34
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Involvement of a gut–retina axis in protection against dietary glycemia-induced age-related macular degeneration

      research-article
      a , a , b , c , d , a , a , e , a , f , g , h , h , i , i , f , j , j , j , j , a , b , c , b , c , b , c , b , c , a , 2
      Proceedings of the National Academy of Sciences of the United States of America
      National Academy of Sciences
      age-related macular degeneration, glycemic index, advanced glycation end-product, gut microbiome, metabolomics

      Read this article at

      ScienceOpenPublisherPMC
      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

          Food is medicine, and diet impacts the risk for and progression of age-related macular degeneration AMD, but we have few clues as to why. We found that wild-type mice fed a high-glycemic-index diet similar in composition to the Western diet developed a disease state that resembles dry AMD. To gain insight into the mechanism, we used LC-MS– and NMR-based metabolomics to discover diet-, metabolic-, and AMD-associated phenotypes. These studies revealed changes in the gut microbiota that altered the production of metabolites that protected against AMD, including serotonin. Changing the diet to a low-glycemic-index diet, even late in life, arrested the development of AMD, offering dietary interventions for AMD.

          Abstract

          Age-related macular degeneration (AMD) is the major cause of blindness in developed nations. AMD is characterized by retinal pigmented epithelial (RPE) cell dysfunction and loss of photoreceptor cells. Epidemiologic studies indicate important contributions of dietary patterns to the risk for AMD, but the mechanisms relating diet to disease remain unclear. Here we investigate the effect on AMD of isocaloric diets that differ only in the type of dietary carbohydrate in a wild-type aged-mouse model. The consumption of a high-glycemia (HG) diet resulted in many AMD features (AMDf), including RPE hypopigmentation and atrophy, lipofuscin accumulation, and photoreceptor degeneration, whereas consumption of the lower-glycemia (LG) diet did not. Critically, switching from the HG to the LG diet late in life arrested or reversed AMDf. LG diets limited the accumulation of advanced glycation end products, long-chain polyunsaturated lipids, and their peroxidation end-products and increased C3-carnitine in retina, plasma, or urine. Untargeted metabolomics revealed microbial cometabolites, particularly serotonin, as protective against AMDf. Gut microbiota were responsive to diet, and we identified microbiota in the Clostridiales order as being associated with AMDf and the HG diet, whereas protection from AMDf was associated with the Bacteroidales order and the LG diet. Network analysis revealed a nexus of metabolites and microbiota that appear to act within a gut–retina axis to protect against diet- and age-induced AMDf. The findings indicate a functional interaction between dietary carbohydrates, the metabolome, including microbial cometabolites, and AMDf. Our studies suggest a simple dietary intervention that may be useful in patients to arrest AMD.

          Related collections

          Most cited references55

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          HMDB: a knowledgebase for the human metabolome

          The Human Metabolome Database (HMDB, http://www.hmdb.ca) is a richly annotated resource that is designed to address the broad needs of biochemists, clinical chemists, physicians, medical geneticists, nutritionists and members of the metabolomics community. Since its first release in 2007, the HMDB has been used to facilitate the research for nearly 100 published studies in metabolomics, clinical biochemistry and systems biology. The most recent release of HMDB (version 2.0) has been significantly expanded and enhanced over the previous release (version 1.0). In particular, the number of fully annotated metabolite entries has grown from 2180 to more than 6800 (a 300% increase), while the number of metabolites with biofluid or tissue concentration data has grown by a factor of five (from 883 to 4413). Similarly, the number of purified compounds with reference to NMR, LC-MS and GC-MS spectra has more than doubled (from 380 to more than 790 compounds). In addition to this significant expansion in database size, many new database searching tools and new data content has been added or enhanced. These include better algorithms for spectral searching and matching, more powerful chemical substructure searches, faster text searching software, as well as dedicated pathway searching tools and customized, clickable metabolic maps. Changes to the user-interface have also been implemented to accommodate future expansion and to make database navigation much easier. These improvements should make the HMDB much more useful to a much wider community of users.
            Bookmark
            • Record: found
            • Abstract: not found
            • Article: not found

            OPLS discriminant analysis: combining the strengths of PLS-DA and SIMCA classification

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

              Oxidative damage-induced inflammation initiates age-related macular degeneration.

              Oxidative damage and inflammation are postulated to be involved in age-related macular degeneration (AMD). However, the molecular signal(s) linking oxidation to inflammation in this late-onset disease is unknown. Here we describe AMD-like lesions in mice after immunization with mouse serum albumin adducted with carboxyethylpyrrole, a unique oxidation fragment of docosahexaenoic acid that has previously been found adducting proteins in drusen from AMD donor eye tissues and in plasma samples from individuals with AMD. Immunized mice develop antibodies to this hapten, fix complement component-3 in Bruch's membrane, accumulate drusen below the retinal pigment epithelium during aging, and develop lesions in the retinal pigment epithelium mimicking geographic atrophy, the blinding end-stage condition characteristic of the dry form of AMD. We hypothesize that these mice are sensitized to the generation of carboxyethylpyrrole adducts in the outer retina, where docosahexaenoic acid is abundant and conditions for oxidative damage are permissive. This new model provides a platform for dissecting the molecular pathology of oxidative damage in the outer retina and the immune response contributing to AMD.
                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
                30 May 2017
                15 May 2017
                : 114
                : 22
                : E4472-E4481
                Affiliations
                [1] aJean Mayer-US Department of Agriculture Human Nutrition Research Center on Aging, Tufts University , Boston, MA 02111;
                [2] bDepartment of Computer Science and Applied Mathematics, Weizmann Institute of Science , Rehovot 7610001, Israel;
                [3] cDepartment of Molecular Cell Biology, Weizmann Institute of Science , Rehovot 7610001, Israel;
                [4] dDepartment of Plant Sciences, Weizmann Institute of Technology , Rehovot 7610001, Israel;
                [5] eDepartment of Pathology and Laboratory Medicine, Tufts University School of Medicine , Boston, MA 02111;
                [6] fDepartment of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine , Boston, MA 02111;
                [7] gLaboratory of Food and Regulation Biology, Graduate School of Agriculture, Tokai University , Kawayou, Minamiaso, Aso-gun, Kumamoto, 869-1404, Japan;
                [8] hDiabetes Research Center, Departments of Medicine and Pathology, Albert Einstein College of Medicine , Bronx, NY 10461;
                [9] iClinical Sciences Research Laboratories, Warwick Medical School, University of Warwick, University Hospital , Coventry CV2 2DX, United Kingdom;
                [10] j Eli and Edythe L. Broad Institute of MIT and Harvard , Cambridge, MA 02142
                Author notes
                2To whom correspondence should be addressed. Email: allen.taylor@ 123456tufts.edu .

                Edited by Catherine Bowes Rickman, Duke University Medical Center, Durham, NC, and accepted by Editorial Board Member Jeremy Nathans April 20, 2017 (received for review February 9, 2017)

                Author contributions: S.R., D.E.S., and A.T. designed research; S.R., S.J., M.-L.C., J. Szelog, C.C., K.D., C.M., N.R., A.A.D., K.A.P., J.M.S., A.W., T.A.-S., and M.L.-P. performed research; R.N., X.-L.D., and M.B. contributed new reagents/analytic tools; S.R., T.K., J. Szymanski, C.M., N.R., P.J.T., J.D.B., C.B.C., and E.S. analyzed data; and S.R. and A.T. wrote the paper.

                1S.J., T.K., and J. Szymanski contributed equally to this work.

                Author information
                http://orcid.org/0000-0001-8078-640X
                Article
                PMC5465926 PMC5465926 5465926 201702302
                10.1073/pnas.1702302114
                5465926
                28507131
                cfc95a26-c2f3-402c-a29e-4d3435c7a9ed
                History
                Page count
                Pages: 10
                Funding
                Funded by: HHS | NIH | National Eye Institute (NEI) 100000053
                Award ID: RO1EY021212
                Funded by: HHS | NIH | National Eye Institute (NEI) 100000053
                Award ID: RO1EY13250
                Funded by: HHS | NIH | National Eye Institute (NEI) 100000053
                Award ID: RO1EY026979
                Funded by: U.S. Department of Agriculture (USDA) 100000199
                Award ID: 58-1950-0-014
                Funded by: U.S. Department of Agriculture (USDA) 100000199
                Award ID: 58-1950-4-003
                Categories
                PNAS Plus
                Biological Sciences
                Medical Sciences
                PNAS Plus

                glycemic index,age-related macular degeneration,advanced glycation end-product,gut microbiome,metabolomics

                Comments

                Comment on this article