12
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Effects of Yulin Tong Bu formula on modulating gut microbiota and fecal metabolite interactions in mice with polycystic ovary syndrome

      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

          Background

          Polycystic ovarian syndrome (PCOS) is a common endocrine disorder characterized by hyperandrogenism, ovarian dysfunction and polycystic ovarian morphology. Gut microbiota dysbiosis and metabolite are associated with PCOS clinical parameters. Yulin Tong Bu formula (YLTB), a traditional Chinese medicine formula, has been recently indicated to be capable of ameliorating polycystic ovary symptoms and correcting abnormal glucose metabolism. However, the therapeutic mechanism of YLTB on PCOS has not been fully elucidated.

          Methods

          A pseudo sterile mouse model was established during this four-day acclimatization phase by giving the animals an antibiotic cocktail to remove the gut microbiota. Here, the therapeutic effects of YLTB on PCOS were investigated using dehydroepiandrosterone plus high-fat diet-induced PCOS mice model. Female prepuberal mice were randomly divided into three groups; namely, the control group, PCOS group and YLTB (38.68 g·kg -1·day -1) group. To test whether this effect is associated with the gut microbiota, we performed 16S rRNA sequencing studies to analyze the fecal microbiota of mice. The relationships among metabolites, gut microbiota, and PCOS phenotypes were further explored by using Spearman correlation analysis. Then, the effect of metabolite ferulic acid was then validated in PCOS mice.

          Results

          Our results showed that YLTB treatment ameliorated PCOS features (ovarian dysfunction, delayed glucose clearance, decreased insulin sensitivity, deregulation of glucolipid metabolism and hormones, etc.) and significantly attenuated PCOS gut microbiota dysbiosis. Spearman correlation analysis showed that metabolites such as ferulic acid and folic acid are negatively correlated with PCOS clinical parameters. The effect of ferulic acid was similar to that of YLTB. In addition, the bacterial species such as Bacteroides dorei and Bacteroides fragilis were found to be positively related to PCOS clinical parameters, using the association study analysis.

          Conclusion

          These results suggest that YLTB treatment systematically regulates the interaction between the gut microbiota and the associated metabolites to ameliorate PCOS, providing a solid theoretical basis for further validation of YLTB effect on human PCOS trials.

          Graphical abstract

          Related collections

          Most cited references64

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

          Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits.

          Soluble dietary fibers promote metabolic benefits on body weight and glucose control, but underlying mechanisms are poorly understood. Recent evidence indicates that intestinal gluconeogenesis (IGN) has beneficial effects on glucose and energy homeostasis. Here, we show that the short-chain fatty acids (SCFAs) propionate and butyrate, which are generated by fermentation of soluble fiber by the gut microbiota, activate IGN via complementary mechanisms. Butyrate activates IGN gene expression through a cAMP-dependent mechanism, while propionate, itself a substrate of IGN, activates IGN gene expression via a gut-brain neural circuit involving the fatty acid receptor FFAR3. The metabolic benefits on body weight and glucose control induced by SCFAs or dietary fiber in normal mice are absent in mice deficient for IGN, despite similar modifications in gut microbiota composition. Thus, the regulation of IGN is necessary for the metabolic benefits associated with SCFAs and soluble fiber. Copyright © 2014 Elsevier Inc. All rights reserved.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Polycystic ovary syndrome: definition, aetiology, diagnosis and treatment

            Polycystic ovary syndrome (PCOS) is one of the most common endocrine and metabolic disorders in premenopausal women. Heterogeneous by nature, PCOS is defined by a combination of signs and symptoms of androgen excess and ovarian dysfunction in the absence of other specific diagnoses. The aetiology of this syndrome remains largely unknown, but mounting evidence suggests that PCOS might be a complex multigenic disorder with strong epigenetic and environmental influences, including diet and lifestyle factors. PCOS is frequently associated with abdominal adiposity, insulin resistance, obesity, metabolic disorders and cardiovascular risk factors. The diagnosis and treatment of PCOS are not complicated, requiring only the judicious application of a few well-standardized diagnostic methods and appropriate therapeutic approaches addressing hyperandrogenism, the consequences of ovarian dysfunction and the associated metabolic disorders. This article aims to provide a balanced review of the latest advances and current limitations in our knowledge about PCOS while also providing a few clear and simple principles, based on current evidence-based clinical guidelines, for the proper diagnosis and long-term clinical management of women with PCOS.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Gut microbiota–bile acid–interleukin-22 axis orchestrates polycystic ovary syndrome

              Polycystic ovary syndrome (PCOS) is characterized by androgen excess, ovulatory dysfunction and polycystic ovaries1, and is often accompanied by insulin resistance2. The mechanism of ovulatory dysfunction and insulin resistance in PCOS remains elusive, thus limiting the development of therapeutics. Improved metabolic health is associated with a relatively high microbiota gene content and increased microbial diversity3,4. This study aimed to investigate the impact of the gut microbiota and its metabolites on the regulation of PCOS-associated ovarian dysfunction and insulin resistance. Here, we report that Bacteroides vulgatus was markedly elevated in the gut microbiota of individuals with PCOS, accompanied by reduced glycodeoxycholic acid and tauroursodeoxycholic acid levels. Transplantation of fecal microbiota from women with PCOS or B. vulgatus-colonized recipient mice resulted in increased disruption of ovarian functions, insulin resistance, altered bile acid metabolism, reduced interleukin-22 secretion and infertility. Mechanistically, glycodeoxycholic acid induced intestinal group 3 innate lymphoid cell IL-22 secretion through GATA binding protein 3, and IL-22 in turn improved the PCOS phenotype. This finding is consistent with the reduced levels of IL-22 in individuals with PCOS. This study suggests that modifying the gut microbiota, altering bile acid metabolism and/or increasing IL-22 levels may be of value for the treatment of PCOS.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Endocrinol (Lausanne)
                Front Endocrinol (Lausanne)
                Front. Endocrinol.
                Frontiers in Endocrinology
                Frontiers Media S.A.
                1664-2392
                06 February 2023
                2023
                : 14
                : 1122709
                Affiliations
                [1] 1 Department of Herbal Medicine, Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, School of traditional Chinese Medicine, Chongqing Medical University , Chongqing, China
                [2] 2 Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University , Chongqing, China
                [3] 3 Department of Physiology, School of Basic Medicine, Chongqing Medical University , Chongqing, China
                [4] 4 Department of Gynecology, Chongqing Hospital of Traditional Chinese Medicine , Chongqing, China
                [5] 5 Department of Pharmacology, Academician Workstation, Changsha Medical University , Changsha, China
                [6] 6 Department of Obstetrics and Gynecology, Chongqing General Hospital, University of Chinese Academy of Sciences , Chongqing, China
                Author notes

                Edited by: Shunfeng Cheng, Qingdao Agricultural University, China

                Reviewed by: Wen-Xiang Liu, Inner Mongolia University, China; Guruswami Gurusubramanian, Mizoram University, India

                *Correspondence: Li-Juan Fu, fulijuan@ 123456cqmu.edu.cn ; Qian Feng, 469280497@ 123456qq.com

                †These authors have contributed equally to this work and share first authorship

                This article was submitted to Reproduction, a section of the journal Frontiers in Endocrinology

                Article
                10.3389/fendo.2023.1122709
                9939769
                1893df70-87a5-4507-b6f8-267a2d4a7d1c
                Copyright © 2023 Su, Wang, Yang, Wu, Xia, Bao, Ding, Feng and Fu

                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
                : 13 December 2022
                : 24 January 2023
                Page count
                Figures: 9, Tables: 4, Equations: 1, References: 64, Pages: 19, Words: 8561
                Funding
                Funded by: National Natural Science Foundation of China , doi 10.13039/501100001809;
                Funded by: Natural Science Foundation of Chongqing , doi 10.13039/501100005230;
                Funded by: Natural Science Foundation of Chongqing , doi 10.13039/501100005230;
                Funded by: Science-Health Joint Medical Scientific Research Project of Chongqing , doi 10.13039/100017501;
                Funded by: Natural Science Foundation of Chongqing , doi 10.13039/501100005230;
                Funded by: Chongqing Municipal Public Health Bureau, Chongqing People's Municipal Government , doi 10.13039/501100003075;
                Funded by: National Natural Science Foundation of China , doi 10.13039/501100001809;
                This work was funded by grants from National Natural Science of Foundation of China (Nos. 82104923, 82171624), Natural Science of Foundation of Chongqing (Nos. cstc2021jcyj-msxmX0236, cstc2021jcyj-msxmX0900, cstc2019jxjl130030) and Chongqing medical scientific research project (Joint project of Chongqing Health Commission and Science and Technology) (No.2022QNXM042), Chongqing Municipal Health Bureau Chinese Medicine Technology Project (serial number: ZY201802044).
                Categories
                Endocrinology
                Original Research

                Endocrinology & Diabetes
                polycystic ovary syndrome,yltb formula,gut microbiota,metabolites,ferulic acid

                Comments

                Comment on this article