5
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Simulated spaceflight-induced cardiac remodeling is modulated by gut microbial-derived trimethylamine N-oxide

      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.

          Summary

          Spaceflight is physically demanding and can negatively affect astronauts’ health. It has been shown that the human gut microbiota and cardiac function are affected by spaceflight and simulated spaceflight. This study investigated the effects of the gut microbiota on simulated spaceflight-induced cardiac remodeling using 10° of head-down bed rest (HDBR) in rhesus macaques and 30° of hindlimb unloading (HU) in mice. The gut microbiota, fecal metabolites, and cardiac remodeling were markedly affected by HDBR in macaques and HU in mice, cardiac remodeling in control mice was affected by the gut microbiota of HU mice and that of HU mice was protected by the gut microbiota of control mice, and there was a correlation between cardiac remodeling and the gut microbial-derived metabolite trimethylamine N-oxide. These findings suggest that spaceflight can affect cardiac remodeling by modulating the gut microbiota and fecal metabolites.

          Graphical abstract

          Highlights

          • Cardiac remodeling correlated with TMAO in HDBR macaques

          • Cardiac remodeling modulated by gut microbiota, which correlated with TMAO in HU mice

          • Simulated spaceflight-induced cardiac remodeling modulated by the gut microbiota

          Abstract

          Microbial genetics; Microbial metabolism; Cell biology; Metabolomics; Model organism; Microgravity sciences

          Related collections

          Most cited references38

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

          From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as Key Bacterial Metabolites.

          A compelling set of links between the composition of the gut microbiota, the host diet, and host physiology has emerged. Do these links reflect cause-and-effect relationships, and what might be their mechanistic basis? A growing body of work implicates microbially produced metabolites as crucial executors of diet-based microbial influence on the host. Here, we will review data supporting the diverse functional roles carried out by a major class of bacterial metabolites, the short-chain fatty acids (SCFAs). SCFAs can directly activate G-coupled-receptors, inhibit histone deacetylases, and serve as energy substrates. They thus affect various physiological processes and may contribute to health and disease.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease

            Metabolomics studies hold promise for discovery of pathways linked to disease processes. Cardiovascular disease (CVD) represents the leading cause of death and morbidity worldwide. A metabolomics approach was used to generate unbiased small molecule metabolic profiles in plasma that predict risk for CVD. Three metabolites of the dietary lipid phosphatidylcholine, namely choline, trimethylamine N-oxide (TMAO), and betaine, were identified and then shown to predict risk for CVD in an independent large clinical cohort. Dietary supplementation of mice with choline, TMAO or betaine promoted up-regulation of multiple macrophage scavenger receptors linked to atherosclerosis, and supplementation with choline or TMAO promoted atherosclerosis. Studies using germ-free mice confirmed a critical role for dietary choline and gut flora in TMAO production, augmented macrophage cholesterol accumulation and foam cell formation. Suppression of intestinal microflora in atherosclerosis-prone mice inhibited dietary choline-enhanced atherosclerosis. Genetic variations controlling expression of flavin monooxygenases (FMOs), an enzymatic source of TMAO, segregated with atherosclerosis in hyperlipidemic mice. Discovery of a relationship between gut flora-dependent metabolism of dietary phosphatidylcholine and CVD pathogenesis provides opportunities for development of both novel diagnostic tests and therapeutic approaches for atherosclerotic heart disease.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Functional interactions between the gut microbiota and host metabolism.

              The link between the microbes in the human gut and the development of obesity, cardiovascular disease and metabolic syndromes, such as type 2 diabetes, is becoming clearer. However, because of the complexity of the microbial community, the functional connections are less well understood. Studies in both mice and humans are helping to show what effect the gut microbiota has on host metabolism by improving energy yield from food and modulating dietary or the host-derived compounds that alter host metabolic pathways. Through increased knowledge of the mechanisms involved in the interactions between the microbiota and its host, we will be in a better position to develop treatments for metabolic disease.
                Bookmark

                Author and article information

                Contributors
                Journal
                iScience
                iScience
                iScience
                Elsevier
                2589-0042
                22 November 2023
                15 December 2023
                22 November 2023
                : 26
                : 12
                : 108556
                Affiliations
                [1 ]National Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
                [2 ]Department of Rheumatology and Immunology, First Medical Center of Chinese PLA General Hospital, Beijing, China
                [3 ]School of Aerospace Medicine, Key Laboratory of Aerospace Medicine of the Ministry of Education, Fourth Military Medical University, Xi’an, China
                [4 ]School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
                [5 ]State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
                [6 ]Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
                [7 ]Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China
                Author notes
                []Corresponding author liuzizhong911@ 123456163.com
                [∗∗ ]Corresponding author sh2ling@ 123456126.com
                [∗∗∗ ]Corresponding author yingxianli@ 123456aliyun.com
                [8]

                These authors contributed equally

                [9]

                Lead contact

                Article
                S2589-0042(23)02633-0 108556
                10.1016/j.isci.2023.108556
                10730869
                38125015
                c62bf943-3704-4f49-90d2-31fc4a32de2a
                © 2023 The Authors.

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                : 28 August 2023
                : 19 October 2023
                : 20 November 2023
                Categories
                Article

                microbial genetics,microbial metabolism,cell biology,metabolomics,model organism,microgravity sciences

                Comments

                Comment on this article