27 research outputs found

    Fungi: Friend or Foe? A Mycobiome Evaluation in Children with Autism and Gastrointestinal Symptoms

    Get PDF
    Gastrointestinal (GI) symptoms often affect children with autism spectrum disorders (ASD) and GI symptoms have been associated with an abnormal fecal microbiome. There is limited evidence of Candida species being more prevalent in children with ASD. We enrolled 20 children with ASD and GI symptoms (ASD + GI), 10 children with ASD but no GI symptoms (ASD - GI), and 20 from typically developing (TD) children in this pilot study. Fecal mycobiome taxa were analyzed by Internal Transcribed Spacer sequencing. GI symptoms (GI Severity Index [GSI]), behavioral symptoms (Social Responsiveness Scale -2 [SRS-2]), inflammation and fungal immunity (fecal calprotectin and serum dectin-1 [ELISA]) were evaluated. We observed no changes in the abundance of total fungal species (alpha diversity) between groups. Samples with identifiable Candida spp. were present in 4 of 19 (21%) ASD + GI, in 5 of 9 (56%) ASD - GI, and in 4 of 16 (25%) TD children (overall P = 0.18). The presence of Candida spp. did not correlate with behavioral or GI symptoms (P = 0.38, P = 0.5, respectively). Fecal calprotectin was normal in all but one child. Finally, there was no significance in serum dectin-1 levels, suggesting no increased fungal immunity in children with ASD. Our data suggest that fungi are present at normal levels in the stool of children with ASD and are not associated with gut inflammation

    Lactobacillus reuteri

    No full text

    Lactobacillus reuteri

    No full text

    Human-derived probiotic Lactobacillus reuteri strains differentially reduce intestinal inflammation

    No full text
    Lactobacillus reuteri (L. reuteri) is a probiotic that inhibits the severity of enteric infections and modulates the immune system. Human-derived L. reuteri strains DSM17938, ATCC PTA4659, ATCC PTA 5289, and ATCC PTA 6475 have demonstrated strain-specific immunomodulation in cultured monocytoid cells, but information about how these strains affect inflammation in intestinal epithelium is limited. We determined the effects of the four different L. reuteri strains on lipopolysaccharide (LPS)-induced inflammation in small intestinal epithelial cells and in the ileum of newborn rats. IPEC-J2 cells (derived from the jejunal epithelium of a neonatal piglet) and IEC-6 cells (derived from the rat crypt) were treated with L. reuteri. Newborn rat pups were gavaged cow milk formula supplemented with L. reuteri strains in the presence or absence of LPS. Protein and mRNA levels of cytokines and histological changes were measured. We demonstrate that even though one L. reuteri strain (DSM 17938) did not inhibit LPS-induced IL-8 production in cultured intestinal cells, all strains significantly reduced intestinal mucosal levels of KC/GRO (∼IL-8) and IFN-γ when newborn rat pups were fed formula containing LPS ± L. reuteri. Intestinal histological damage produced by LPS plus cow milk formula was also significantly reduced by all four strains. Cow milk formula feeding (without LPS) produced mild gut inflammation, evidenced by elevated mucosal IFN-γ and IL-13 levels, a process that could be suppressed by strain 17938. Other cytokines and chemokines were variably affected by the different strains, and there was no toxic effect of L. reuteri on intestinal cells or mucosa. In conclusion, L. reuteri strains differentially modulate LPS-induced inflammation. Probiotic interactions with both epithelial and nonepithelial cells in vivo must be instrumental in modulating intrinsic anti-inflammatory effects in the intestine. We suggest that the terms anti- and proinflammatory be used only to describe the effects of a probiotic in the living host

    <em>Lactobacillus reuteri</em> DSM 17938 Changes the Frequency of Foxp3<sup>+</sup> Regulatory T Cells in the Intestine and Mesenteric Lymph Node in Experimental Necrotizing Enterocolitis

    Get PDF
    <div><p>Necrotizing enterocolitis (NEC) is an inflammatory disease of the intestine in premature infants. <i>Lactobacillus reuteri</i> DSM 17938 improves survival and reduces the incidence and severity of NEC in a rodent model. Foxp3<sup>+</sup> regulatory T cells (Tregs) maintain intestinal homeostasis by controlling inflammation and inducing tolerance. To determine whether there are insufficient numbers of Tregs to control inflammation in NEC and to determine if LR17938 increases the frequency of Tregs, we studied selected groups of newborn Sprague-Dawley rats according to feeding plan: dam±LR17938, formula±LR17938, and NEC±LR17938. NEC was induced by gavage feeding with special formula and exposure to hypoxic conditions. Lymphocytes isolated from ileum, mesenteric lymph nodes (MLN), spleen and thymus were labeled for T cell surface markers (CD3, CD4, CD8) and intracellular Foxp3; and labeled cells were analyzed by flow cytometry. The percentage of CD3<sup>+</sup> T cells and Foxp3<sup>+</sup> Tregs in the ileum significantly decreased in pups with NEC, compared to normal controls. Feeding LR17938 to neonatal rats with NEC increased the % of Foxp3<sup>+</sup> T cells in the ileum while decreasing the percentage of cells in the MLN. Administration of LR17938 to dam-fed rats significantly increased Foxp3<sup>+</sup>Tregs in the ileum as early as day of life (DOL)1 but did not produce an increase in Tregs in formula-fed rats on DOL1. These results suggest that factors in breast milk may enhance the early immunomodulatory effects of LR17938. An anti-inflammatory effect of LR17938 in NEC was associated with the modulation of immune responses and induction and what appears to be migration of Foxp3<sup>+</sup> Tregs to the diseased gut. Probiotic-facilitated development of Tregs might play an important role in the prevention of NEC.</p> </div

    Changes in intestinal Toll-like receptors and cytokines precede histological injury in a rat model of necrotizing enterocolitis

    No full text
    It is unclear whether the broad inflammatory response shown in neonatal necrotizing enterocolitis (NEC) is the cause or the effect of tissue injury. Toll-like receptors (TLRs) on intestinal dendritic, mononuclear, and epithelial cells recognize bacterial ligands and damaged tissues, thus activating the inflammatory response. The present study aimed to determine whether active TLR signaling would precede histological injury in NEC. Newborn rat pups were divided into four groups: dam fed, dam fed-hypoxic, formula fed, and formula fed-hypoxic (NEC). The ileal tissues were evaluated for NEC scores at 24, 48, 72, and 120 h. Quantitative real-time reverse transcription-polymerase chain reaction and immunohistochemistry were used to measure and localize intestinal TLRs. Cytokines were assessed by a multispot cytokine array. Among the four groups, ileal injury was seen only after 72 h of formula feeding and hypoxia. We found selective induction of mRNA levels in NEC compared with dam-fed controls for TLR2 > TLR4 > TLR1 = TLR3, TLR7, and TLR9 > TLR6 (P < 0.01); TLR5 was downregulated (P < 0.01). All TLR changes started at 48 h, before any histological evidence of NEC. Both Th1-type cytokines (IFN-γ, IL-1β, TNF-α, and KC/GRO) and Th2-type cytokines (IL-4, IL-5 and IL-13) were significantly increased in NEC but also in nondamaged formula-fed rat ileum. In conclusion, the intestinal expression of TLRs and cytokines precedes histological injury in the experimental NEC

    Effect of breast milk or formula feeding with or without <i>Lactobacillus reuteri</i> DSM 17938 on the frequency of Foxp3<sup>+</sup> T cells in the ileum and mesenteric lymph node (MLN) of rats during DOL 0 and DOL 4.

    No full text
    <p>Newborn rats were fed immediately after birth for 4 days. (a) and (c): Changes of single CD4<sup>+</sup>Foxp3<sup>+</sup> Tregs (%) in the ileum (a) and MLN (c); (b) and (d): changes of double CD4<sup>+</sup>CD8<sup>+</sup>Foxp3<sup>+</sup> T cells in the ileum (b) and MLN (d). The percentage (%) of cells is represented as the mean ± SE of N = 9 observations (pups) in each DOL of each feeding group (total N = 45 for each feeding group).* Dam-fed +17938 vs. Dam-fed, **p<0.01, ***p<0.001. # Formula vs. Dam-fed, # p<0.05, ## p<0.01. &: Formula-fed +17938 vs. Formula-fed, & p<0.05, && p<0.01.</p

    Effect of <i>Lactobacillus reuteri</i> DSM 17938 on survival, CD3<sup>+</sup> T cells and Foxp3<sup>+</sup> T cells in the ileum of rats with NEC.

    No full text
    <p>(a) Survival (%) of different groups of rat pups (dam-fed, NEC, and NEC+17938). (b) Changes of % CD3<sup>+</sup> T cells of lymphocytes in different groups. (c) Immunohistochemical staining of CD3<sup>+</sup> cells in rat pup ileum, 200 × magnification. (d) Number of CD3<sup>+</sup> T cells per 5 villi in IHC. (e) Percentage of CD4<sup>+</sup>Foxp3<sup>+</sup>Tregs in each group on day 4 of NEC. (f) Percentage of double positive CD4<sup>+</sup>CD8<sup>+</sup>Foxp3<sup>+</sup> T cells. The numbers represent percent of T cells (means ± SE). Each dot represents one animal in each group in the figures b, e, and f. Dam-fed: N = 15, NEC: N = 13, NEC+17938: N = 20.</p
    corecore