11
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Vitamin B12 Deficiency Alters the Gut Microbiota in a Murine Model of Colitis

      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

          Purpose: Inflammatory bowel disease (IBD) refers to a spectrum of autoimmune diseases, which result in chronic intestinal inflammation. Previous findings suggest a role for diet, nutrition and dysbiosis of the gut microbiota in both the development and progression of the condition. Vitamin B12 is a key cofactor of methionine synthase and is produced solely by microbes. Previous work links increased levels of homocysteine, a substrate of methionine synthase, MetH, to IBD indicating a potential role for vitamin B12 deficiency in intestinal injury and inflammation. This study assessed the role of vitamin B12 in shaping the gut microbiota and determining responses to intestinal injury using a reproducible murine model of colitis.

          Methods: The effects of vitamin B12 supplementation and deficiency were assessed in vivo; 3-week-old post-weanling C57Bl/6 mice were divided into three dietary treatment groups: (1) sufficient vitamin B12 (50 mg/Kg), (2) deficient vitamin B12 (0 mg/Kg) and (3) supplemented vitamin B12 (200 mg/Kg) for a period of 4 weeks. Intestinal injury was induced with 2% dextran sodium sulphate (DSS) via drinking water for 5 days. The impact of varying levels of dietary vitamin B12 on gut microbiota composition was assessed using 16S rRNA gene sequencing from fecal samples collected at day 0 and day 28 of the dietary intervention, and 7 days following induction of colitis on day 38, when blood and colonic tissues were also collected.

          Results: No significant alterations were found in the gut microbiota composition of disease-free animals in response to dietary interventions. By contrast, after DSS-induced colitis, >30 genera were significantly altered in vitamin B12 deficient mice. Altered B12 levels produced no significant effect on composite disease-activity scores; however, administration of a B12 deficient diet resulted in reduced DSS-induced epithelial tissue damage.

          Conclusions: Vitamin B12 supplementation does not alter the gut microbiota composition under healthy conditions, but does contribute to differential microbial responses and intestinal dysbiosis following the induction of experimental colitis.

          Related collections

          Most cited references35

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

          Lymphoid tissue genesis induced by commensals through NOD1 regulates intestinal homeostasis.

          Intestinal homeostasis is critical for efficient energy extraction from food and protection from pathogens. Its disruption can lead to an array of severe illnesses with major impacts on public health, such as inflammatory bowel disease characterized by self-destructive intestinal immunity. However, the mechanisms regulating the equilibrium between the large bacterial flora and the immune system remain unclear. Intestinal lymphoid tissues generate flora-reactive IgA-producing B cells, and include Peyer's patches and mesenteric lymph nodes, as well as numerous isolated lymphoid follicles (ILFs). Here we show that peptidoglycan from Gram-negative bacteria is necessary and sufficient to induce the genesis of ILFs in mice through recognition by the NOD1 (nucleotide-binding oligomerization domain containing 1) innate receptor in epithelial cells, and beta-defensin 3- and CCL20-mediated signalling through the chemokine receptor CCR6. Maturation of ILFs into large B-cell clusters requires subsequent detection of bacteria by toll-like receptors. In the absence of ILFs, the composition of the intestinal bacterial community is profoundly altered. Our results demonstrate that intestinal bacterial commensals and the immune system communicate through an innate detection system to generate adaptive lymphoid tissues and maintain intestinal homeostasis.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            The ribosomal database project (RDP-II): introducing myRDP space and quality controlled public data

            Substantial new features have been implemented at the Ribosomal Database Project in response to the increased importance of high-throughput rRNA sequence analysis in microbial ecology and related disciplines. The most important changes include quality analysis, including chimera detection, for all available rRNA sequences and the introduction of myRDP Space, a new web component designed to help researchers place their own data in context with the RDP's data. In addition, new video tutorials describe how to use RDP features. Details about RDP data and analytical functions can be found at the RDP-II website ().
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Homocysteine metabolism.

              J Selhub (1999)
              Homocysteine is a sulfur amino acid whose metabolism stands at the intersection of two pathways: remethylation to methionine, which requires folate and vitamin B12 (or betaine in an alternative reaction); and transsulfuration to cystathionine, which requires pyridoxal-5'-phosphate. The two pathways are coordinated by S-adenosylmethionine, which acts as an allosteric inhibitor of the methylenetetrahydrofolate reductase reaction and as an activator of cystathionine beta-synthase. Hyperhomocysteinemia, a condition that recent epidemiological studies have shown to be associated with increased risk of vascular disease, arises from disrupted homocysteine metabolism. Severe hyperhomocysteinemia is due to rare genetic defects resulting in deficiencies in cystathionine beta synthase, methylenetetrahydrofolate reductase, or in enzymes involved in methyl-B12 synthesis and homocysteine methylation. Mild hyperhomocysteinemia seen in fasting conditions is due to mild impairment in the methylation pathway (i.e. folate or B12 deficiencies or methylenetetrahydrofolate reductase thermolability). Post-methionine-load hyperhomocysteinemia may be due to heterozygous cystathionine beta-synthase defect or B6 deficiency. Early studies with nonphysiological high homocysteine levels showed a variety of deleterious effects on endothelial or smooth muscle cells in culture. More recent studies with human beings and animals with mild hyperhomocysteinemia provided encouraging results in the attempt to understand the mechanism that underlies this relationship between mild elevations of plasma homocysteine and vascular disease. The studies with animal models indicated the possibility that the effect of elevated homocysteine is multifactorial, affecting both the vascular wall structure and the blood coagulation system.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Nutr
                Front Nutr
                Front. Nutr.
                Frontiers in Nutrition
                Frontiers Media S.A.
                2296-861X
                05 June 2020
                2020
                : 7
                : 83
                Affiliations
                [1] 1Cell Biology Program, Research Institute, Hospital for Sick Children , Toronto, ON, Canada
                [2] 2Division of Gastroenterology, Hepatology and Nutrition, Department of Paediatrics , Toronto, ON, Canada
                [3] 3Department of Laboratory Medicine and Pathobiology, University of Toronto , Toronto, ON, Canada
                [4] 4Physiology and Experimental Medicine, Research Institute, Hospital for Sick Children , Toronto, ON, Canada
                [5] 5Department of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University , Hamilton, ON, Canada
                [6] 6Division of General and Thoracic Surgery, Hospital for Sick Children , Toronto, ON, Canada
                [7] 7Farncombe Family Digestive Health Institute, McMaster University , Hamilton, ON, Canada
                Author notes

                Edited by: Silvia Turroni, University of Bologna, Italy

                Reviewed by: Nobuhiko Kamada, University of Michigan, United States; Ravinder Nagpal, Wake Forest School of Medicine, United States

                *Correspondence: Philip M. Sherman philip.sherman@ 123456sickkids.ca

                This article was submitted to Nutrition and Microbes, a section of the journal Frontiers in Nutrition

                †Present address: Eberhard Lurz, Division of Gastroenterology and Hepatology, Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany

                †These authors have shared co-first authorship

                Article
                10.3389/fnut.2020.00083
                7291859
                32582756
                dd9b7ec3-7c70-445a-a80c-b83ef4b51d52
                Copyright © 2020 Lurz, Horne, Määttänen, Wu, Botts, Li, Rossi, Johnson-Henry, Pierro, Surette and Sherman.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 06 March 2020
                : 07 May 2020
                Page count
                Figures: 6, Tables: 1, Equations: 0, References: 50, Pages: 12, Words: 7336
                Funding
                Funded by: Canadian Institutes of Health Research 10.13039/501100000024
                Categories
                Nutrition
                Original Research

                vitamin b12,inflammation,microbiome,inflammatory bowel disease,colitis

                Comments

                Comment on this article