1
|
Wang S, Xu Z, Wang Z, Yi X, Wu J. M6A methyltransferase METTL3 promotes glucose metabolism hub gene expression and induces metabolic dysfunction-associated steatotic liver disease (MASLD). BMC Genomics 2025; 26:188. [PMID: 39994526 PMCID: PMC11853331 DOI: 10.1186/s12864-025-11377-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 02/18/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND N6-methyladenosine (m6A) RNA modification plays a crucial role in various biological events and is implicated in various metabolic-related diseases. However, its role in MASLD remains unclear. This study aims to investigate the impact of METTL3 on MASLD through multi-omics analysis, with a focus on exploring its potential mechanisms of action. METHODS An MASLD mouse model was established by feeding C57BL/6J mice a high-fat diet for 12 weeks. A METTL3 stable overexpression AML12 cell model was also constructed via lentiviral transfection. Subsequent transcriptomic and proteomic analyses, as well as integrated analysis between different omics datasets, were conducted. RESULTS METTL3 expression was significantly increased in the MASLD mouse model. Through our transcriptomic and proteomic analyses, we identified 848 genes with significant inconsistencies between the transcriptomic and proteomic datasets. GO/ KEGG enrichment analyses identified terms that may be involved in post-transcriptional modifications, particularly METTL3-mediated m6A modification. Subsequently, through integrated proteomic analysis of the METTL3-overexpressed AML12 cell model and the MASLD mouse model, we selected the top 20 co-upregulated and co-downregulated GO/ KEGG terms as the main biological processes influenced by METTL3 during MASLD. By intersecting with pathways obtained from previous integrated analyses, we identified GO/ KEGG terms affected by METTL3-induced m6A modification. Protein-protein interaction analysis of proteins involved in these pathways highlighted GAPDH and TPI1 as two key hub genes. CONCLUSIONS During MASLD, METTL3 regulates the glycolytic pathway through m6A modification, influencing the occurrence and development of the disease via the key hub genes GAPDH and TPI1. These findings expand our understanding of MASLD and provide strong evidence for potential therapeutic targets and drug development.
Collapse
Affiliation(s)
- Shuowen Wang
- Gastroenterology Department, Children's Hospital Capital Institute of Pediatrics, Beijing, 100020, China
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
- Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Ziying Xu
- Bacteriology Department, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Zijun Wang
- Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Xiaoyu Yi
- Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Jianxin Wu
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China.
- Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, 100020, China.
- Beijing Tongren Hospital, Capital Medical University, Beijing, 100005, China.
| |
Collapse
|
2
|
Tsinopoulou VR, Kotanidou EP, Athanasiadis N, Sapountzi E, Bacopoulou F, Ntzani E, Galli-Tsinopoulou A, Christoforidis A. The Role of the Gut Microbiome in Youth with Polycystic Ovary Syndrome: A Systematic Review. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1872. [PMID: 38136074 PMCID: PMC10741487 DOI: 10.3390/children10121872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a common endocrine disorder that affects women of reproductive age and female adolescents. The diagnosis of PCOS is difficult during puberty due to overlapping of the criteria with normal variations of menstruation during this age period. There are insufficient data on the gut microbiome and PCOS and potential mechanisms linking the two. The present systematic review aimed to detect dysbiosis patterns in youth with PCOS, compared with healthy controls. METHODS One hundred seventy-eight studies were identified by a databases search and sixty-eight by a full-text assessment for eligibility; four were included in the systematic review and underwent quality control. RESULTS The results of the study were controversial in accordance to findings from the literature. A change in gut microbiome α diversity was found in PCOS adolescents, with no significant alterations in β diversity. Almost all studies found Firmicutes, Bacteroidetes, and Actinobacteria in abundance in both groups, with changes in family composition and fluctuations at the phylum level. A statistically significant association between these changes and clinical or biochemical features of the syndrome was described. CONCLUSIONS This systematic review confirmed gut microbiota dysbiosis in youth with PCOS. However, further data are needed to clarify these changes and to build a strategy to prevent the syndrome.
Collapse
Affiliation(s)
- Vasiliki-Rengina Tsinopoulou
- Program of Postgraduate Studies “Adolescent Medicine and Adolescent Health Care”, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.P.K.); (A.G.-T.); (A.C.)
- 2nd Department of Pediatrics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA University General Hospital, Stilponos Kyriakidi 1, 54636 Thessaloniki, Greece
| | - Eleni P. Kotanidou
- Program of Postgraduate Studies “Adolescent Medicine and Adolescent Health Care”, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.P.K.); (A.G.-T.); (A.C.)
- 2nd Department of Pediatrics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA University General Hospital, Stilponos Kyriakidi 1, 54636 Thessaloniki, Greece
| | - Nikolaos Athanasiadis
- 3rd Department of Obstetrics and Gynecology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Ippokration General Hospital, 54642 Thessaloniki, Greece
| | - Evdoxia Sapountzi
- 2nd Department of Pediatrics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA University General Hospital, Stilponos Kyriakidi 1, 54636 Thessaloniki, Greece
| | - Flora Bacopoulou
- Center for Adolescent Medicine and UNESCO Chair in Adolescent Health Care, 1st Department of Pediatrics, School of Medicine, National and Kapodistrian University of Athens, Aghia Sophia Children’s Hospital Athens, 11527 Athens, Greece;
| | - Evangelia Ntzani
- Department of Hygiene and Epidemiology, School of Medicine, University of Ioannina, 45500 Ioannina, Greece;
| | - Assimina Galli-Tsinopoulou
- Program of Postgraduate Studies “Adolescent Medicine and Adolescent Health Care”, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.P.K.); (A.G.-T.); (A.C.)
- 2nd Department of Pediatrics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, AHEPA University General Hospital, Stilponos Kyriakidi 1, 54636 Thessaloniki, Greece
| | - Athanasios Christoforidis
- Program of Postgraduate Studies “Adolescent Medicine and Adolescent Health Care”, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (E.P.K.); (A.G.-T.); (A.C.)
- 1st Department of Pediatrics, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Ippokration General Hospital, 54636 Thessaloniki, Greece
| |
Collapse
|
3
|
Sawada K, Chung H, Softic S, Moreno-Fernandez ME, Divanovic S. The bidirectional immune crosstalk in metabolic dysfunction-associated steatotic liver disease. Cell Metab 2023; 35:1852-1871. [PMID: 37939656 PMCID: PMC10680147 DOI: 10.1016/j.cmet.2023.10.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is an unabated risk factor for end-stage liver diseases with no available therapies. Dysregulated immune responses are critical culprits of MASLD pathogenesis. Independent contributions from either the innate or adaptive arms of the immune system or their unidirectional interplay are commonly studied in MASLD. However, the bidirectional communication between innate and adaptive immune systems and its impact on MASLD remain insufficiently understood. Given that both innate and adaptive immune cells are indispensable for the development and progression of inflammation in MASLD, elucidating pathogenic contributions stemming from the bidirectional interplay between these two arms holds potential for development of novel therapeutics for MASLD. Here, we review the immune cell types and bidirectional pathways that influence the pathogenesis of MASLD and highlight potential pharmacologic approaches to combat MASLD based on current knowledge of this bidirectional crosstalk.
Collapse
Affiliation(s)
- Keisuke Sawada
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - Hak Chung
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Samir Softic
- Department of Pediatrics and Gastroenterology, University of Kentucky, Lexington, KY 40536, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Maria E Moreno-Fernandez
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
4
|
Sola-Leyva A, Pérez-Prieto I, Molina NM, Vargas E, Ruiz-Durán S, Leonés-Baños I, Canha-Gouveia A, Altmäe S. Microbial composition across body sites in polycystic ovary syndrome: a systematic review and meta-analysis. Reprod Biomed Online 2023; 47:129-150. [PMID: 37208218 DOI: 10.1016/j.rbmo.2023.03.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 04/03/2023]
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine disorder affecting reproductive-aged women, but the cause remains unclear. Recent evidence has linked microbial composition with PCOS; however, the results are inconsistent. The aim of this systematic review was to gather current knowledge of the microbes across body sites (oral cavity, blood, vagina/cervix, gut) in women with PCOS, and meta-analyse the microbial diversity in PCOS. For this purpose, a systematic search using PubMed, Web of Science, Cochrane and Scopus was carried out. After selection, 34 studies met the inclusion criteria. Most of the studies associated changes in the microbiome with PCOS, whereas heterogeneity of the studies in terms of ethnicity, body mass index (BMI) and methodology, among other confounders, made it difficult to corroborate this relationship. In fact, 19 out of 34 of the studies were categorised as having high risk of bias when the quality assessment was conducted. Our meta-analysis on the gut microbiome of 14 studies demonstrated that women with PCOS possess significantly lower microbial alpha diversity compared with controls (SMD = -0.204; 95% CI -0.360 to -0.048; P = 0.010; I2 = 5.508, by Shannon Index), which may contribute to the development of PCOS. Nevertheless, future studies should specifically overcome the shortcomings of the current studies by through well planned and conducted studies with larger sample sizes, proper negative and positive controls and adequate case-control matching.
Collapse
Affiliation(s)
- Alberto Sola-Leyva
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Inmaculada Pérez-Prieto
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Nerea M Molina
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Eva Vargas
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain; Systems Biology Unit, Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaén, Jaén, Spain
| | - Susana Ruiz-Durán
- Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain; UGC Obstetricia y Ginecología. HU Virgen de las Nieves, Granada, Spain
| | - Irene Leonés-Baños
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain
| | - Analuce Canha-Gouveia
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain; Department of Physiology, Faculty of Veterinary, University of Murcia, Campus Mare Nostrum, IMIB-Arrixaca, 30100 Murcia, Spain
| | - Signe Altmäe
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, Granada, Spain; Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
5
|
Liu J, Liu Y, Li X. Effects of intestinal flora on polycystic ovary syndrome. Front Endocrinol (Lausanne) 2023; 14:1151723. [PMID: 36967808 PMCID: PMC10036050 DOI: 10.3389/fendo.2023.1151723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 02/20/2023] [Indexed: 03/11/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine disorder in women of reproductive age. Its clinical characteristics are mainly oligo-ovulation or anovulation, hyperandrogenemia (HA) and insulin resistance (IR). PCOS is considered to be one of the main causes of infertility in women of childbearing age, and its pathogenesis is still unclear. Intestinal flora, known as the "second genome" of human beings, is closely related to metabolic diseases, immune diseases and infectious diseases. At the same time, mounting evidence suggests that intestinal flora can regulate insulin synthesis and secretion, affect androgen metabolism and follicular development, and is involved in the occurrence of chronic inflammation and obesity. The imbalance of intestinal flora is caused by the abnormal interaction between intestinal flora and host cells caused by the change of intestinal microbial diversity, which is related to the occurrence and development of PCOS. The adjustment of intestinal flora may be a potential direction for the treatment of PCOS.
Collapse
Affiliation(s)
- Jiayue Liu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, Haikou Key Laboratory of Li Nationality Medicine, School of Pharmacy, Hainan Medical University, Haikou, China
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Cardiovascular Diseases Institute of the First Affiliated Hospital, Hainan Medical University, Haikou, China
- College of Pharmacy, Heilongjiang Provincial Key Laboratory of New Drug Development and Pharmacotoxicological Evaluation, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Ying Liu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, Haikou Key Laboratory of Li Nationality Medicine, School of Pharmacy, Hainan Medical University, Haikou, China
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Cardiovascular Diseases Institute of the First Affiliated Hospital, Hainan Medical University, Haikou, China
- College of Pharmacy, Heilongjiang Provincial Key Laboratory of New Drug Development and Pharmacotoxicological Evaluation, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Xiaoliang Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, Haikou Key Laboratory of Li Nationality Medicine, School of Pharmacy, Hainan Medical University, Haikou, China
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Cardiovascular Diseases Institute of the First Affiliated Hospital, Hainan Medical University, Haikou, China
- College of Pharmacy, Heilongjiang Provincial Key Laboratory of New Drug Development and Pharmacotoxicological Evaluation, Jiamusi University, Jiamusi, Heilongjiang, China
- *Correspondence: Xiaoliang Li,
| |
Collapse
|
6
|
Bai X, Ma J, Wu X, Qiu L, Huang R, Zhang H, Huang H, Chen X. Impact of Visceral Obesity on Structural and Functional Alterations of Gut Microbiota in Polycystic Ovary Syndrome (PCOS): A Pilot Study Using Metagenomic Analysis. Diabetes Metab Syndr Obes 2023; 16:1-14. [PMID: 36760592 PMCID: PMC9843473 DOI: 10.2147/dmso.s388067] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/26/2022] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVE We aimed to identify structural and functional alterations of gut microbiota associated with visceral obesity in adult women with polycystic ovary syndrome (PCOS). METHODS Twenty-seven adults with PCOS underwent stool and fasting blood collection, oral glucose tolerance testing, and visceral fat area (VFA) measurement via dual-bioimpedance technique. Metagenomic analysis was used to analyze gut microbiota. RESULTS PCOS patients were divided into three groups: visceral obesity group (PCOS-VO, n=9, age 28.33±5.68 years, BMI 37.06±4.27 kg/m2, VFA 128.67±22.45 cm2), non-visceral obesity group (PCOS-NVO, n=10, age 25.40±4.53, BMI 30.74±3.95, VFA 52.00±24.04), normal BMI group (PCOS-NB, n=8, age 27.88±2.53, BMI 21.56±2.20, VFA 27.00±21.18), with no statistical difference in age (P>0.05) and significantly statistical differences in BMI and VFA (P<0.05). The groups showed a significant difference in microbial β-diversity between PCOS-VO and PCOS-NVO (P=0.002) and no difference between PCOS-NVO and PCOS-NB (P=0.177). Bacteroidetes was the phylum with the highest relative abundance among all patients, followed by Firmicutes. Those with visceral obesity had a higher abundance of Prevotella, Megamonas, and Dialister genera, positively correlated with metabolic markers (r>0.4, P<0.05), and lower abundance of Phascolarctobacterium and Neisseria genera, negatively correlated with metabolic markers (r<-0.4, P<0.05). Functional annotation analysis showed significant differences in relative abundance of ribosome pathway, fatty acid biosynthesis pathway, and sphingolipid signaling pathway between groups, affecting lipid homeostasis and visceral fat accumulation. CONCLUSION Alteration in β-diversity of gut microbiota exists in PCOS with visceral obesity versus those without visceral obesity and relates to functional differences in ribosomes, fatty acid biosynthesis, and sphingolipid signaling pathways.
Collapse
Affiliation(s)
- Xuefeng Bai
- Department of Endocrinology, Second Affiliated Hospital of Fujian Medical University, Quanzhou City, Fujian Province, People’s Republic of China
| | - Jiangxin Ma
- Department of Endocrinology, Second Affiliated Hospital of Fujian Medical University, Quanzhou City, Fujian Province, People’s Republic of China
| | - Xiaohong Wu
- Department of Endocrinology, Second Affiliated Hospital of Fujian Medical University, Quanzhou City, Fujian Province, People’s Republic of China
| | - Lingling Qiu
- Department of Reproductive Medicine, Second Affiliated Hospital of Fujian Medical University, Quanzhou City, Fujian Province, People’s Republic of China
| | - Rongfu Huang
- Department of Clinical Laboratory, Second Affiliated Hospital of Fujian Medical University, Quanzhou City, Fujian Province, People’s Republic of China
| | - Haibin Zhang
- Department of Endocrinology, Second Affiliated Hospital of Fujian Medical University, Quanzhou City, Fujian Province, People’s Republic of China
| | - Huibin Huang
- Department of Endocrinology, Second Affiliated Hospital of Fujian Medical University, Quanzhou City, Fujian Province, People’s Republic of China
- Correspondence: Huibin Huang; Xiaoyu Chen, Department of Endocrinology, the Second Affiliated Hospital of Fujian Medical University, No. 950 Donghai Street, Fengze District, Quanzhou City, Fujian Province, 362000, People’s Republic of China, Tel +86-13313872001; +86-13600739755, Email ;
| | - Xiaoyu Chen
- Department of Endocrinology, Second Affiliated Hospital of Fujian Medical University, Quanzhou City, Fujian Province, People’s Republic of China
| |
Collapse
|
7
|
Suturina L, Belkova N, Igumnov I, Lazareva L, Danusevich I, Nadeliaeva I, Sholokhov L, Rashidova M, Belenkaya L, Belskikh A, Sharifulin E, Ievleva K, Babaeva N, Egorova I, Salimova M, Kuzmin M, Tiumentseva D, Klimenko E, Sidorova T, Atalyan A. Polycystic Ovary Syndrome and Gut Microbiota: Phenotype Matters. Life (Basel) 2022; 13:7. [PMID: 36675956 PMCID: PMC9861125 DOI: 10.3390/life13010007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/05/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Abnormalities in gut microbiota diversity are considered important mechanisms in metabolic disorders in polycystic ovarian syndrome (PCOS). However, the data on the association of these disorders with the PCOS phenotype remain controversial. The objectives of this study were to estimate the alpha diversity of the gut microbiota of healthy women and PCOS patients depending on phenotype. The study participants (184 premenopausal women: 63 with PCOS, 121 without PCOS) were recruited during the annual employment assessment in the Irkutsk Region and the Buryat Republic (Russia) in 2016-2019. For PCOS diagnosis, we used the Rotterdam (2003) criteria and definitions of PCOS phenotypes. Five indexes of alpha diversity (ASV, Shannon, Simpson, Chao, and ACE) were estimated for the gut microbiota in all participants using amplicon metasequencing. As a result, two out of five alpha diversity indexes showed a statistical difference between the non-PCOS and PCOS groups. We did not find a significant difference in the alpha diversity of gut microbiota in the subgroups of women with hyperandrogenic PCOS phenotypes vs non-androgenic phenotype D and the group of women with the presence of only one of the PCOS criteria. Nevertheless, "classic" PCOS phenotypes demonstrated the most significant decrease in alpha diversity compared with healthy women without any signs of PCOS.
Collapse
|
8
|
Al-Bulish MSM, Cao W, Yang R, Wang Y, Xue C, Tang Q. Docosahexaenoic acid-rich fish oil alleviates hepatic steatosis in association with regulation of gut microbiome in ob/ob mice. Food Res Int 2022; 157:111373. [PMID: 35761631 DOI: 10.1016/j.foodres.2022.111373] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 05/04/2022] [Accepted: 05/10/2022] [Indexed: 11/04/2022]
Abstract
It remains to study whether docosahexaenoic acid-rich fish oil (DHA-FO) improves hepatic lipid metabolism by leptin-independent mechanisms. We used ob/ob mice as a model to investigate the effects of DHA-FO on hepatic steatosis. DHA-FO inhibited lipid droplets (LD) formation in liver of ob/ob mice. Probably because DHA-FO consumption prevented the accumulation of oleic acid, and suppressed the synthesis of triglycerides and cholesteryl esters. These beneficial effects might be concerned with the promotion of short chain fatty acids (SCFAs) production. Furthermore, DHA-FO could reverse gut bacteria dysbiosis, including increasing the abundance of SCFAs producers (e.g. Akkermansia and unclassified_Muribaculaceae), and suppressing the proliferation of conditional pathogenic bacteria, such as unclassified_Lachnospiraceae. DHA-FO also promoted colonic microbial function ("Glycerolipid metabolism") associated with lipid metabolism. As a potential ingredient for functional food, DHA-FO reduced LD accumulation, which might be associated with modulation of obesity-linked gut microbiome in ob/ob mice.
Collapse
Affiliation(s)
| | - Wanxiu Cao
- Marine Biomedical Research Institute of Qingdao, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Ruili Yang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - Yuming Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China; Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Qingjuan Tang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266003, China.
| |
Collapse
|
9
|
Tayachew B, Vanden Brink H, Garcia-Reyes Y, Rahat H, D'Alessandro A, Frank DN, Robertson CE, Silveira L, Kelsey M, Pyle L, Cree-Green M. Combined Oral Contraceptive Treatment Does Not Alter the Gut Microbiome but Affects Amino Acid Metabolism in Sera of Obese Girls With Polycystic Ovary Syndrome. Front Physiol 2022; 13:887077. [PMID: 35800349 PMCID: PMC9255376 DOI: 10.3389/fphys.2022.887077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/16/2022] [Indexed: 12/30/2022] Open
Abstract
Background: The gut microbiome is altered in obese adolescents with polycystic ovary syndrome (PCOS), and is associated with free testosterone, metabolic markers, and insulin resistance. Combined oral contraceptives (OCP) are a primary treatment for PCOS and decrease testosterone, but the effect on the serum metabolome or gut microbiome in obese adolescents with PCOS is unknown. Objective: Contrast gut microbiome profiles, targeted serum metabolomics, hormone levels, and metabolic measures in adolescents with PCOS and obesity with and without OCP treatment. Methods: Adolescent girls with obesity and PCOS underwent stool and fasting blood collection and MRI for hepatic fat fraction. Fecal bacteria were profiled by high-throughput 16S rRNA gene sequencing and fasting serum metabolomics performed with high resolution mass spectrometry. Groups were contrasted using t-tests and principle least squares discrimination analysis (PLS-DA). Associations between bacterial taxa, baseline metabolic measures, hormone levels and the metabolome were conducted using Spearman analysis. Analyses were adjusted for false discovery rate. Results: 29 adolescents with obesity [Untreated N = 21, 16 ± 1.2 years, BMI%ile 36.5 ± 3.0; OCP N = 8, 15.5 ± 0.9 years, BMI%ile 32.5 ± 3.9] participated. Of the untreated adolescents, N = 14 contributed serum for metabolomic analysis. Participants on OCP therapy had lower free testosterone and free androgen index (p < 0.001) and higher sex hormone binding globulin. There was no difference in measures of fasting glucose, insulin, lipids or HOMA-IR between groups. PLS-DA of serum metabolomics showed discrimination between the groups, secondary amino acid changes. Untreated and OCP had similar stool microbiome α-diversity based on evenness (p = 0.28), richness (p = 0.39), and Shannon diversity (p = 0.24) and global microbial composition (β-diversity, p = 0.56). There were no differences in % relative abundance at any level. Bacterial α-diversity was negatively associated with serum long chain fatty acids and branched chain amino acids. A higher %relative abundance of family Ruminococcaceae was significantly associated with serum bile acids and HOMA-IR. Conclusion: Despite hormone and serum amino acid differences, girls treated with OCP had similar metabolic and gut microbiome profiles compared to the untreated PCOS group. The association between bacterial α-diversity, Ruminococcaceae, clinical markers and long chain fatty acids suggests a potential role of the gut microbiome in the pathogenesis of the metabolic comorbidities in PCOS.
Collapse
Affiliation(s)
- Beza Tayachew
- Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States,Department of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Heidi Vanden Brink
- Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States,Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Yesenia Garcia-Reyes
- Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Haseeb Rahat
- Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Angelo D'Alessandro
- Department of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Daniel N. Frank
- Department of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Charles E. Robertson
- Department of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Lori Silveira
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States,Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, United States
| | - Megan Kelsey
- Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States,Center for Women’s Health Research, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Laura Pyle
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States,Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, United States
| | - Melanie Cree-Green
- Division of Pediatric Endocrinology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States,Center for Women’s Health Research, University of Colorado Anschutz Medical Campus, Aurora, CO, United States,*Correspondence: Melanie Cree-Green,
| |
Collapse
|
10
|
Doycheva I, Ehrmann DA. Nonalcoholic fatty liver disease and obstructive sleep apnea in women with polycystic ovary syndrome. Fertil Steril 2022; 117:897-911. [PMID: 35512974 DOI: 10.1016/j.fertnstert.2022.03.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 12/12/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) and obstructive sleep apnea are frequently associated with polycystic ovary syndrome (PCOS) but remain underrecognized. Women with PCOS have a 2-4 times higher risk of NAFLD independent of body mass index than healthy weight-matched controls. Insulin resistance and hyperandrogenemia together play a central role in the pathogenesis of NAFLD. Timely diagnosis of NAFLD is important because its progression can lead to nonalcoholic steatohepatitis and/or advanced liver fibrosis that can eventually result in liver-related mortality. The presence of NAFLD has also been associated with increased risks of type 2 diabetes, cardiovascular events, overall mortality, and extrahepatic cancers. The treatment of NAFLD in PCOS should include lifestyle interventions. Glucagon-like peptide 1 receptor agonists have shown promising results in patients with PCOS and NAFLD, but future randomized trails are needed to confirm this benefit. Likewise, the use of combined oral estrogen-progestin contraceptives may provide a benefit by decreasing hyperandrogenemia. Sleep disordered breathing is common among women with PCOS and is responsible for a number of cardiometabolic derangements. Obstructive sleep apnea is most often found in overweight and obese women with PCOS, but as is the case with NAFLD, its prevalence exceeds that of women who are of similar weight without PCOS. Left untreated, obstructive sleep apnea can precipitate or exacerbate insulin resistance, glucose intolerance, and hypertension.
Collapse
Affiliation(s)
- Iliana Doycheva
- Section of Endocrinology, Diabetes, and Metabolism, University of Chicago, Chicago, Illinois
| | - David A Ehrmann
- Section of Endocrinology, Diabetes, and Metabolism, University of Chicago, Chicago, Illinois.
| |
Collapse
|
11
|
Li H, Liu NN, Li JR, Dong B, Wang MX, Tan JL, Wang XK, Jiang J, Lei L, Li HY, Sun H, Jiang JD, Peng ZG. Combined Use of Bicyclol and Berberine Alleviates Mouse Nonalcoholic Fatty Liver Disease. Front Pharmacol 2022; 13:843872. [PMID: 35250593 PMCID: PMC8889073 DOI: 10.3389/fphar.2022.843872] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 01/12/2022] [Indexed: 11/20/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), ranging from simple steatosis to nonalcoholic steatohepatitis (NASH), is a liver disease worldwide without approved therapeutic drugs. Anti-inflammatory and hepatoprotective drug bicyclol and multi-pharmacological active drug berberine, respectively, have shown beneficial effects on NAFLD in murine nutritional models and patients, though the therapeutic mechanisms remain to be illustrated. Here, we investigated the combined effects of bicyclol and berberine on mouse steatosis induced by Western diet (WD), and NASH induced by WD/CCl4. The combined use of these was rather safe and better reduced the levels of transaminase in serum and triglycerides and cholesterol in the liver than their respective monotherapy, accompanied with more significantly attenuating hepatic inflammation, steatosis, and ballooning in mice with steatosis and NASH. The combined therapy also significantly inhibited fibrogenesis, characterized by the decreased hepatic collagen deposition and fibrotic surface. As per mechanism, bicyclol enhanced lipolysis and β-oxidation through restoring the p62-Nrf2-CES2 signaling axis and p62-Nrf2-PPARα signaling axis, respectively, while berberine suppressed de novo lipogenesis through downregulating the expression of acetyl-CoA carboxylase and fatty acid synthetase, along with enrichment of lipid metabolism-related Bacteroidaceae (family) and Bacteroides (genus). Of note, the combined use of bicyclol and berberine did not influence each other but enhanced the overall therapeutic role in the amelioration of NAFLD. Conclusion: Combined use of bicyclol and berberine might be a new available strategy to treat NAFLD.
Collapse
Affiliation(s)
- Hu Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan-Nan Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian-Rui Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Biao Dong
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mei-Xi Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jia-Li Tan
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xue-Kai Wang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Jiang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Lei
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hong-Ying Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Han Sun
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian-Dong Jiang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zong-Gen Peng
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Key Laboratory of Biotechnology of Antibiotics, The National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
12
|
Hassan S, Kaakinen MA, Draisma H, Zudina L, Ganie MA, Rashid A, Balkhiyarova Z, Kiran GS, Vogazianos P, Shammas C, Selvin J, Antoniades A, Demirkan A, Prokopenko I. Bifidobacterium Is Enriched in Gut Microbiome of Kashmiri Women with Polycystic Ovary Syndrome. Genes (Basel) 2022; 13:379. [PMID: 35205422 PMCID: PMC8871983 DOI: 10.3390/genes13020379] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/08/2022] [Accepted: 02/15/2022] [Indexed: 12/12/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a very common endocrine condition in women in India. Gut microbiome alterations were shown to be involved in PCOS, yet it is remarkably understudied in Indian women who have a higher incidence of PCOS as compared to other ethnic populations. During the regional PCOS screening program among young women, we recruited 19 drug naive women with PCOS and 20 control women at the Sher-i-Kashmir Institute of Medical Sciences, Kashmir, North India. We profiled the gut microbiome in faecal samples by 16S rRNA sequencing and included 40/58 operational taxonomic units (OTUs) detected in at least 1/3 of the subjects with relative abundance (RA) ≥ 0.1%. We compared the RAs at a family/genus level in PCOS/non-PCOS groups and their correlation with 33 metabolic and hormonal factors, and corrected for multiple testing, while taking the variation in day of menstrual cycle at sample collection, age and BMI into account. Five genera were significantly enriched in PCOS cases: Sarcina, Megasphaera, and previously reported for PCOS Bifidobacterium, Collinsella and Paraprevotella confirmed by different statistical models. At the family level, the relative abundance of Bifidobacteriaceae was enriched, whereas Peptococcaceae was decreased among cases. We observed increased relative abundance of Collinsella and Paraprevotella with higher fasting blood glucose levels, and Paraprevotella and Alkalibacterium with larger hip, waist circumference, weight, and Peptococcaceae with lower prolactin levels. We also detected a novel association between Eubacterium and follicle-stimulating hormone levels and between Bifidobacterium and alkaline phosphatase, independently of the BMI of the participants. Our report supports that there is a relationship between gut microbiome composition and PCOS with links to specific reproductive health metabolic and hormonal predictors in Indian women.
Collapse
Affiliation(s)
- Saqib Hassan
- Section of Genetics and Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK; (S.H.); (M.A.K.); (H.D.); (Z.B.)
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India;
| | - Marika A. Kaakinen
- Section of Genetics and Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK; (S.H.); (M.A.K.); (H.D.); (Z.B.)
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, University of Surrey, Guildford GU2 7XH, UK; (L.Z.); (A.D.)
| | - Harmen Draisma
- Section of Genetics and Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK; (S.H.); (M.A.K.); (H.D.); (Z.B.)
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, University of Surrey, Guildford GU2 7XH, UK; (L.Z.); (A.D.)
| | - Liudmila Zudina
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, University of Surrey, Guildford GU2 7XH, UK; (L.Z.); (A.D.)
| | - Mohd A. Ganie
- Department of Endocrinology, Sheri-Kashmir Institute of Medical Sciences (SKIMS), Srinagar 190011, India; (M.A.G.); (A.R.)
| | - Aafia Rashid
- Department of Endocrinology, Sheri-Kashmir Institute of Medical Sciences (SKIMS), Srinagar 190011, India; (M.A.G.); (A.R.)
| | - Zhanna Balkhiyarova
- Section of Genetics and Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK; (S.H.); (M.A.K.); (H.D.); (Z.B.)
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, University of Surrey, Guildford GU2 7XH, UK; (L.Z.); (A.D.)
| | - George S. Kiran
- Department of Food Science and Technology, School of Life Sciences, Pondicherry University, Puducherry 605014, India;
| | | | | | - Joseph Selvin
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India;
| | | | - Ayse Demirkan
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, University of Surrey, Guildford GU2 7XH, UK; (L.Z.); (A.D.)
- Department of Genetics, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands
| | - Inga Prokopenko
- Section of Genetics and Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London SW7 2AZ, UK; (S.H.); (M.A.K.); (H.D.); (Z.B.)
- Section of Statistical Multi-Omics, Department of Clinical and Experimental Medicine, University of Surrey, Guildford GU2 7XH, UK; (L.Z.); (A.D.)
- Laboratory UMR 8199-EGID, Institut Pasteur de Lille, CNRS, University of Lille, F-59000 Lille, France
| |
Collapse
|
13
|
Falzarano C, Lofton T, Osei-Ntansah A, Oliver T, Southward T, Stewart S, Andrisse S. Nonalcoholic Fatty Liver Disease in Women and Girls With Polycystic Ovary Syndrome. J Clin Endocrinol Metab 2022; 107:258-272. [PMID: 34491336 DOI: 10.1210/clinem/dgab658] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Indexed: 12/15/2022]
Abstract
CONTEXT Nonalcoholic fatty liver disease (NAFLD) describes a spectrum of liver damage due to excessive hepatic lipid accumulation. Recent research has demonstrated a high prevalence of NAFLD in women with polycystic ovary syndrome (PCOS). RESULTS Strong associations independent of body mass index (BMI) have been found between high androgen levels characteristic of PCOS, as well as insulin resistance, and the presence of NAFLD in these women, suggesting that these factors contribute to liver injury more significantly than obesity. Current studies indicate the occurrence of NAFLD in normal weight women with PCOS in addition to the commonly researched women who are overweight and obese. While the majority of studies address NAFLD in adult, premenopausal women (ages 25-40 years), the occurrence of NAFLD in young and adolescent women has gone largely unaddressed. Research in this field lacks diversity; a majority of studies either focus on populations of White women or are missing demographic information entirely. CONCLUSIONS Future studies should include larger, more racially and ethnically inclusive populations and particular attention should be paid to how excess androgens and insulin resistance contribute to the increased risk of NAFLD seen in women with PCOS of varying weights, ages, and ethnicities. OBJECTIVE AND METHODS Here, we review NAFLD in women with PCOS with subsections focused on the impact of hyperandrogenism, BMI, insulin resistance and age. Most notably, we present the most up-to-date racially and ethnically diverse worldwide prevalence of NAFLD in women with PCOS compared with women without PCOS (51.56% vs 29.64%, P < .001, respectively).
Collapse
Affiliation(s)
- Claire Falzarano
- Howard University College of Medicine, Physiology and Biophysics, Washington, DC, 20059, USA
| | - Taylor Lofton
- Howard University College of Medicine, Physiology and Biophysics, Washington, DC, 20059, USA
| | - Adjoa Osei-Ntansah
- Howard University College of Medicine, Physiology and Biophysics, Washington, DC, 20059, USA
| | - Trinitee Oliver
- Howard University College of Medicine, Physiology and Biophysics, Washington, DC, 20059, USA
| | - Taylor Southward
- Howard University College of Medicine, Physiology and Biophysics, Washington, DC, 20059, USA
| | - Salim Stewart
- Howard University College of Medicine, Physiology and Biophysics, Washington, DC, 20059, USA
| | - Stanley Andrisse
- Howard University College of Medicine, Physiology and Biophysics, Washington, DC, 20059, USA
| |
Collapse
|
14
|
Wang L, Zhang P, Li C, Xu F, Chen J. A polysaccharide from Rosa roxburghii Tratt fruit attenuates high-fat diet-induced intestinal barrier dysfunction and inflammation in mice by modulating the gut microbiota. Food Funct 2021; 13:530-547. [PMID: 34932054 DOI: 10.1039/d1fo03190b] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Obesity-induced colonic inflammation-stimulated colitis is one of the main causes of colorectal cancer. Dietary polysaccharides are considered an effective agent for relieving obesity-induced inflammatory diseases such as diabetes and colitis. In this work, the protective effects of a polysaccharide (RTFP) extracted from Rosa roxburghii Tratt fruit on barrier dysfunction and inflammation were investigated using obesity-induced colitis model mice. RTFP treatment repaired intestinal barrier dysfunction by increasing the expression of tight junction proteins (ZO-1, claudin-1, and occludin) and reducing the levels of inflammatory cytokines, intestinal permeability, and colonic oxidative stress in mice fed a high-fat diet. Most significantly, RTFP decreased gut inflammation and ameliorated the metabolic dysbiosis of intestinal microflora by decreasing the Firmicutes/Bacteroidetes ratio, reducing the levels of serum D-lactic acid and lipopolysaccharides, and inhibiting the TLR4/NF-κB signaling pathway. Furthermore, RTFP significantly increased the abundance of beneficial bacteria (Ruminococcaceae, Muribaculaceae, Akkermansiaceae, etc.) but decreased the abundance of pathogenic bacteria. These findings indicate that RTFP can be used as a natural anti-inflammatory agent to reduce chronic obesity-induced colitis.
Collapse
Affiliation(s)
- Lei Wang
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou, 450001, China.
| | - Pan Zhang
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou, 450001, China.
| | - Chao Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Fei Xu
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou, 450001, China.
| | - Jie Chen
- College of Food Science and Engineering, Henan University of Technology, Zhengzhou, 450001, China.
| |
Collapse
|