Burrin DG. 2009. Intestinal metabolism of sulfur amino acids. Nutr Res Rev 22:175–187. doi: 10.1017/S0954422409990138. [DOI] [PubMed] [Google Scholar]
- 72.Zhao Y, Qian L. 2014. Homocysteine-mediated intestinal epithelial barrier dysfunction in the rat model of irritable bowel syndrome caused by maternal separation. Acta Biochim Biophys Sin (Shanghai) 46:917–919. doi: 10.1093/abbs/gmu076. [DOI] [PubMed] [Google Scholar]
- 73.Bauchart-Thevret C, Stoll B, Chacko S, Burrin DG. 2009. Sulfur amino acid deficiency upregulates intestinal methionine cycle activity and suppresses epithelial growth in neonatal pigs. Am J Physiol Endocrinol Metab 296:E1239–50. doi: 10.1152/ajpendo.91021.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Ruth MR, Field CJ. 2013. The immune modifying effects of amino acids on gut-associated lymphoid tissue. J Anim Sci Biotechnol 4:27. doi: 10.1186/2049-1891-4-27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Yang X, Twitchell E, Li G, Wen K, Weiss M, Kocher J, Lei S, Ramesh A, Ryan EP, Yuan L. 2015. High protective efficacy of rice bran against human rotavirus diarrhea via enhancing probiotic growth, gut barrier function, and innate immunity. Sci Rep 5:15004. doi: 10.1038/srep15004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.de Jong PR, Gonzalez-Navajas JM, Jansen NJ. 2016. The digestive tract as the origin of systemic inflammation. Crit Care 20:279. doi: 10.1186/s13054-016-1458-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Lei S, Ramesh A, Twitchell E, Wen K, Bui T, Weiss M, Yang X, Kocher J, Li G, Giri-Rachman E, Trang NV, Jiang X, Ryan EP, Yuan L. 2016. High protective efficacy of probiotics and rice bran against human norovirus infection and diarrhea in gnotobiotic pigs. Front Microbiol 7:1699. doi: 10.3389/fmicb.2016.01699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Wegiel B, Otterbein LE. 2012. Go green: the anti-inflammatory effects of biliverdin reductase. Front Pharmacol 3:47. doi: 10.3389/fphar.2012.00047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Smith SG, El-Far MA. 1980. The effect of fasting and protein calorie malnutrition on the liver porphyrins. Int J Biochem 12:979–980. doi: 10.1016/0020-711X(80)90196-2. [DOI] [PubMed] [Google Scholar]
- 80.Lykke M, Hother AL, Hansen CF, Friis H, Molgaard C, Michaelsen KF, Briend A, Larsen T, Sangild PT, Thymann T. 2013. Malnutrition induces gut atrophy and increases hepatic fat infiltration: studies in a pig model of childhood malnutrition. Am J Transl Res 5:543–554. [PMC free article] [PubMed] [Google Scholar]
- 81.Norris GH, Blesso CN. 2017. Dietary and endogenous sphingolipid metabolism in chronic inflammation. Nutrients 9:1180. doi: 10.3390/nu9111180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Pavicic T, Wollenweber U, Farwick M, Korting HC. 2007. Anti-microbial and -inflammatory activity and efficacy of phytosphingosine: an in vitro and in vivo study addressing acne vulgaris. Int J Cosmet Sci 29:181–190. doi: 10.1111/j.1467-2494.2007.00378.x. [DOI] [PubMed] [Google Scholar]
- 83.Brundige DR, Maga EA, Klasing KC, Murray JD. 2010. Consumption of pasteurized human lysozyme transgenic goats' milk alters serum metabolite profile in young pigs. Transgenic Res 19:563–574. doi: 10.1007/s11248-009-9334-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Khanapure SP, Garvey DS, Janero DR, Letts LG. 2007. Eicosanoids in inflammation: biosynthesis, pharmacology, and therapeutic frontiers. Curr Top Med Chem 7:311–340. doi: 10.2174/156802607779941314. [DOI] [PubMed] [Google Scholar]
- 85.Pompeia C, Lima T, Curi R. 2003. Arachidonic acid cytotoxicity: can arachidonic acid be a physiological mediator of cell death? Cell Biochem Funct 21:97–104. doi: 10.1002/cbf.1012. [DOI] [PubMed] [Google Scholar]
- 86.Zarny LA, Bernstein LH. 1995. Serum cholesterol: an indicator of malnutrition. J Am Diet Assoc 95:A25. doi: 10.1016/S0002-8223(95)00436-X. [DOI] [Google Scholar]
- 87.Koeth RA, Levison BS, Culley MK, Buffa JA, Wang Z, Gregory JC, Org E, Wu Y, Li L, Smith JD, Tang WHW, DiDonato JA, Lusis AJ, Hazen SL. 2014. gamma-Butyrobetaine is a proatherogenic intermediate in gut microbial metabolism of L-carnitine to TMAO. Cell Metab 20:799–812. doi: 10.1016/j.cmet.2014.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Flanagan JL, Simmons PA, Vehige J, Willcox MD, Garrett Q. 2010. Role of carnitine in disease. Nutr Metab (Lond) 7:30. doi: 10.1186/1743-7075-7-30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Beardsley GP, Moroson BA, Taylor EC, Moran RG. 1989. A new folate antimetabolite, 5,10-dideaza-5,6,7,8-tetrahydrofolate is a potent inhibitor of de novo purine synthesis. J Biol Chem 264:328–333. doi: 10.1016/S0021-9258(17)31261-9. [DOI] [PubMed] [Google Scholar]
- 90.Greene S, Watanabe K, Braatz-Trulson J, Lou L. 1995. Inhibition of dihydroorotate dehydrogenase by the immunosuppressive agent leflunomide. Biochem Pharmacol 50:861–867. doi: 10.1016/0006-2952(95)00255-X. [DOI] [PubMed] [Google Scholar]
- 91.Hoppe-Seyler K, Sauer P, Lohrey C, Hoppe-Seyler F. 2012. The inhibitors of nucleotide biosynthesis leflunomide, FK778, and mycophenolic acid activate hepatitis B virus replication in vitro. Hepatology 56:9–16. doi: 10.1002/hep.25602. [DOI] [PubMed] [Google Scholar]
- 92.Nelson JA, Carpenter JW, Rose LM, Adamson DJ. 1975. Mechanisms of action of 6-thioguanine, 6-mercaptopurine, and 8-azaguanine. Cancer Res 35:2872–2878. [PubMed] [Google Scholar]
- 93.Qing M, Zou G, Wang QY, Xu HY, Dong H, Yuan Z, Shi PY. 2010. Characterization of dengue virus resistance to brequinar in cell culture. Antimicrob Agents Chemother 54:3686–3695. doi: 10.1128/AAC.00561-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Silva HT, Jr, Cao W, Shorthouse RA, Loffler M, Morris RE. 1997. In vitro and in vivo effects of leflunomide, brequinar, and cyclosporine on pyrimidine biosynthesis. Transplant Proc 29:1292–1293. doi: 10.1016/S0041-1345(96)00523-4. [DOI] [PubMed] [Google Scholar]
- 95.Wang QY, Bushell S, Qing M, Xu HY, Bonavia A, Nunes S, Zhou J, Poh MK, Florez de Sessions P, Niyomrattanakit P, Dong H, Hoffmaster K, Goh A, Nilar S, Schul W, Jones S, Kramer L, Compton T, Shi PY. 2011. Inhibition of dengue virus through suppression of host pyrimidine biosynthesis. J Virol 85:6548–6556. doi: 10.1128/JVI.02510-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Wang Y, Wang W, Xu L, Zhou X, Shokrollahi E, Felczak K, van der Laan LJ, Pankiewicz KW, Sprengers D, Raat NJ, Metselaar HJ, Peppelenbosch MP, Pan Q. 2016. Cross talk between nucleotide synthesis pathways with cellular immunity in constraining hepatitis e virus replication. Antimicrob Agents Chemother 60:2834–2848. doi: 10.1128/AAC.02700-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Mogensen KM, Lasky-Su J, Rogers AJ, Baron RM, Fredenburgh LE, Rawn J, Robinson MK, Massarro A, Choi AM, Christopher KB. 2017. Metabolites associated with malnutrition in the intensive care unit are also associated with 28-day mortality. JPEN J Parenter Enteral Nutr 41:188–197. doi: 10.1177/0148607116656164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Braidy N, Berg J, Clement J, Khorshidi F, Poljak A, Jayasena T, Grant R, Sachdev P. 2019. Role of nicotinamide adenine dinucleotide and related precursors as therapeutic targets for age-related degenerative diseases: rationale, biochemistry, pharmacokinetics, and outcomes. Antioxid Redox Signal 30:251–294. doi: 10.1089/ars.2017.7269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Braidy N, Guillemin G, Grant R. 2008. Promotion of cellular NAD(+) anabolism: therapeutic potential for oxidative stress in ageing and Alzheimer's disease. Neurotox Res 13:173–184. doi: 10.1007/BF03033501. [DOI] [PubMed] [Google Scholar]
- 100.Grant RS, Passey R, Matanovic G, Smythe G, Kapoor V. 1999. Evidence for increased de novo synthesis of NAD in immune-activated RAW264.7 macrophages: a self-protective mechanism? Arch Biochem Biophys 372:1–7. doi: 10.1006/abbi.1999.1381. [DOI] [PubMed] [Google Scholar]
- 101.Meyer RC, Bohl EH, Kohler EM. 1964. Procurement and maintenance of germ-free seine for microbiological investigations. Appl Microbiol 12:295–300. doi: 10.1128/am.12.4.295-300.1964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Chattha KS, Vlasova AN, Kandasamy S, Rajashekara G, Saif LJ. 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. doi: 10.4049/jimmunol.1300678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Zhang W, Azevedo MS, Wen K, Gonzalez A, Saif LJ, Li G, Yousef AE, Yuan L. 2008. Probiotic Lactobacillus acidophilus enhances the immunogenicity of an oral rotavirus vaccine in gnotobiotic pigs. Vaccine 26:3655–3661. doi: 10.1016/j.vaccine.2008.04.070. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Ward LA, Yuan L, Rosen BI, To TL, Saif LJ. 1996. Development of mucosal and systemic lymphoproliferative responses and protective immunity to human group A rotaviruses in a gnotobiotic pig model. Clin Diagn Lab Immunol 3:342–350. doi: 10.1128/cdli.3.3.342-350.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Miyazaki A, Kandasamy S, Michael H, Langel SN, Paim FC, Chepngeno J, Alhamo MA, Fischer DD, Huang HC, Srivastava V, Kathayat D, Deblais L, Rajashekara G, Saif LJ, Vlasova AN. 2018. Protein deficiency reduces efficacy of oral attenuated human rotavirus vaccine in a human infant fecal microbiota transplanted gnotobiotic pig model. Vaccine 36:6270–6281. doi: 10.1016/j.vaccine.2018.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Kandasamy S, Chattha KS, Vlasova AN, Saif LJ. 2014. Prenatal vitamin A deficiency impairs adaptive immune responses to pentavalent rotavirus vaccine (RotaTeq(R)) in a neonatal gnotobiotic pig model. Vaccine 32:816–824. doi: 10.1016/j.vaccine.2013.12.039. [DOI] [PubMed] [Google Scholar]
- 107.Chattha KS, Kandasamy S, Vlasova AN, Saif LJ. 2013. Vitamin A deficiency impairs adaptive B and T cell responses to a prototype monovalent attenuated human rotavirus vaccine and virulent human rotavirus challenge in a gnotobiotic piglet model. PLoS One 8:e82966. doi: 10.1371/journal.pone.0082966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Chattha KS, Vlasova AN, Kandasamy S, Esseili MA, Siegismund C, Rajashekara G, Saif LJ. 2013. Probiotics and colostrum/milk differentially affect neonatal humoral immune responses to oral rotavirus vaccine. Vaccine 31:1916–1923. doi: 10.1016/j.vaccine.2013.02.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Annamalai T, Saif LJ, Lu Z, Jung K. 2015. Age-dependent variation in innate immune responses to porcine epidemic diarrhea virus infection in suckling versus weaned pigs. Vet Immunol Immunopathol 168:193–202. doi: 10.1016/j.vetimm.2015.09.006. [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.
Supplementary Materials
Excel file, relative intensities of dysregulated metabolites. Download Table S1, XLSX file, 0.02 MB (21KB, xlsx) .
Copyright © 2022 Michael et al.
This content is distributed under the terms of the Creative Commons Attribution 4.0 International license.
Excel file, correlations of immune responses with significantly altered metabolites in plasma and fecal metabolomic and lipidomic analysis. Download Table S2, XLSX file, 0.01 MB (14.3KB, xlsx) .
Copyright © 2022 Michael et al.
This content is distributed under the terms of the Creative Commons Attribution 4.0 International license.






