16
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Mechanisms by which sialylated milk oligosaccharides impact bone biology in a gnotobiotic mouse model of infant undernutrition

      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.

          Significance

          Identifying components of breast milk that influence postnatal development though their effects on the gut microbiota and immune system could provide new therapeutic approaches for childhood undernutrition, including heretofore treatment-refractory linear growth faltering (stunting). Plasma biomarkers of osteoclast-mediated bone resorption and osteoblast-driven bone formation in stunted Bangladeshi children provided evidence for elevated osteoclastic activity. Gnotobiotic mice, colonized with a stunted infant’s gut microbiota, exhibited decreased bone resorption when consuming diets supplemented with a purified bovine oligosaccharide mixture dominated by sialylated structures found in human breast milk. Supplementation decreased osteoclastogenesis while sparing osteoblast activity; the microbiota, intestinal cell populations, and immune mediators contribute to these responses. The influence of milk oligosaccharides on the gut microbiota–bone axis has diagnostic and therapeutic implications.

          Abstract

          Undernutrition in children is a pressing global health problem, manifested in part by impaired linear growth (stunting). Current nutritional interventions have been largely ineffective in overcoming stunting, emphasizing the need to obtain better understanding of its underlying causes. Treating Bangladeshi children with severe acute malnutrition with therapeutic foods reduced plasma levels of a biomarker of osteoclastic activity without affecting biomarkers of osteoblastic activity or improving their severe stunting. To characterize interactions among the gut microbiota, human milk oligosaccharides (HMOs), and osteoclast and osteoblast biology, young germ-free mice were colonized with cultured bacterial strains from a 6-mo-old stunted infant and fed a diet mimicking that consumed by the donor population. Adding purified bovine sialylated milk oligosaccharides (S-BMO) with structures similar to those in human milk to this diet increased femoral trabecular bone volume and cortical thickness, reduced osteoclasts and their bone marrow progenitors, and altered regulators of osteoclastogenesis and mediators of Th2 responses. Comparisons of germ-free and colonized mice revealed S-BMO-dependent and microbiota-dependent increases in cecal levels of succinate, increased numbers of small intestinal tuft cells, and evidence for activation of a succinate-induced tuft cell signaling pathway linked to Th2 immune responses. A prominent fucosylated HMO, 2′-fucosyllactose, failed to elicit these changes in bone biology, highlighting the structural specificity of the S-BMO effects. These results underscore the need to further characterize the balance between, and determinants of, osteoclastic and osteoblastic activity in stunted infants/children, and suggest that certain milk oligosaccharides may have therapeutic utility in this setting.

          Related collections

          Most cited references51

          • Record: found
          • Abstract: found
          • Article: found

          Maternal and child undernutrition and overweight in low-income and middle-income countries

          The Lancet, 382(9890), 427-451
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Evidence-based interventions for improvement of maternal and child nutrition: what can be done and at what cost?

            The Lancet, 382(9890), 452-477
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Tuft cells, taste-chemosensory cells, orchestrate parasite type 2 immunity in the gut.

              The intestinal epithelium forms an essential barrier between a host and its microbiota. Protozoa and helminths are members of the gut microbiota of mammals, including humans, yet the many ways that gut epithelial cells orchestrate responses to these eukaryotes remain unclear. Here we show that tuft cells, which are taste-chemosensory epithelial cells, accumulate during parasite colonization and infection. Disruption of chemosensory signaling through the loss of TRMP5 abrogates the expansion of tuft cells, goblet cells, eosinophils, and type 2 innate lymphoid cells during parasite colonization. Tuft cells are the primary source of the parasite-induced cytokine interleukin-25, which indirectly induces tuft cell expansion by promoting interleukin-13 production by innate lymphoid cells. Our results identify intestinal tuft cells as critical sentinels in the gut epithelium that promote type 2 immunity in response to intestinal parasites.
                Bookmark

                Author and article information

                Journal
                Proc Natl Acad Sci U S A
                Proc. Natl. Acad. Sci. U.S.A
                pnas
                pnas
                PNAS
                Proceedings of the National Academy of Sciences of the United States of America
                National Academy of Sciences
                0027-8424
                1091-6490
                11 June 2019
                28 May 2019
                28 May 2019
                : 116
                : 24
                : 11988-11996
                Affiliations
                [1] aEdison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine , St. Louis, MO 63110;
                [2] bCenter for Gut Microbiome and Nutrition Research, Washington University School of Medicine , St. Louis, MO 63110;
                [3] cDepartment of Genetics, Washington University School of Medicine , St. Louis, MO 63110;
                [4] dGenome Technology Access Center, Washington University School of Medicine , St. Louis, MO 63110;
                [5] eFoods for Health Institute, University of California, Davis , CA 95616;
                [6] fDepartment of Food Science and Technology, University of California , Davis, CA 95616;
                [7] gInternational Centre for Diarrhoeal Disease Research , 1212 Dhaka, Bangladesh
                Author notes
                1To whom correspondence should be addressed. Email: jgordon@ 123456wustl.edu .

                Contributed by Jeffrey I. Gordon, April 10, 2019 (sent for review December 24, 2018; reviewed by Martin J. Blaser and Sharon M. Donovan)

                Author contributions: C.A.C., M.J.B., S.H., T.A., and J.I.G. designed research; C.A.C., P.P.A., V.L.K., J.C., J.L.G., S.H., and T.A. performed research; R.D.H., D.B., and D.A.M. contributed new reagents/analytic tools; C.A.C., P.P.A., V.L.K., M.C.H., J.C., V.S., M.J.B., and J.I.G. analyzed data; and C.A.C., M.J.B., and J.I.G. wrote the paper.

                Reviewers: M.J.B., NYU Langone Medical Center; and S.M.D., University of Illinois.

                Author information
                http://orcid.org/0000-0002-8225-8261
                http://orcid.org/0000-0001-6520-677X
                Article
                201821770
                10.1073/pnas.1821770116
                6575181
                31138692
                88c04012-79b9-4fb6-81ba-2e7408a913e7
                Copyright © 2019 the Author(s). Published by PNAS.

                This open access article is distributed under Creative Commons Attribution-NonCommercial-NoDerivatives License 4.0 (CC BY-NC-ND).

                History
                Page count
                Pages: 9
                Funding
                Funded by: HHS | NIH | National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) 100000062
                Award ID: DK30292
                Award Recipient : Matthew C. Hibberd Award Recipient : Jiye Cheng Award Recipient : Jeffrey I. Gordon
                Funded by: HHS | NIH | National Center for Complementary and Integrative Health (NCCIH) 100008460
                Award ID: AT008759
                Award Recipient : David A. Mills
                Funded by: HHS | NIH | National Institute of Allergy and Infectious Diseases (NIAID) 100000060
                Award ID: AI007172-38
                Award Recipient : Carrie Cowardin
                Funded by: HHS | NIH | National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) 100000069
                Award ID: AR057235
                Award Recipient : Jeffrey I. Gordon
                Funded by: Bill and Melinda Gates Foundation (Bill & Melinda Gates Foundation) 100000865
                Award ID: OPP1134649; 1169434
                Award Recipient : Carrie Cowardin Award Recipient : Philip P. Ahern Award Recipient : Vanderlene L. Kung Award Recipient : Matthew C. Hibberd Award Recipient : Jiye Cheng Award Recipient : Janaki L Guruge Award Recipient : Vinaik Sundaresan Award Recipient : Michael J. Barratt Award Recipient : Jeffrey I. Gordon
                Categories
                PNAS Plus
                Biological Sciences
                Medical Sciences
                PNAS Plus

                childhood undernutrition,stunting,gut microbiota,bone growth,breast milk oligosaccharides

                Comments

                Comment on this article