2
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Bofutsushosan improves gut barrier function with a bloom of Akkermansia muciniphila and improves glucose metabolism in mice with diet-induced obesity

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Obesity and insulin resistance are associated with dysbiosis of the gut microbiota and impaired intestinal barrier function. Herein, we report that Bofutsushosan (BFT), a Japanese herbal medicine, Kampo, which has been clinically used for constipation in Asian countries, ameliorates glucose metabolism in mice with diet–induced obesity. A 16S rRNA sequence analysis of fecal samples showed that BFT dramatically increased the relative abundance of Verrucomicrobia, which was mainly associated with a bloom of Akkermansia muciniphila (AKK). BFT decreased the gut permeability as assessed by FITC-dextran gavage assay, associated with increased expression of tight-junction related protein, claudin-1, in the colon. The BFT treatment group also showed significant decreases of the plasma endotoxin level and expression of the hepatic lipopolysaccharide-binding protein. Antibiotic treatment abrogated the metabolic effects of BFT. Moreover, many of these changes could be reproduced when the cecal contents of BFT-treated donors were transferred to antibiotic-pretreated high fat diet-fed mice. These data demonstrate that BFT modifies the gut microbiota with an increase in AKK, which may contribute to improving gut barrier function and preventing metabolic endotoxemia, leading to attenuation of diet-induced inflammation and glucose intolerance. Understanding the interaction between a medicine and the gut microbiota may provide insights into new pharmacological targets to improve glucose metabolism.

          Related collections

          Most cited references20

          • Record: found
          • Abstract: found
          • Article: not found

          Short-chain fatty acids and ketones directly regulate sympathetic nervous system via G protein-coupled receptor 41 (GPR41).

          The maintenance of energy homeostasis is essential for life, and its dysregulation leads to a variety of metabolic disorders. Under a fed condition, mammals use glucose as the main metabolic fuel, and short-chain fatty acids (SCFAs) produced by the colonic bacterial fermentation of dietary fiber also contribute a significant proportion of daily energy requirement. Under ketogenic conditions such as starvation and diabetes, ketone bodies produced in the liver from fatty acids are used as the main energy sources. To balance energy intake, dietary excess and starvation trigger an increase or a decrease in energy expenditure, respectively, by regulating the activity of the sympathetic nervous system (SNS). The regulation of metabolic homeostasis by glucose is well recognized; however, the roles of SCFAs and ketone bodies in maintaining energy balance remain unclear. Here, we show that SCFAs and ketone bodies directly regulate SNS activity via GPR41, a Gi/o protein-coupled receptor for SCFAs, at the level of the sympathetic ganglion. GPR41 was most abundantly expressed in sympathetic ganglia in mouse and humans. SCFA propionate promoted sympathetic outflow via GPR41. On the other hand, a ketone body, β-hydroxybutyrate, produced during starvation or diabetes, suppressed SNS activity by antagonizing GPR41. Pharmacological and siRNA experiments indicated that GPR41-mediated activation of sympathetic neurons involves Gβγ-PLCβ-MAPK signaling. Sympathetic regulation by SCFAs and ketone bodies correlated well with their respective effects on energy consumption. These findings establish that SCFAs and ketone bodies directly regulate GPR41-mediated SNS activity and thereby control body energy expenditure in maintaining metabolic homeostasis.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            The Mucin degrader Akkermansia muciniphila is an abundant resident of the human intestinal tract.

            A 16S rRNA-targeted probe, MUC-1437, was designed and validated in order to determine the presence and numbers of cells of Akkermansia muciniphila, a mucin degrader, in the human intestinal tract. As determined by fluorescent in situ hybridization, A. muciniphila accounted more than 1% of the total fecal cells and was shown to be a common bacterial component of the human intestinal tract.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Dietary Polyphenols Promote Growth of the Gut Bacterium Akkermansia muciniphila and Attenuate High-Fat Diet–Induced Metabolic Syndrome

              Dietary polyphenols protect against metabolic syndrome, despite limited absorption and digestion, raising questions about their mechanism of action. We hypothesized that one mechanism may involve the gut microbiota. To test this hypothesis, C57BL/6J mice were fed a high-fat diet (HFD) containing 1% Concord grape polyphenols (GP). Relative to vehicle controls, GP attenuated several effects of HFD feeding, including weight gain, adiposity, serum inflammatory markers (tumor necrosis factor [TNF]α, interleukin [IL]-6, and lipopolysaccharide), and glucose intolerance. GP lowered intestinal expression of inflammatory markers (TNFα, IL-6, inducible nitric oxide synthase) and a gene for glucose absorption (Glut2). GP increased intestinal expression of genes involved in barrier function (occludin) and limiting triglyceride storage (fasting-induced adipocyte factor). GP also increased intestinal gene expression of proglucagon, a precursor of proteins that promote insulin production and gut barrier integrity. 16S rRNA gene sequencing and quantitative PCR of cecal and fecal samples demonstrated that GP dramatically increased the growth of Akkermansia muciniphila and decreased the proportion of Firmicutes to Bacteroidetes, consistent with prior reports that similar changes in microbial community structure can protect from diet-induced obesity and metabolic disease. These data suggest that GP act in the intestine to modify gut microbial community structure, resulting in lower intestinal and systemic inflammation and improved metabolic outcomes. The gut microbiota may thus provide the missing link in the mechanism of action of poorly absorbed dietary polyphenols.
                Bookmark

                Author and article information

                Contributors
                shihof@med.u-toyama.ac.jp
                tobe@med.u-toyama.ac.jp
                Journal
                Sci Rep
                Sci Rep
                Scientific Reports
                Nature Publishing Group UK (London )
                2045-2322
                26 March 2020
                26 March 2020
                2020
                : 10
                : 5544
                Affiliations
                [1 ]ISNI 0000 0001 2171 836X, GRID grid.267346.2, First Department of Internal Medicine, Faculty of Medicine, , University of Toyama, ; Toyama, Japan
                [2 ]ISNI 0000 0001 0702 8004, GRID grid.255137.7, Department of Endocrinology and Metabolism, , Dokkyo Medical University, ; Tochigi, Japan
                [3 ]ISNI 0000 0001 2171 836X, GRID grid.267346.2, Department of Metabolism and Nutrition, Graduate School of Medicine and Pharmaceutical Sciences for Research, , University of Toyama, ; Toyama, Japan
                [4 ]GRID grid.452851.f, Department of community Medical Support, , Toyama University Hospital, ; Toyama, Japan
                [5 ]ISNI 0000 0001 0689 9676, GRID grid.412803.c, Department of Liberal Arts and Sciences, Faculty of Engineering, , Toyama Prefectural University, ; Toyama, Japan
                [6 ]ISNI 0000 0001 2171 836X, GRID grid.267346.2, Division of Nutritional Biochemistry, Institute of Natural Medicine, , University of Toyama, ; Toyama, Japan
                [7 ]ISNI 0000 0001 2171 836X, GRID grid.267346.2, Department of Pathology, , University of Toyama, ; Toyama, Japan
                [8 ]ISNI 0000 0001 0689 9676, GRID grid.412803.c, Department of Pharmaceutical Engineering, Faculty of Engineering, , Toyama Prefectural University, ; Toyama, Japan
                Author information
                http://orcid.org/0000-0001-5539-7605
                Article
                62506
                10.1038/s41598-020-62506-w
                7099031
                32218475
                ba2334ad-b48e-44e4-b27f-0d27c01b770c
                © The Author(s) 2020

                Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 25 September 2019
                : 10 March 2020
                Funding
                Funded by: FundRef https://doi.org/10.13039/501100001691, MEXT | Japan Society for the Promotion of Science (JSPS);
                Award ID: 17K09821
                Award Recipient :
                Funded by: FundRef https://doi.org/10.13039/100007449, Takeda Science Foundation;
                Funded by: FundRef https://doi.org/10.13039/501100005865, Mochida Memorial Foundation for Medical and Pharmaceutical Research;
                Funded by: FundRef https://doi.org/10.13039/100009619, Japan Agency for Medical Research and Development (AMED);
                Award ID: JP18gm6010023h0001
                Award Recipient :
                Funded by: Yakult Bio-Science Foundation
                Funded by: FundRef https://doi.org/10.13039/100008732, Uehara Memorial Foundation;
                Funded by: FundRef https://doi.org/10.13039/100007428, Naito Foundation;
                Funded by: FundRef https://doi.org/10.13039/501100004398, Mitsubishi Foundation;
                Funded by: Strategic PRomotion for practical application of INnovative medical Technology, AMED
                Categories
                Article
                Custom metadata
                © The Author(s) 2020

                Uncategorized
                obesity
                Uncategorized
                obesity

                Comments

                Comment on this article