14 research outputs found

    Image_3_Lactobacillus paracasei CNCM I-5220-derived postbiotic protects from the leaky-gut.TIF

    No full text
    The maintenance of intestinal barrier function is essential for preventing different pathologies, such as the leaky gut syndrome (LGS), which is characterized by the passage of harmful agents, like bacteria, toxins, and viruses, into the bloodstream. Intestinal barrier integrity is controlled by several players, including the gut microbiota. Various molecules, called postbiotics, are released during the natural metabolic activity of the microbiota. Postbiotics can regulate host–microbe interactions, epithelial homeostasis, and have overall benefits for our health. In this work, we used in vitro and in vivo systems to demonstrate the role of Lactobacillus paracasei CNCM I-5220-derived postbiotic (LP-PBF) in preserving intestinal barrier integrity. We demonstrated in vitro that LP-PBF restored the morphology of tight junctions (TJs) that were altered upon Salmonella typhimurium exposure. In vivo, LP-PBF protected the gut vascular barrier and blocked S. typhimurium dissemination into the bloodstream. Interestingly, we found that LP-PBF interacts not only with the host cells, but also directly with S. typhimurium blocking its biofilm formation, partially due to the presence of biosurfactants. This study highlights that LP-PBF is beneficial in maintaining gut homeostasis due to the synergistic effect of its different components. These results suggest that LP-PBF could be utilized in managing several pathologies displaying an impaired intestinal barrier function.</p

    Image_1_Lactobacillus paracasei CNCM I-5220-derived postbiotic protects from the leaky-gut.TIF

    No full text
    The maintenance of intestinal barrier function is essential for preventing different pathologies, such as the leaky gut syndrome (LGS), which is characterized by the passage of harmful agents, like bacteria, toxins, and viruses, into the bloodstream. Intestinal barrier integrity is controlled by several players, including the gut microbiota. Various molecules, called postbiotics, are released during the natural metabolic activity of the microbiota. Postbiotics can regulate host–microbe interactions, epithelial homeostasis, and have overall benefits for our health. In this work, we used in vitro and in vivo systems to demonstrate the role of Lactobacillus paracasei CNCM I-5220-derived postbiotic (LP-PBF) in preserving intestinal barrier integrity. We demonstrated in vitro that LP-PBF restored the morphology of tight junctions (TJs) that were altered upon Salmonella typhimurium exposure. In vivo, LP-PBF protected the gut vascular barrier and blocked S. typhimurium dissemination into the bloodstream. Interestingly, we found that LP-PBF interacts not only with the host cells, but also directly with S. typhimurium blocking its biofilm formation, partially due to the presence of biosurfactants. This study highlights that LP-PBF is beneficial in maintaining gut homeostasis due to the synergistic effect of its different components. These results suggest that LP-PBF could be utilized in managing several pathologies displaying an impaired intestinal barrier function.</p

    Image_4_Lactobacillus paracasei CNCM I-5220-derived postbiotic protects from the leaky-gut.TIF

    No full text
    The maintenance of intestinal barrier function is essential for preventing different pathologies, such as the leaky gut syndrome (LGS), which is characterized by the passage of harmful agents, like bacteria, toxins, and viruses, into the bloodstream. Intestinal barrier integrity is controlled by several players, including the gut microbiota. Various molecules, called postbiotics, are released during the natural metabolic activity of the microbiota. Postbiotics can regulate host–microbe interactions, epithelial homeostasis, and have overall benefits for our health. In this work, we used in vitro and in vivo systems to demonstrate the role of Lactobacillus paracasei CNCM I-5220-derived postbiotic (LP-PBF) in preserving intestinal barrier integrity. We demonstrated in vitro that LP-PBF restored the morphology of tight junctions (TJs) that were altered upon Salmonella typhimurium exposure. In vivo, LP-PBF protected the gut vascular barrier and blocked S. typhimurium dissemination into the bloodstream. Interestingly, we found that LP-PBF interacts not only with the host cells, but also directly with S. typhimurium blocking its biofilm formation, partially due to the presence of biosurfactants. This study highlights that LP-PBF is beneficial in maintaining gut homeostasis due to the synergistic effect of its different components. These results suggest that LP-PBF could be utilized in managing several pathologies displaying an impaired intestinal barrier function.</p

    Image_2_Lactobacillus paracasei CNCM I-5220-derived postbiotic protects from the leaky-gut.TIF

    No full text
    The maintenance of intestinal barrier function is essential for preventing different pathologies, such as the leaky gut syndrome (LGS), which is characterized by the passage of harmful agents, like bacteria, toxins, and viruses, into the bloodstream. Intestinal barrier integrity is controlled by several players, including the gut microbiota. Various molecules, called postbiotics, are released during the natural metabolic activity of the microbiota. Postbiotics can regulate host–microbe interactions, epithelial homeostasis, and have overall benefits for our health. In this work, we used in vitro and in vivo systems to demonstrate the role of Lactobacillus paracasei CNCM I-5220-derived postbiotic (LP-PBF) in preserving intestinal barrier integrity. We demonstrated in vitro that LP-PBF restored the morphology of tight junctions (TJs) that were altered upon Salmonella typhimurium exposure. In vivo, LP-PBF protected the gut vascular barrier and blocked S. typhimurium dissemination into the bloodstream. Interestingly, we found that LP-PBF interacts not only with the host cells, but also directly with S. typhimurium blocking its biofilm formation, partially due to the presence of biosurfactants. This study highlights that LP-PBF is beneficial in maintaining gut homeostasis due to the synergistic effect of its different components. These results suggest that LP-PBF could be utilized in managing several pathologies displaying an impaired intestinal barrier function.</p

    Effects of goat whey on cell lines.

    No full text
    <p>(A) Nitrite (NO) production and (B) interleukin (IL)-6 levels in Raw 264 and CMT-93 cells, respectively, in basal or LPS-stimulated conditions (100 ng/mL and 1 μg/mL, respectively). Data are expressed as the mean ± SEM. The bars with different letters are significantly different (one-way ANOVA post hoc Tukey’s test, P < 0.05).</p

    Effects of goat whey on gene expression by RT-qPCR and immunofluorescence of the intestinal mucosal barrier proteins as measured.

    No full text
    <p>Colonic gene expression of the barrier function mediators gene expression (A) Mucin (MUC)-2, (B) MUC-3, (C) occludin, (D) zonula occludens (ZO)-1 analyzed by real-time qPCR and normalized with the housekeeping gene, Glyceraldehyde-3-phosphate dehydrohenase (GAPDH) in dinitrobenzene-sulphonic acid (DNBS) mice colitis 4 days after damage induction. Representative confocal photomicrographs of ZO-1 (E) immunoreactivity (green) in colons of the animals from each group; the sections are nuclear counterstained with DAPI (blue): (E.1) Healthy group had moderated ZO-1 labelling; (E.2) ZO-1 labelling was almost absence in DNBS control group; (E.3) ZO-1 labelling (red arrow) was strong in the treated group with goat whey; (E.4) Densitometric analysis confirmed a significant increases in ZO-1 in goat whey. Data are expressed as the means ± SEM. the groups with different letters differ significantly (one-way ANOVA post hoc Tukey’s test, P < 0.05).</p

    Effects of goat whey on the colonic mucosa of colitic mice as assessed by histological examination.

    No full text
    <p>Sections of the colonic mucosa were stained with haematoxylin and eosin (x100): (A) Healthy, (B) DNBS control, and (C) Goat Whey. (D) Microscopic scores were assigned to the different groups according to the criteria described by Zea-Iriarte et al. (1996) [<a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0185382#pone.0185382.ref026" target="_blank">26</a>] and (E) Myeloperoxidase activity–MPO. Data are expressed as the means ± SEM (n = 12/group), and the groups with different letters differ significantly (one-way ANOVA post hoc Tukey’s test, P < 0.05).</p

    Effects of goat whey on pro-inflammatory cytokines as measured by ELISA.

    No full text
    <p>Distal colon tissue samples were cultured overnight. The supernatants were assessed for cytokine levels using kits from R&D Systems (Minneapolis, MN, USA) following the manufacturer’s protocols. The cytokine levels in the supernatant were expressed as the concentration in pg/mL. (A) Interleukin (IL)-6 and (B) tumour necrosis factor (TNF)-α production in colonic tissues from mice with 2,4-dinitrobenzenesulfonic acid (DNBS)-induced colitis. Data are expressed as the mean ± SEM (n = 12). The groups with different letters are significantly different (one-way ANOVA post hoc Tukey’s test, P < 0.05).</p

    Effect of goat whey on IL-17 expression in colitic mice.

    No full text
    <p>Representative confocal photomicrographs of IL-17 (Panel A) immunoreactivity (green) in colons of the animals from each group; the sections are nuclear counterstained with DAPI (blue): (A.1) Healthy group had absent or weak IL-17 labelling in all mucosa layers; (A.2) IL-17 labelling was strong in the DNBS control group; (A.3) weak to moderate IL-17 labelling (red arrow) was seen in the group treated with goat whey; (A.4) Densitometric analysis confirmed a significant reduction in IL-17 immunoreactivity in goat whey. Data are expressed as the means ± SEM; the groups with different letters differ significantly (one-way ANOVA post hoc Tukey’s test, P < 0.05).</p

    Effects of goat whey on the gene expression of pro-inflammatory cytokines as measured by RT-qPCR.

    No full text
    <p>Colonic gene expression of the pro-inflammatory cytokines (A) Interleukin (IL)-1β, (B) IL-6, (C) tumour necrosis factor (TNF)-α, (D) inducible nitric oxide synthase (iNOS), (E) matrix metalloproteinase (MMP)-9, and (F) intercellular adhesion molecule (ICAM)-1 analyzed by real-time qPCR and normalized with the housekeeping gene, Glyceraldehyde-3-phosphate dehydrohenase (GAPDH) in dinitrobenzene-sulphonic acid (DNBS) mice colitis 4 days after damage induction. Data are expressed as the mean ± SEM (n = 12/group). The groups with different letters are significantly different (one-way ANOVA post hoc Tukey’s test, P < 0.05).</p
    corecore