1
|
Yilmaz EG, Hacıosmanoğlu N, Jordi SBU, Yilmaz B, Inci F. Revolutionizing IBD research with on-chip models of disease modeling and drug screening. Trends Biotechnol 2025; 43:1062-1078. [PMID: 39523166 DOI: 10.1016/j.tibtech.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/30/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024]
Abstract
Inflammatory bowel disease (IBD) comprises chronic inflammatory conditions with complex mechanisms and diverse manifestations, posing significant clinical challenges. Traditional animal models and ethical concerns in human studies necessitate innovative approaches. This review provides an overview of human intestinal architecture in health and inflammation, emphasizing the role of microfluidics and on-chip technologies in IBD research. These technologies allow precise manipulation of cellular and microbial interactions in a physiologically relevant context, simulating the intestinal ecosystem microscopically. By integrating cellular components and replicating 3D tissue architecture, they offer promising models for studying host-microbe interactions, wound healing, and therapeutic approaches. Continuous refinement of these technologies promises to advance IBD understanding and therapy development, inspiring further innovation and cross-disciplinary collaboration.
Collapse
Affiliation(s)
- Eylul Gulsen Yilmaz
- UNAM-National Nanotechnology Research Center, Bilkent University, 06800 Ankara, Turkey; Institute of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey
| | - Nedim Hacıosmanoğlu
- UNAM-National Nanotechnology Research Center, Bilkent University, 06800 Ankara, Turkey; Institute of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey
| | - Sebastian Bruno Ulrich Jordi
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, 3010, Bern, Switzerland; Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, 3008, Bern, Switzerland
| | - Bahtiyar Yilmaz
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, 3010, Bern, Switzerland; Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, 3008, Bern, Switzerland.
| | - Fatih Inci
- UNAM-National Nanotechnology Research Center, Bilkent University, 06800 Ankara, Turkey; Institute of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey.
| |
Collapse
|
2
|
Kennedy MS, Freiburger A, Cooper M, Beilsmith K, St George ML, Kalski M, Cham C, Guzzetta A, Ng SC, Chan FK, DeLeon O, Rubin D, Henry CS, Bergelson J, Chang EB. Diet outperforms microbial transplant to drive microbiome recovery in mice. Nature 2025:10.1038/s41586-025-08937-9. [PMID: 40307551 DOI: 10.1038/s41586-025-08937-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 03/25/2025] [Indexed: 05/02/2025]
Abstract
A high-fat, low-fibre Western-style diet (WD) induces microbiome dysbiosis characterized by reduced taxonomic diversity and metabolic breadth1,2, which in turn increases risk for a wide array of metabolic3-5, immune6 and systemic pathologies. Recent work has established that WD can impair microbiome resilience to acute perturbations such as antibiotic treatment7,8, although little is known about the mechanism of impairment and the specific consequences for the host of prolonged post-antibiotic dysbiosis. Here we characterize the trajectory by which the gut microbiome recovers its taxonomic and functional profile after antibiotic treatment in mice on regular chow (RC) or WD, and find that only mice on RC undergo a rapid successional process of recovery. Metabolic modelling indicates that a RC diet promotes the development of syntrophic cross-feeding interactions, whereas in mice on WD, a dominant taxon monopolizes readily available resources without releasing syntrophic byproducts. Intervention experiments reveal that an appropriate dietary resource environment is both necessary and sufficient for rapid and robust microbiome recovery, whereas microbial transplant is neither. Furthermore, prolonged post-antibiotic dysbiosis in mice on WD renders them susceptible to infection by the intestinal pathogen Salmonella enterica serovar Typhimurium. Our data challenge widespread enthusiasm for faecal microbiota transplant (FMT) as a strategy to address dysbiosis, and demonstrate that specific dietary interventions are, at a minimum, an essential prerequisite for effective FMT, and may afford a safer, more natural and less invasive alternative.
Collapse
Affiliation(s)
- M S Kennedy
- Medical Scientist Training Program, Pritzker School of Medicine, The University of Chicago, Chicago, IL, USA
- Department of Ecology & Evolution, The University of Chicago, Chicago, IL, USA
| | - A Freiburger
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, USA
- Department of Chemical Engineering, Northwestern University, Evanston, IL, USA
| | - M Cooper
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - K Beilsmith
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, USA
| | - M L St George
- Department of Medicine, The University of Chicago, Chicago, IL, USA
- Medical Scientist Training Program, University of Illinois Chicago, Chicago, IL, USA
| | - M Kalski
- Department of Medicine, The University of Chicago, Chicago, IL, USA
- Stritch School of Medicine, Loyola University Chicago, Chicago, IL, USA
| | - C Cham
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - A Guzzetta
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - S C Ng
- Microbiota I-Center (MagIC), Department of Medicine and Therapeutics, LKS Institute of Health Science, Institute of Digestive Disease, Center for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China
- New Cornerstone Science Laboratory, The Chinese University of Hong Kong, Hong Kong, China
| | - F K Chan
- Microbiota I-Center (MagIC), Department of Medicine and Therapeutics, LKS Institute of Health Science, Institute of Digestive Disease, Center for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - O DeLeon
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - D Rubin
- Inflammatory Bowel Disease Center, University of Chicago Medicine, Chicago, IL, USA
| | - C S Henry
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, USA
| | - J Bergelson
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - E B Chang
- Department of Medicine, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
3
|
Zhang ZM, Liu H, Zuo HL, Wang YN, Sun AL, Chen J, Shi XZ. Unraveling the toxic trio: Combined effects of thifluzamide, enrofloxacin, and microplastics on Mytilus coruscus. JOURNAL OF HAZARDOUS MATERIALS 2025; 494:138441. [PMID: 40311431 DOI: 10.1016/j.jhazmat.2025.138441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 04/26/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
The presence of pesticides, antibiotics, and microplastics in aquatic environments poses a significant threat because of their persistence and potential harm to aquatic life and human health. However, few studies have explored their combined effects on bioaccumulation and toxicity in edible bivalves. This study examined the bioaccumulation and toxicological impacts of thifluzamide (TF) and enrofloxacin (ENR) on oxidative stress, neurotoxicity, detoxification, and metabolism in Mytilus coruscus after 4 weeks of exposure at the environmental level. The findings indicated that coexposure to TF and ENR or the presence of microplastic polystyrene (PS) increased TF and ENR accumulation in mussels and caused oxidative damage, as evidenced by elevated catalase and glutathione transferase activities and increased malondialdehyde (MDA) levels. Notably, compared with single exposures, coexposure to PS+TF, PS+ENR, or TF+ENR generally increased the MDA content, reduced acetylcholinesterase activity, and increased detoxification gene expression. Metabolomic analysis revealed that TF, ENR, and PS, either alone or combined, significantly disrupted multiple metabolic pathways by altering levels of glycerophospholipids, eicosanoids, amino acids, and nucleotides. Coexposure particularly worsened glycerophospholipid and arachidonic acid metabolism disturbances. These results suggest that combined exposure to TF, ENR or PS exacerbated the ecotoxicological effects of TF and ENR on M. coruscus. Taken together, the results of the present study could enhance our understanding of the environmental effects resulting from multipollutant interactions and their potential risks to seafood security.
Collapse
Affiliation(s)
- Ze-Ming Zhang
- State Key Laboratory for Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, PR China; Key Laboratory of Aquacultural Biotechnology (Ningbo University), Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo 315211, PR China
| | - Hao Liu
- State Key Laboratory for Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, PR China; Key Laboratory of Aquacultural Biotechnology (Ningbo University), Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo 315211, PR China
| | - Hong-Lin Zuo
- State Key Laboratory for Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, PR China
| | - Yi-Nan Wang
- State Key Laboratory for Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, PR China; Key Laboratory of Aquacultural Biotechnology (Ningbo University), Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo 315211, PR China
| | - Ai-Li Sun
- Key Laboratory of Aquacultural Biotechnology (Ningbo University), Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo 315211, PR China
| | - Jiong Chen
- State Key Laboratory for Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, PR China; Key Laboratory of Aquacultural Biotechnology (Ningbo University), Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo 315211, PR China
| | - Xi-Zhi Shi
- State Key Laboratory for Quality and Safety of Agro-products, School of Marine Sciences, Ningbo University, Ningbo 315211, PR China; Key Laboratory of Aquacultural Biotechnology (Ningbo University), Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo 315211, PR China.
| |
Collapse
|
4
|
Han X, Ma P, Liu C, Yao C, Yi Y, Du Z, Liu P, Zhang M, Xu J, Meng X, Liu Z, Wang W, Ren R, Xie L, Han X, Xiao K. Pathogenic profiles and lower respiratory tract microbiota in severe pneumonia patients using metagenomic next-generation sequencing. ADVANCED BIOTECHNOLOGY 2025; 3:13. [PMID: 40279015 PMCID: PMC12031718 DOI: 10.1007/s44307-025-00064-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 03/15/2025] [Accepted: 03/29/2025] [Indexed: 04/26/2025]
Abstract
INTRODUCTION The homeostatic balance of the lung microbiota is important for the maintenance of normal physiological function of the lung, but its role in pathological processes such as severe pneumonia is poorly understood. METHODS We screened 34 patients with community-acquired pneumonia (CAP) and 12 patients with hospital-acquired pneumonia (HAP), all of whom were admitted to the respiratory intensive care unit. Clinical samples, including bronchoalveolar lavage fluid (BALF), sputum, peripheral blood, and tissue specimens, were collected along with traditional microbiological test results, routine clinical test data, and clinical treatment information. The pathogenic spectrum of lower respiratory tract pathogens in critically ill respiratory patients was characterized through metagenomic next-generation sequencing (mNGS). Additionally, we analyzed the composition of the commensal microbiota and its correlation with clinical characteristics. RESULTS The sensitivity of the mNGS test for pathogens was 92.2% and the specificity 71.4% compared with the clinical diagnosis of the patients. Using mNGS, we detected more fungi and viruses in the lower respiratory tract of CAP-onset severe pneumonia patients, whereas bacterial species were predominant in HAP-onset patients. On the other hand, using mNGS data, commensal microorganisms such as Fusobacterium yohimbe were observed in the lower respiratory tract of patients with HAP rather than those with CAP, and most of these commensal microorganisms were associated with hospitalization or the staying time in ICU, and were significantly and positively correlated with the total length of stay. CONCLUSION mNGS can be used to effectively identify pathogenic pathogens or lower respiratory microbiome associated with pulmonary infectious diseases, playing a crucial role in the early and accurate diagnosis of these conditions. Based on the findings of this study, it is possible that a novel set of biomarkers and predictive models could be developed in the future to efficiently identify the cause and prognosis of patients with severe pneumonia.
Collapse
Affiliation(s)
- Xinjie Han
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, China
- Chinese PLA Medical School, Beijing, China
| | - Peng Ma
- MatriDx Biotechnology Co., Ltd, Hangzhou, China
| | - Chang Liu
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, China
- School of Medicine, Nankai University, Tianjin, China
| | - Chen Yao
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yaxing Yi
- MatriDx Biotechnology Co., Ltd, Hangzhou, China
| | - Zhenshan Du
- MatriDx Biotechnology Co., Ltd, Hangzhou, China
| | - Pengfei Liu
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Minlong Zhang
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jianqiao Xu
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiaoyun Meng
- Department of Urology, 8th Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zidan Liu
- MatriDx Biotechnology Co., Ltd, Hangzhou, China
| | - Weijia Wang
- MatriDx Biotechnology Co., Ltd, Hangzhou, China
| | - Ruotong Ren
- MatriDx Biotechnology Co., Ltd, Hangzhou, China
- Foshan Branch, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Lixin Xie
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, China.
| | - Xu Han
- MatriDx Biotechnology Co., Ltd, Hangzhou, China.
| | - Kun Xiao
- College of Pulmonary & Critical Care Medicine, 8th Medical Center of Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
5
|
Cheng X, Liang Y, Ji K, Feng M, Du X, Jiao D, Wu X, Zhong C, Cong H, Yang G. Enhanced propionate and butyrate metabolism in cecal microbiota contributes to cold-stress adaptation in sheep. MICROBIOME 2025; 13:103. [PMID: 40275300 PMCID: PMC12023611 DOI: 10.1186/s40168-025-02096-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/17/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND During cold stress, gut microbes play crucial roles in orchestrating energy metabolism to enhance environmental adaptation. In sheep, hindgut microbes ferment carbohydrates to generate short-chain fatty acids (SCFAs) as an energy source. However, the mechanisms by which hindgut microbes and their metabolites interact with the host to facilitate adaptation to cold environments remain ambiguous. Herein, we simulated a winter environment (- 20 °C) and provided a rationed diet to compare the cold adaptation mechanisms between Hulunbuir and Hu sheep. RESULTS Our findings show that cold exposure enhances SCFA metabolism in the sheep cecum. In Hu sheep, acetate, butyrate, and total SCFA concentrations increased, whereas in Hulunbuir sheep, propionate and butyrate concentrations increased, with a notable increase in total SCFAs. Notably, butyrate concentration was higher in Hulunbuir sheep than in Hu sheep under cold stress. Following cold exposure, the proinflammatory cytokine IL-1β levels increased in both breeds. In addition, Hu sheep showed increased IL-10, whereas Hulunbuir sheep exhibited elevated secretory IgA levels. The cecal microbiota responded differently, Hu sheep showed no notable changes in alpha and beta diversity, whereas Hulunbuir sheep exhibited considerable alterations. In Hu sheep, the abundance of fungi, specifically Blastocystis sp. subtype 4, decreased, and that of several Lachnospiraceae species (Roseburia hominis, Faecalicatena contorta, and Ruminococcus gnavus) involved in SCFA metabolism increased. Pathways related to carbohydrate metabolism, such as starch and sucrose metabolism, galactose metabolism, and pentose and glucuronate interconversions, were upregulated. In Hulunbuir sheep, the abundance of Treponema bryantii, Roseburia sp. 499, and Prevotella copri increased, with upregulation in pathways related to amino acid metabolism and energy metabolism. Cold exposure increased node connectivity within the symbiotic networks of both breeds, with increased network vulnerability in Hu sheep. Following cold exposure, the microbial community of Hulunbuir sheep showed a decrease in the influence of stochastic processes on community assembly, with a corresponding increase in the role of environmental selection. Conversely, no such shift was evident in Hu sheep. Further transcriptomic analysis revealed distinct regulatory mechanisms between breeds. In Hu sheep, protein synthesis, energy metabolism, and thermogenesis pathways were substantially upregulated. By contrast, Hulunbuir sheep showed considerable upregulation of immune pathways and energy conservation through reduced ribosome synthesis. Correlation analysis indicated that butyrate holds a central position in both networks, with Hulunbuir sheep exhibiting a more complex and tightly regulated network involving SCFAs, microbiota, microbial functions, and transcriptomes. Partial least squares path modeling revealed that cold exposure substantially altered the cecal microbiota and transcriptomes of Hulunbuir sheep, affecting SCFAs and cytokines. CONCLUSIONS The findings of this study suggest that under cold exposure, Hu sheep enhance acetate fermentation and rely on tissue thermogenesis for adaptation. By contrast, Hulunbuir sheep exhibit changes in microbial diversity and function, leading to increased propionate and butyrate metabolism. This may promote physiological energy conservation and innate immune defense, balancing heat loss and enhancing cold adaptation.
Collapse
Affiliation(s)
- Xindong Cheng
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanping Liang
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kaixi Ji
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute of Animal Husbandry and Veterinary Medicine, Shandong Academy of Agricultural Sciences/Shandong Key Laboratory of Animal Microecologics and Efficient Breeding of Livestock and Poultry, Jinan, 250100, China
| | - Mengyu Feng
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xia Du
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Dan Jiao
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiukun Wu
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Extreme Environmental Microbial Resources and Engineering, Lanzhou, 730000, China
| | - Chongyue Zhong
- Dongying Animal Husbandry and Veterinary Station, Dongying, 257000, China
| | - Haitao Cong
- Shandong Huakun Rural Revitalization Institute Co., Ltd, Jinan, 250014, China
| | - Guo Yang
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
6
|
Liu X, Guan B, Hu Z, Hu X, Liu S, Yang K, Zhou L, Yu L, Yang J, Chen S, Chen Q, Liu D, Liu G, Pan H. Combined traditional Chinese medicine and probiotics (TCMP) alleviates lipid accumulation and improves metabolism in high-fat diet mice via the microbiota-gut-liver axis. Food Res Int 2025; 207:116064. [PMID: 40086971 DOI: 10.1016/j.foodres.2025.116064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/09/2025] [Accepted: 02/22/2025] [Indexed: 03/16/2025]
Abstract
Lipid accumulation and metabolic disorders caused by a high-fat diet (HFD) pose significant threats to human health, and place a substantial burden on individuals and society. In this study, a novel combination comprising three traditional Chinese herbs (lotus leaf, hawthorn, and leaf of Chinese holly) and a probiotic (Bifidobacterium lactis BPL-1) (TCMP) was prepared. Then, its effects on growth performance, fat accumulation, hepatic function and gut microbiota in mice fed a high-fat diet were investigated. According to the results, TCMP significantly reduced adipose tissue fat accumulation, improved hepatic lipid metabolism, and ameliorated glucose homeostasis in HFD-fed mice. Notably, TCMP not only improved the abundance and diversity of gut microbiota and increased the content of beneficial intestinal bacteria related to lipid metabolism (especially Bifidobacterium animalis), but also increased the production of short-chain fatty acids, including2-methylbutyrate, isovaleric acid and isobutyric acid. Additionally, multi-omics (transcriptome and metabolome) analysis revealed that TCMP significantly inhibited the expression of genes involved in the lipid biosynthesis process and modulated the purine and glycerophospholipid metabolism caused by a high-fat diet, thereby achieving the purpose of reducing fat accumulation and regulating lipid metabolism. Taken together, our finding demonstrates the potential of TCMP as a promising therapeutic candidate for combatting obesity and lipid metabolism disorders induced by a high-fat diet.
Collapse
Affiliation(s)
- Xiayu Liu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| | - Boyuan Guan
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| | - Ziyi Hu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| | - Xiaoyan Hu
- Guangdong Qingyunshan Chinese Medicine Innovation Co., Ltd., Shaoguan 512000, China
| | - Shuaixing Liu
- Guangdong Qingyunshan Chinese Medicine Innovation Co., Ltd., Shaoguan 512000, China
| | - Ke Yang
- Guangdong Qingyunshan Chinese Medicine Innovation Co., Ltd., Shaoguan 512000, China
| | - Liping Zhou
- Guangdong Qingyunshan Chinese Medicine Innovation Co., Ltd., Shaoguan 512000, China
| | - Longli Yu
- Guangdong Qingyunshan Chinese Medicine Innovation Co., Ltd., Shaoguan 512000, China
| | - Jinyan Yang
- Guangdong Qingyunshan Chinese Medicine Innovation Co., Ltd., Shaoguan 512000, China
| | - Shiguo Chen
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| | - Qihe Chen
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| | - Donghong Liu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| | - Guanchen Liu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China.
| | - Haibo Pan
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China.
| |
Collapse
|
7
|
Zhao S, Cao H, Sun F, Xu M, Wang X, Jiang J, Luo L, Zeng L. Investigating the modulatory effects of Pu-erh tea on the gut microbiota in ameliorating hyperuricemia induced by circadian rhythm disruption. Food Funct 2025; 16:2669-2686. [PMID: 40029218 DOI: 10.1039/d4fo05659k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Circadian rhythm disruption (CRD) can induce a variety of metabolic disorders. Our previous laboratory studies have shown that Pu-erh tea could alleviate CRD-induced syndromes, including obesity, intestinal dysfunction, and tryptophan metabolism disorders. However, its potential protective mechanism against CRD-induced hyperuricaemia remains unclear. In this work, we found that polyphenols of Pu-erh tea were significantly released in the stage of intestinal digestion, which might promote their interaction with gut microbes. Through animal experiments, C57BL6/J mice were given water or different doses of Pu-erh tea for 60 days, followed by a 90-day CRD, the lifestyle of modern individuals who frequently stay up late. Our results indicated that CRD mice exhibited high serum uric acid levels and gut microbiota disorders. Pu-erh tea intake significantly reshaped the gut microbiome, especially increasing the abundance of Bifidobacterium, Akkermansia and Faecalibaculum, and increased the production of short-chain fatty acids (SCFAs), especially acetic acid, which restored the function of the intestinal barrier. This improvement further regulated oxidative stress pathways (NRF2/HO-1), reduced systemic inflammatory response (IL-6, IL-1β, and TNF-α), restored hepatic function (SOD, MOD, CAT, and GSH) and modulated the activity of enzymes related to UA metabolism in the liver (XOD and ADA). Finally, Pu-erh tea intake promoted the excretion of UA and reduced the levels of UA and xanthine in the serum. Moreover, the results of antibiotic experiments showed that the UA improvement effect of Pu-erh tea depended on the existence of the gut microbiota. Collectively, Pu-erh tea intake has the potential to prevent CRD-induced hyperuricaemia by reshaping the gut microbiota.
Collapse
Affiliation(s)
- Sibo Zhao
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, College of Food Science, Southwest University, Beibei, Chongqing, 400715, China.
- Chongqing Key Laboratory of Specialty Food Co-Built by Sichuan and Chongqing, Southwest University, Chongqing, 400715, China
| | - Hongli Cao
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, College of Food Science, Southwest University, Beibei, Chongqing, 400715, China.
- Chongqing Key Laboratory of Specialty Food Co-Built by Sichuan and Chongqing, Southwest University, Chongqing, 400715, China
| | - Fanwei Sun
- Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Mianhong Xu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, College of Food Science, Southwest University, Beibei, Chongqing, 400715, China.
- Chongqing Key Laboratory of Specialty Food Co-Built by Sichuan and Chongqing, Southwest University, Chongqing, 400715, China
| | - Xinghua Wang
- College of Tea, Yunnan Agricultural University, Puer, Yunnan, 665000, China
| | - Jielin Jiang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, College of Food Science, Southwest University, Beibei, Chongqing, 400715, China.
- Chongqing Key Laboratory of Specialty Food Co-Built by Sichuan and Chongqing, Southwest University, Chongqing, 400715, China
- Menghai Tea Factory·TAETEA Group, Xishuangbanna Dai Autonomous Prefecture, Yunnan 666200, China
| | - Liyong Luo
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, College of Food Science, Southwest University, Beibei, Chongqing, 400715, China.
- Chongqing Key Laboratory of Specialty Food Co-Built by Sichuan and Chongqing, Southwest University, Chongqing, 400715, China
| | - Liang Zeng
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, College of Food Science, Southwest University, Beibei, Chongqing, 400715, China.
- Chongqing Key Laboratory of Specialty Food Co-Built by Sichuan and Chongqing, Southwest University, Chongqing, 400715, China
| |
Collapse
|
8
|
Tisza MJ, Lloyd RE, Hoffman K, Smith DP, Rewers M, Javornik Cregeen SJ, Petrosino JF. Longitudinal phage-bacteria dynamics in the early life gut microbiome. Nat Microbiol 2025; 10:420-430. [PMID: 39856391 PMCID: PMC11790489 DOI: 10.1038/s41564-024-01906-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 12/04/2024] [Indexed: 01/27/2025]
Abstract
Microbial colonization of the human gut occurs soon after birth, proceeds through well-studied phases and is affected by lifestyle and other factors. Less is known about phage community dynamics during infant gut colonization due to small study sizes, an inability to leverage large databases and a lack of appropriate bioinformatics tools. Here we reanalysed whole microbial community shotgun sequencing data of 12,262 longitudinal samples from 887 children from four countries across four years of life as part of the The Environmental Determinants of Diabetes in the Young (TEDDY) study. We developed an extensive metagenome-assembled genome catalogue using the Marker-MAGu pipeline, which comprised 49,111 phage taxa from existing human microbiome datasets. This was used to identify phage marker genes and their integration into the MetaPhlAn 4 bacterial marker gene database enabled simultaneous assessment of phage and bacterial dynamics. We found that individual children are colonized by hundreds of different phages, which are more transitory than bacteria, accumulating a more diverse phage community over time. Type 1 diabetes correlated with a decreased rate of change in bacterial and viral communities in children aged one and two. The addition of phage data improved the ability of machine learning models to discriminate samples by country. Finally, although phage populations were specific to individuals, we observed trends of phage ecological succession that correlated well with putative host bacteria. This resource improves our understanding of phage-bacteria interactions in the developing early life microbiome.
Collapse
Affiliation(s)
- Michael J Tisza
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Richard E Lloyd
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Kristi Hoffman
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Daniel P Smith
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Marian Rewers
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, USA
| | - Sara J Javornik Cregeen
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA.
| | - Joseph F Petrosino
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
9
|
Xu X, Wang Y, Wu X, Cai T, Dong L, Liang S, Zhu L, Song X, Dong Y, Zheng Y, Li L, Sun W. Administration of Alistipes indistinctus prevented the progression from nonalcoholic fatty liver disease to nonalcoholic steatohepatitis by enhancing the gut barrier and increasing Lactobacillus spp. Biochem Biophys Res Commun 2024; 741:151033. [PMID: 39579531 DOI: 10.1016/j.bbrc.2024.151033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/05/2024] [Accepted: 11/19/2024] [Indexed: 11/25/2024]
Abstract
Metabolic-associated fatty liver disease (MAFLD) is an important public health problem, and the gut microbiota has become a new treatment target for MAFLD. Previously, A. indistinctus, a core gut bacterium, was shown to potentially contribute to the prevention of MAFLD. However, the effect and mechanism of A. indistinctus on MAFLD are still unclear and need to be investigated. This study primarily evaluated whether A. indistinctus can improve gut microbiota disorders and prevent the progression from nonalcoholic fatty liver (NAFL) to nonalcoholic steatohepatitis (NASH) in mice fed a high-fat diet (HFD). First, we observed that A. indistinctus significantly improved lipid metabolism disorders and reduced hepatic inflammation induced by HFD consumption in mice. We found that A. indistinctus improved gut barrier function and inhibited the LPS/TLR4/NF-κB pathway, thereby reducing hepatic inflammation. Moreover, 16S rRNA V3-V4 analyses revealed that A. indistinctus could significantly change the structure of the gut microbiota and increase the abundance of L. johnsonii by promoting its growth. Finally, we showed that L. johnsonii administration significantly improved lipid metabolism disorders and reduced hepatic lipid accumulation induced by HFD consumption in mice. In summary, A. indistinctus administration significantly reduces hepatic inflammation by improving gut barrier function and improves lipid metabolism disorders by promoting the growth of L. johnsonii. Our research improves the understanding of the gut microbiota and provides a basis for future therapeutic use of A. indistinctus.
Collapse
Affiliation(s)
- Xiaoxue Xu
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, People's Republic of China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100000, People's Republic of China
| | - Yanrong Wang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, People's Republic of China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100000, People's Republic of China
| | - Xiaofei Wu
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, People's Republic of China
| | - Tianqi Cai
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, People's Republic of China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100000, People's Republic of China
| | - Ling Dong
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, People's Republic of China
| | - Shufei Liang
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, People's Republic of China
| | - Linghui Zhu
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100000, People's Republic of China
| | - Xinhua Song
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, People's Republic of China
| | - Yang Dong
- Monitoring and Statistical Research Center, National Administration of Traditional Chinese Medicine, Beijing, 100021, People's Republic of China
| | - Yanfei Zheng
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100000, People's Republic of China.
| | - Lingru Li
- National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, 100000, People's Republic of China.
| | - Wenlong Sun
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong, 255000, People's Republic of China.
| |
Collapse
|
10
|
Chen Y, Yan S, Yang J, Zhang Y, Suo H, Song J. Integrated microbiome and metabolome analysis reveals the key role of taurohyocholate in the treatment of hyperuricemia with Lacticaseibacillus rhamnosus 2016SWU.05.0601. Food Res Int 2024; 197:115234. [PMID: 39593318 DOI: 10.1016/j.foodres.2024.115234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/15/2024] [Accepted: 10/18/2024] [Indexed: 11/28/2024]
Abstract
The incidence of hyperuricemia (HUA) is on the rise, posing a significant threat to human health. Several probiotics have shown potential in treating HUA; however, the critical role of intestinal metabolites in this therapy remains inadequately understood. Our study demonstrated that Lacticaseibacillus rhamnosus 2016SWU.05.0601 not only reduced the expression levels of xanthine dehydrogenase and the content and activity of xanthine oxidase in the liver but also regulated the uric acid transporters expression in the kidney, thereby attenuating HUA in mice. Additionally, L. rhamnosus 2016SWU.05.0601 modulated the gut microbiota and metabolite abundance in HUA mice. Correlation analysis revealed that the gut microbiota metabolite taurohyocholate played a vital role in the treatment of HUA by L. rhamnosus 2016SWU.05.0601, as confirmed in HUA cell models. Our research provides a significant theoretical basis for elucidating the mechanisms by which probiotics alleviate HUA and for developing functional ingredients for HUA treatment.
Collapse
Affiliation(s)
- Yanchao Chen
- College of Food Science, Southwest University, Chongqing 400715, PR China
| | - Shenglan Yan
- College of Food Science, Southwest University, Chongqing 400715, PR China
| | - Jing Yang
- School of Environment and Resources, Chongqing Technology and Business University, Chongqing 400067, PR China
| | - Yu Zhang
- College of Food Science, Southwest University, Chongqing 400715, PR China
| | - Huayi Suo
- College of Food Science, Southwest University, Chongqing 400715, PR China.
| | - Jiajia Song
- College of Food Science, Southwest University, Chongqing 400715, PR China.
| |
Collapse
|
11
|
Singh AK, Durairajan SSK, Iyaswamy A, Williams LL. Elucidating the role of gut microbiota dysbiosis in hyperuricemia and gout: Insights and therapeutic strategies. World J Gastroenterol 2024; 30:4404-4410. [PMID: 39494101 PMCID: PMC11525862 DOI: 10.3748/wjg.v30.i40.4404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 09/14/2024] [Accepted: 09/26/2024] [Indexed: 10/16/2024] Open
Abstract
Hyperuricemia (HUA) is a condition associated with a high concentration of uric acid (UA) in the bloodstream and can cause gout and chronic kidney disease. The gut microbiota of patients with gout and HUA is significantly altered compared to that of healthy people. This article focused on the complex interconnection between alterations in the gut microbiota and the development of this disorder. Some studies have suggested that changes in the composition, diversity, and activity of microbes play a key role in establishing and progressing HUA and gout pathogenesis. Therefore, we discussed how the gut microbiota contributes to HUA through purine metabolism, UA excretion, and intestinal inflammatory responses. We examined specific changes in the composition of the gut microbiota associated with gout and HUA, highlighting key bacterial taxa and the metabolic pathways involved. Additionally, we discussed the effect of conventional gout treatments on the gut microbiota composition, along with emerging therapeutic approaches that target the gut microbiome, such as the use of probiotics and prebiotics. We also provided insights into a study regarding the gut microbiota as a possible novel therapeutic intervention for gout treatment and dysbiosis-related diagnosis.
Collapse
Affiliation(s)
- Abhay Kumar Singh
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur, TN 610005, India
| | - Siva Sundara Kumar Durairajan
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur, TN 610005, India
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Ashok Iyaswamy
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641021, India
| | - Leonard L Williams
- Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, Greensboro, NC 27411, United States
| |
Collapse
|
12
|
Li J, Liang J, Liu Y, Sun W, Sun W. Basal metabolic rate mediates the causal relationship between gut microbiota and osteoarthritis: a two-step bidirectional Mendelian randomization study. Front Microbiol 2024; 15:1371679. [PMID: 39411433 PMCID: PMC11473340 DOI: 10.3389/fmicb.2024.1371679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
Background The relationship between gut microbiota and osteoarthritis (OA) occurrence remains unclear. Existing research needs to clearly understand how basal metabolic rate (BMR) regulates this relationship. Therefore, using a two-step bidirectional Mendelian Randomization approach, our study aims to investigate whether BMR levels mediate the causal relationship between gut microbiota and OA. Methods In this study, we examined publicly available summary statistics from Genome-Wide Association Studies (GWAS) to determine the correlation between gut microbiota and OA. The analysis included one primary dataset and two secondary datasets. Initially, a two-step, two-sample, and reverse MR analysis was performed to identify the causal relationship between gut microbiota and OA. Subsequently, a two-step MR analysis revealed that the relationship between microbiota and OA is mediated by BMR. Sensitivity analyses confirmed the robustness of the study results. Results In our analysis of the primary dataset, we discovered a positive correlation between three taxa and the outcome of OA, and eight taxa exhibited a negative correlation with the OA outcome. Through comparisons with the secondary dataset and multiple testing corrections, we found a negative association between the class Actinobacteria (OR=0.992886277, p-value = 0.003) and the likelihood of OA occurrence. Notably, knee osteoarthritis (KOA) and hip osteoarthritis (HOA) had a strong negative correlation (OR = 0.927237553/0.892581219). Our analysis suggests that BMR significantly mediates the causal pathway from Actinobacteria to OA, with a mediated effect of 2.59%. Additionally, BMR mediates 3.98% of the impact in the path from the order Bifidobacteriales and the family Bifidobacteriaceae to OA. Besides these findings, our reverse analysis did not indicate any significant effect of OA on gut microbiota or BMR. Conclusion Our research results indicate that an increase in the abundance of specific gut microbial taxa is associated with a reduced incidence of OA, and BMR levels mediate this causal relationship. Further large-scale randomized controlled trials are necessary to validate the causal impact of gut microbiota on the risk of OA. This study provides new insights into the potential prevention of OA by modulating the gut microbiota.
Collapse
Affiliation(s)
- Jiachen Li
- Department of Orthopedics, Shenzhen Second People's Hospital/First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
- Shantou University Medical College, Shantou, China
| | - Jianhui Liang
- Department of Orthopedics, Shenzhen Second People's Hospital/First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
- Shantou University Medical College, Shantou, China
| | - Yang Liu
- Department of Orthopedics, Shenzhen Second People's Hospital/First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Weichao Sun
- Department of Orthopedics, Shenzhen Second People's Hospital/First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
- The Central Laboratory, Shenzhen Second People's Hospital/First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Wei Sun
- Department of Orthopedics, Shenzhen Second People's Hospital/First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| |
Collapse
|
13
|
Hu Y, Wang S, Wang R, Zhang Y, Yuan Q, Yuan C. Total saponins from Panax japonicus regulated the intestinal microbiota to alleviate lipid metabolism disorders in aging mice. Arch Gerontol Geriatr 2024; 125:105500. [PMID: 38851092 DOI: 10.1016/j.archger.2024.105500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/21/2024] [Accepted: 05/25/2024] [Indexed: 06/10/2024]
Abstract
Total saponins from Panax japonicus (TSPJ) have many beneficial physiological activities, particularly in alleviating the damages of aging and abnormal lipid metabolism. This work used mice models to investigate if TSPJ reduced obesity and regulated metabolic functions via the intestinal microbiota, the disturbance of which has been shown to cause aging-related diseases. The results showed that TSPJ significantly reduced the weight and blood lipid level of aging mice. Further analyses showed that TSPJ significantly inhibited adipogenesis, changed the composition of the intestinal flora, and protected the integrity of the intestinal barrier. It was inferred from the accumulated experimental data that TSPJ helped to combat obesity in aging mice by regulating the intestinal microbiota and promoting microbial metabolism.
Collapse
Affiliation(s)
- Yaqi Hu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China; College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Shuwen Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China; College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Rui Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China; College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Yifan Zhang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China; College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Qi Yuan
- Third-Grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, China; College of Medicine and Health Science, China Three Gorges University, Yichang, 443002, China
| | - Chengfu Yuan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, China; College of Basic Medical Science, China Three Gorges University, Yichang 443002, China.
| |
Collapse
|
14
|
Helal P, Xia W, Sardar P, Conway‐Morris A, Conway‐Morris A, Pedicord VA, Serfontein J. Changes in the Firmicutes to Bacteriodetes ratio in the gut microbiome in individuals with anorexia nervosa following inpatient treatment: A systematic review and a case series. Brain Behav 2024; 14:e70014. [PMID: 39295072 PMCID: PMC11410858 DOI: 10.1002/brb3.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 09/21/2024] Open
Abstract
OBJECTIVE Anorexia nervosa has the highest mortality rate among psychiatric illnesses. Current treatments remain ineffective for a large fraction of patients. This may be due to unclear mechanisms behind its development and maintenance. Studies exploring the role of the gut microbiome have revealed inconsistent evidence of dysbiosis. This article aims to investigate changes in the gut microbiome, particularly, mean differences in the Firmicutes to Bacteroidetes ratio, in adolescent and adult individuals with anorexia nervosa following inpatient treatment. METHODS Longitudinal studies investigating gut microbiome composition in inpatient populations of anorexia nervosa before and after treatment were systematically reviewed. Additionally, gut microbiome compositions were characterized in three acute anorexia nervosa inpatients early after admission and after 4-12 weeks of treatment. RESULTS Review results indicated an increase in the Firmicutes to Bacteroidetes ratio in individuals with anorexia nervosa after treatment. These however did not match values of their healthy counterparts. In the case-series samples, the reverse occurred with samples taken 4 weeks after treatment. In the patient who provided an extra sample 12 weeks after treatment, similar results to the studies included in the review were observed. Furthermore, Firmicutes to Bacteroidetes ratio values in the case-series samples were notably higher in the two patients who had chronic anorexia nervosa. DISCUSSION Differences in methodologies, small sample sizes, and insufficient data limited the generalizability of the outcomes of the reviewed studies. Results suggest a potentially unique microbiome signature in individuals with chronic anorexia nervosa, which may explain different outcomes in this group of patients.
Collapse
Affiliation(s)
- Passent Helal
- Adult Eating Disorders Service, Ward S3 InpatientsAddenbrooke's HospitalCambridgeUK
| | - Wangmingyu Xia
- Department of Medicine, Cambridge Institute of Therapeutic Immunology & Infectious DiseaseUniversity of CambridgeCambridgeUK
| | - Puspendu Sardar
- Department of Medicine, Cambridge Institute of Therapeutic Immunology & Infectious DiseaseUniversity of CambridgeCambridgeUK
| | - Anna Conway‐Morris
- Adult Eating Disorder Service, Ward S3 OutpatientsAddenbrooke's HospitalCambridgeUK
- School of PsychiatryNHS EnglandFulbournCambridgeUK
| | - Andrew Conway‐Morris
- Division of Anaesthesia, Department of MedicineUniversity of CambridgeCambridgeUK
- Division of Immunology, Department of PathologyUniversity of CambridgeCambridgeUK
- John V Farman Intensive Care UnitAddenbrooke's HospitalCambridgeUK
| | - Virginia A. Pedicord
- Department of Medicine, Cambridge Institute of Therapeutic Immunology & Infectious DiseaseUniversity of CambridgeCambridgeUK
| | - Jaco Serfontein
- Adult Eating Disorders Service, Ward S3 InpatientsAddenbrooke's HospitalCambridgeUK
| |
Collapse
|
15
|
Ezenabor EH, Adeyemi AA, Adeyemi OS. Gut Microbiota and Metabolic Syndrome: Relationships and Opportunities for New Therapeutic Strategies. SCIENTIFICA 2024; 2024:4222083. [PMID: 39041052 PMCID: PMC11262881 DOI: 10.1155/2024/4222083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/10/2024] [Accepted: 07/04/2024] [Indexed: 07/24/2024]
Abstract
Since its discovery, numerous studies have shown the role of the microbiota in well-being and disease. The gut microbiota represents an essential factor that plays a multidirectional role that affects not just the gut but also other parts of the body, including the brain, endocrine system, humoral system, immune system, and metabolic pathways, as well as host-microbiome interactions. Through a comprehensive analysis of existing literature using the desktop research methodology, this review elucidates the mechanisms by which gut microbiota dysbiosis contributes to metabolic dysfunction, including obesity, dyslipidaemia, hypertension, atherosclerosis, hyperuricemia, and hyperglycaemia. Furthermore, it examines the bidirectional communication pathways between gut microbiota and host metabolism, highlighting the role of microbial-derived metabolites, immune modulation, and gut barrier integrity in shaping metabolic homeostasis. Importantly, the review identifies promising therapeutic strategies targeting the gut microbiota as potential interventions for metabolic syndrome, including probiotics, prebiotics, symbiotics, dietary modifications, and faecal microbiota transplantation. By delineating the bidirectional interactions between gut microbiota and metabolic syndrome, the review not only advances our understanding of disease pathophysiology but also underscores the potential for innovative microbiota-based interventions to mitigate the global burden of metabolic syndrome and its associated complications.
Collapse
Affiliation(s)
- Emmanuel Henry Ezenabor
- Department of BiochemistryMedicinal Biochemistry, Nanomedicine & Toxicology LaboratoryBowen University, Iwo 232102, Osun State, Nigeria
| | - Aishat Abimbola Adeyemi
- Department of BiochemistryMedicinal Biochemistry, Nanomedicine & Toxicology LaboratoryBowen University, Iwo 232102, Osun State, Nigeria
| | - Oluyomi Stephen Adeyemi
- Department of BiochemistryMedicinal Biochemistry, Nanomedicine & Toxicology LaboratoryBowen University, Iwo 232102, Osun State, Nigeria
| |
Collapse
|
16
|
Lee J, Wellenstein K, Rahnavard A, Nelson AT, Holter MM, Cummings BP, Yeliseyev V, Castoldi A, Clish CB, Bry L, Siegel D, Kahn BB. Beneficial metabolic effects of PAHSAs depend on the gut microbiota in diet-induced obese mice but not in chow-fed mice. Proc Natl Acad Sci U S A 2024; 121:e2318691121. [PMID: 38968121 PMCID: PMC11252816 DOI: 10.1073/pnas.2318691121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 05/31/2024] [Indexed: 07/07/2024] Open
Abstract
Dietary lipids play an essential role in regulating the function of the gut microbiota and gastrointestinal tract, and these luminal interactions contribute to mediating host metabolism. Palmitic Acid Hydroxy Stearic Acids (PAHSAs) are a family of lipids with antidiabetic and anti-inflammatory properties, but whether the gut microbiota contributes to their beneficial effects on host metabolism is unknown. Here, we report that treating chow-fed female and male germ-free (GF) mice with PAHSAs improves glucose tolerance, but these effects are lost upon high fat diet (HFD) feeding. However, transfer of feces from PAHSA-treated, but not vehicle-treated, chow-fed conventional mice increases insulin sensitivity in HFD-fed GF mice. Thus, the gut microbiota is necessary for, and can transmit, the insulin-sensitizing effects of PAHSAs in HFD-fed GF male mice. Analyses of the cecal metagenome and lipidome of PAHSA-treated mice identified multiple lipid species that associate with the gut commensal Bacteroides thetaiotaomicron (Bt) and with insulin sensitivity resulting from PAHSA treatment. Supplementing live, and to some degree, heat-killed Bt to HFD-fed female mice prevented weight gain, reduced adiposity, improved glucose tolerance, fortified the colonic mucus barrier and reduced systemic inflammation compared to HFD-fed controls. These effects were not observed in HFD-fed male mice. Furthermore, ovariectomy partially reversed the beneficial Bt effects on host metabolism, indicating a role for sex hormones in mediating the Bt probiotic effects. Altogether, these studies highlight the fact that PAHSAs can modulate the gut microbiota and that the microbiota is necessary for the beneficial metabolic effects of PAHSAs in HFD-fed mice.
Collapse
Affiliation(s)
- Jennifer Lee
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA02215
| | - Kerry Wellenstein
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA02215
| | - Ali Rahnavard
- Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Washington, DC20052
| | - Andrew T. Nelson
- Division of Pharmaceutical Chemistry, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA92093
| | - Marlena M. Holter
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Cornell University, Ithaca, NY14850
| | - Bethany P. Cummings
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, CA95817
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California Davis School of Veterinary Medicine, Davis, CA95616
| | - Vladimir Yeliseyev
- Massachusetts Host-Microbiome Center, Department of Pathology, Brigham & Women's Hospital and Harvard Medical School, Boston, MA02115
| | - Angela Castoldi
- Laboratory of Immunopathology Keizo Asami, Federal University of Pernambuco, Recife50670-901, Brazil
| | - Clary B. Clish
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
| | - Lynn Bry
- Massachusetts Host-Microbiome Center, Department of Pathology, Brigham & Women's Hospital and Harvard Medical School, Boston, MA02115
| | - Dionicio Siegel
- Division of Pharmaceutical Chemistry, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA92093
| | - Barbara B. Kahn
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA02215
| |
Collapse
|
17
|
Xie J, Luo M, Chen Q, Zhang Q, Qin L, Wang Y, Zhao Y, He Y. Hypolipidemic effect and gut microbiota regulation of Gypenoside aglycones in rats fed a high-fat diet. JOURNAL OF ETHNOPHARMACOLOGY 2024; 328:118066. [PMID: 38499259 DOI: 10.1016/j.jep.2024.118066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 03/20/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Gynostemma pentaphyllum (Thunb.) Makino has traditional applications in Chinese medicine to treat lipid abnormalities. Gypenosides (GPs), the main bioactive components of Gynostemma pentaphyllum, have been reported to exert hypolipidemic effects through multiple mechanisms. The lipid-lowering effects of GPs may be attributed to the aglycone portion resulting from hydrolysis of GPs by the gut microbiota. However, to date, there have been no reports on whether gypenoside aglycones (Agl), the primary bioactive constituents, can ameliorate hyperlipidemia by modulating the gut microbiota. AIM OF THE STUDY This study explored the potential therapeutic effects of gypenoside aglycone (Agl) in a rat model of high-fat diet (HFD)-induced hyperlipidemia. METHODS A hyperlipidemic rat model was established by feeding rats with a high-fat diet. Agl was administered orally, and serum lipid levels were analyzed. Molecular techniques, including RT-polymerase chain reaction (PCR) and fecal microbiota sequencing, were used to investigate the effects of Agl on lipid metabolism and gut microbiota composition. RESULTS Agl administration significantly reduced serum levels of total cholesterol (TC), triglycerides (TG), and low-density lipoprotein cholesterol (LDL-C) and mitigated hepatic damage induced by HFD. Molecular investigations have revealed the modulation of key lipid metabolism genes and proteins by Agl. Notably, Agl treatment enriched the gut microbiota with beneficial genera, including Lactobacillus, Akkermansia, and Blautia and promoted specific shifts in Lactobacillus murinus, Firmicutes bacterium CAG:424, and Allobaculum stercoricanis. CONCLUSION This comprehensive study established Agl as a promising candidate for the treatment of hyperlipidemia. It also exhibits remarkable hypolipidemic and hepatoprotective properties. The modulation of lipid metabolism-related genes, along with the restoration of gut microbiota balance, provides mechanistic insights. Thus, Agl has great potential for clinical applications in hyperlipidemia management.
Collapse
Affiliation(s)
- Jian Xie
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi, 563000, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China; 2011 Cooperative Inovational Center for Guizhou Traditional Chinese Medicine and Ethnic Medicine Zunyi Medical University, Zunyi, 563000, China; Department of Medical Genetics, Zunyi Medical University, Zunyi, 563000, China.
| | - Mingxia Luo
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi, 563000, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China; 2011 Cooperative Inovational Center for Guizhou Traditional Chinese Medicine and Ethnic Medicine Zunyi Medical University, Zunyi, 563000, China.
| | - Qiuyi Chen
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi, 563000, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China; 2011 Cooperative Inovational Center for Guizhou Traditional Chinese Medicine and Ethnic Medicine Zunyi Medical University, Zunyi, 563000, China.
| | - Qianru Zhang
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi, 563000, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China; 2011 Cooperative Inovational Center for Guizhou Traditional Chinese Medicine and Ethnic Medicine Zunyi Medical University, Zunyi, 563000, China.
| | - Lin Qin
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi, 563000, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China; 2011 Cooperative Inovational Center for Guizhou Traditional Chinese Medicine and Ethnic Medicine Zunyi Medical University, Zunyi, 563000, China.
| | - Yuhe Wang
- Department of Pharmacy, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, China.
| | - Yongxia Zhao
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi, 563000, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China; 2011 Cooperative Inovational Center for Guizhou Traditional Chinese Medicine and Ethnic Medicine Zunyi Medical University, Zunyi, 563000, China.
| | - Yuqi He
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Zunyi Medical University, Zunyi, 563000, China; Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, 563000, China; 2011 Cooperative Inovational Center for Guizhou Traditional Chinese Medicine and Ethnic Medicine Zunyi Medical University, Zunyi, 563000, China.
| |
Collapse
|
18
|
Zhuo LB, Yang Y, Xiao C, Li F, Lin L, Xi Y, Fu Y, Zheng JS, Chen YM. Gut microbiota-bile acid axis mediated the beneficial associations between dietary lignans and hyperuricemia: a prospective study. Food Funct 2024; 15:6438-6449. [PMID: 38775706 DOI: 10.1039/d4fo00961d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Background: The escalating prevalence of hyperuricemia is emerging as a significant public health concern. The association between dietary lignans and hyperuricemia is yet to be fully elucidated. Our study aims to evaluate the relationships between dietary lignan intake and hyperuricemia among middle-aged and elderly Chinese individuals, with an additional focus on investigating the underlying mechanisms. Methods: Dietary lignan intake was measured using a validated Food Frequency Questionnaire in 3801 participants at the baseline. Among them, 2552 participants were included in the longitudinal study with a median follow-up of 10.5 years. The gut microbiota was analyzed by shotgun metagenome sequencing in 1789 participants, and the targeted fecal metabolome was determined in 987 participants using UPLC-MS/MS at the midpoint of follow-up. Results: The multivariable-adjusted HRs (95% CIs) for hyperuricemia incidence in the highest quartile (vs. the lowest quartile) of dietary intake of total lignans, matairesinol, pinoresinol, and secoisolariciresinol were 0.93 (0.78-1.10), 0.77 (0.66-0.90), 0.83 (0.70-0.97), and 0.85 (0.73-1.00), respectively. The gut microbial and fecal metabolic compositions were significantly different across the dietary lignan groups and the hyperuricemia groups. The beneficial associations between dietary lignans and hyperuricemia might be mediated by several gut microbes (e.g., Fusobacterium mortiferum and Blautia sp. CAG-257) and the downstream bile acid products (e.g., NorCA, glycochenodeoxycholic acid, and glycoursodeoxycholic acid). Conclusion: We found that dietary lignans were inversely associated with hyperuricemia incidence, and the gut microbiota-bile acid axis might mediate this association. Our findings provide new perspectives on precise therapeutic targets and underlying mechanisms for conditions associated with elevated uric acid.
Collapse
Affiliation(s)
- Lai-Bao Zhuo
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Yingdi Yang
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Congmei Xiao
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, School of Medicine and School of Life Sciences, Westlake University, Hangzhou 310030, China.
- Shenzhen Bao'an Center for Chronic Diseases Control, Shenzhen, China
| | - Fanqin Li
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Lishan Lin
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Yue Xi
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Yuanqing Fu
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, School of Medicine and School of Life Sciences, Westlake University, Hangzhou 310030, China.
- Shenzhen Bao'an Center for Chronic Diseases Control, Shenzhen, China
| | - Ju-Sheng Zheng
- Zhejiang Key Laboratory of Multi-Omics in Infection and Immunity, School of Medicine and School of Life Sciences, Westlake University, Hangzhou 310030, China.
| | - Yu-Ming Chen
- Department of Epidemiology, Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
19
|
Guo X, Han J, Hong L, Huang Y, Li S, Zhang L, Yan W, Dong P, Yang Y, Cao Y. Associations of Early Gut Microbiome and Metabolome with Growth and Body Composition of Preterm Infants Within the First 6 Months. Breastfeed Med 2024; 19:435-444. [PMID: 38501370 DOI: 10.1089/bfm.2023.0258] [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] [Indexed: 03/20/2024]
Abstract
Objectives: This study aimed to explore the associations of growth and body composition with gut microbiome and metabolome in preterm infants. Materials and Methods: A prospective cohort study including 73 human milk-fed very preterm infants was conducted. During hospitalization, fecal samples were collected to detect microbes and metabolites using 16S rRNA gene sequencing and liquid chromatography-mass spectrometry. Growth and body composition indices were measured at term equivalent age (TEA) and 6 months of corrected age (CA). Associations of the fecal microbiome and metabolome profiles with growth and body composition indices, as well as their changes, were analyzed. Results: A higher abundance of Streptococcus was associated with a lower fat-free mass (FFM) z-score at 6 months of CA (p = 0.002) and a smaller increase in FFM z-score from TEA to 6 months of CA (p = 0.018). Higher levels of 3'-sialyllactose and 6'-sialyllactose (6'-SL) in feces were correlated with a lower z-score of percentage body fat (PBF) (p = 0.018 and 0.020, respectively) and a lower z-score of fat mass (p = 0.044 and 0.043, respectively) at 6 months of CA. A higher level of 6'-SL in feces was correlated with a greater increase in FFM z-score from TEA to 6 months of CA (p = 0.021). Conclusions: This study sheds light on the role of specific microbial-host interactions in metabolic changes in preterm infants, indicating the potential role of sialylated human milk oligosaccharides in optimizing body composition.
Collapse
Affiliation(s)
- Xinhui Guo
- Department of Neonatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, People's Republic of China
| | - Junyan Han
- Department of Neonatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, People's Republic of China
| | - Luyang Hong
- Department of Neonatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, People's Republic of China
| | - Yihuang Huang
- Department of Neonatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, People's Republic of China
| | - Shujuan Li
- Department of Neonatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, People's Republic of China
| | - Lan Zhang
- Department of Neonatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, People's Republic of China
| | - Weili Yan
- Department of Clinical Epidemiology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, People's Republic of China
| | - Ping Dong
- Department of Child Healthcare, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, People's Republic of China
| | - Yi Yang
- NHC Key Laboratory of Neonatal Diseases, Fudan University, Children's Hospital of Fudan University, Shanghai, People's Republic of China
| | - Yun Cao
- Department of Neonatology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, People's Republic of China
- NHC Key Laboratory of Neonatal Diseases, Fudan University, Children's Hospital of Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
20
|
Wu J, Aga L, Tang L, Li H, Wang N, Yang L, Zhang N, Wang X, Wang X. Lacticaseibacillus paracasei JS-3 Isolated from "Jiangshui" Ameliorates Hyperuricemia by Regulating Gut Microbiota and iTS Metabolism. Foods 2024; 13:1371. [PMID: 38731742 PMCID: PMC11083236 DOI: 10.3390/foods13091371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Background: A diet high in purines can impair the function of the gut microbiota and disrupt purine metabolism, which is closely associated with the onset of hyperuricemia. Dietary regulation and intestinal health maintenance are key approaches for controlling uric acid (UA) levels. Investigating the impacts of fermented foods offers potential dietary interventions for managing hyperuricemia. Methods: In this study, we isolated a strain with potent UA-degrading capabilities from "Jiangshui", a fermented food product from Gansu, China. We performed strain identification and assessed its probiotic potential. Hyperuricemic quails, induced by a high-purine diet, were used to assess the UA degradation capability of strain JS-3 by measuring UA levels in serum and feces. Additionally, the UA degradation pathways were elucidated through analyses of the gut microbiome and fecal metabolomics. Results: JS-3, identified as Lacticaseibacillus paracasei, was capable of eliminating 16.11% of uric acid (UA) within 72 h, rapidly proliferating and producing acid within 12 h, and surviving in the gastrointestinal tract. Using hyperuricemic quail models, we assessed JS-3's UA degradation capacity. Two weeks after the administration of JS-3 (2 × 108 cfu/d per quail), serum uric acid (SUA) levels significantly decreased to normal levels, and renal damage in quails was markedly improved. Concurrently, feces from the JS-3 group demonstrated a significant degradation of UA, achieving up to 49% within 24 h. 16S rRNA sequencing revealed JS-3's role in gut microbiota restoration by augmenting the probiotic community (Bifidobacterium, Bacteroides unclassified_f-Lachnospiraceae, and norank_fynorank_o-Clostridia_UCG-014) and diminishing the pathogenic bacteria (Macrococus and Lactococcus). Corresponding with the rise in short-chain fatty acid (SCFA)-producing bacteria, JS-3 significantly increased SCFA levels (p < 0.05, 0.01). Additionally, JS-3 ameliorated metabolic disturbances in hyperuricemic quails, influencing 26 abnormal metabolites predominantly linked to purine, tryptophan, and bile acid metabolism, thereby enhancing UA degradation and renal protection. Conclusions: For the first time, we isolated and identified an active probiotic strain, JS-3, from the "Jiangshui" in Gansu, used for the treatment of hyperuricemia. It modulates host-microbiome interactions, impacts the metabolome, enhances intestinal UA degradation, reduces levels of SUA and fecal UA, alleviates renal damage, and effectively treats hyperuricemia without causing gastrointestinal damage. In summary, JS-3 can serve as a probiotic with potential therapeutic value for the treatment of hyperuricemia.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xueyong Wang
- School of Chinese Meteria Medica, Beijing University of Chinese Medicine, Northeast Corner of Intersection of Sunshine South Street and Baiyang East Road, Fang-Shan District, Beijing 102488, China; (J.W.); (L.T.); (H.L.); (N.W.)
| |
Collapse
|
21
|
Zhou Y, Zeng Y, Wang R, Pang J, Wang X, Pan Z, Jin Y, Chen Y, Yang Y, Ling W. Resveratrol Improves Hyperuricemia and Ameliorates Renal Injury by Modulating the Gut Microbiota. Nutrients 2024; 16:1086. [PMID: 38613119 PMCID: PMC11013445 DOI: 10.3390/nu16071086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 03/25/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024] Open
Abstract
Resveratrol (RES) has been reported to prevent hyperuricemia (HUA); however, its effect on intestinal uric acid metabolism remains unclear. This study evaluated the impact of RES on intestinal uric acid metabolism in mice with HUA induced by a high-fat diet (HFD). Moreover, we revealed the underlying mechanism through metagenomics, fecal microbiota transplantation (FMT), and 16S ribosomal RNA analysis. We demonstrated that RES reduced the serum uric acid, creatinine, urea nitrogen, and urinary protein levels, and improved the glomerular atrophy, unclear renal tubule structure, fibrosis, and renal inflammation. The results also showed that RES increased intestinal uric acid degradation. RES significantly changed the intestinal flora composition of HFD-fed mice by enriching the beneficial bacteria that degrade uric acid, reducing harmful bacteria that promote inflammation, and improving microbial function via the upregulation of purine metabolism. The FMT results further showed that the intestinal microbiota is essential for the effect of RES on HUA, and that Lactobacillus may play a key role in this process. The present study demonstrated that RES alleviates HFD-induced HUA and renal injury by regulating the gut microbiota composition and the metabolism of uric acid.
Collapse
Affiliation(s)
- Yuqing Zhou
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Y.Z.); (Y.Z.); (J.P.); (X.W.); (Z.P.); (Y.J.); (Y.C.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China;
| | - Yupeng Zeng
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Y.Z.); (Y.Z.); (J.P.); (X.W.); (Z.P.); (Y.J.); (Y.C.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China;
| | - Ruijie Wang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China;
- Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou 510080, China
- Department of Nutrition, School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Juan Pang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Y.Z.); (Y.Z.); (J.P.); (X.W.); (Z.P.); (Y.J.); (Y.C.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China;
| | - Xin Wang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Y.Z.); (Y.Z.); (J.P.); (X.W.); (Z.P.); (Y.J.); (Y.C.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China;
| | - Zhijun Pan
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Y.Z.); (Y.Z.); (J.P.); (X.W.); (Z.P.); (Y.J.); (Y.C.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China;
| | - Yufeng Jin
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Y.Z.); (Y.Z.); (J.P.); (X.W.); (Z.P.); (Y.J.); (Y.C.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China;
| | - Yu Chen
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Y.Z.); (Y.Z.); (J.P.); (X.W.); (Z.P.); (Y.J.); (Y.C.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China;
| | - Yan Yang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China;
- Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou 510080, China
- Department of Nutrition, School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen 518107, China
| | - Wenhua Ling
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; (Y.Z.); (Y.Z.); (J.P.); (X.W.); (Z.P.); (Y.J.); (Y.C.)
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou 510080, China;
- Guangdong Engineering Technology Center of Nutrition Transformation, Guangzhou 510080, China
| |
Collapse
|
22
|
Xu CL, Wang C, Li GB, Zhao T, Zhou RL, Chen J. Antibiotic administration aggravates asthma by disrupting gut microbiota and the intestinal mucosal barrier in an asthma mouse model. Exp Ther Med 2024; 27:157. [PMID: 38476896 PMCID: PMC10928978 DOI: 10.3892/etm.2024.12445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 01/29/2024] [Indexed: 03/14/2024] Open
Abstract
In humans, gut microbiota can determine the health status. The regulatory mechanisms of the gut microbiota in asthma must be elucidated. Although antibiotics (ABXs) can clear infections, they markedly alter the composition and abundance of gut microbiota. The present study used ABX-treated mice to examine the time-dependent effects of ABX administration on the gut microbiota and intestinal mucosal barrier. The mouse asthma model was established using ovalbumin (OVA) and gavaged with an ABX cocktail for different durations (1 or 2 weeks) and stacked sequences. The pathology of the model, model 2, OVA-ABX, OVA-ABX 2, ABX-OVA and ABX-OVA was severe when compared with the control group as evidenced by the following results: i) significantly increased pulmonary and colonic inflammatory cell infiltration; ii) enhanced pause values and iii) OVA-induced immunoglobulin E (IgE) and TGF-β expression levels, and significantly downregulated Tight Junction Protein 1 (TJP1), claudin 1 and Occludin expression levels. Furthermore, the intestinal bacterial load in the OVA-ABX and OVA-ABX 2 groups was significantly lower than that in the ABX-OVA and ABX-OVA 2 groups, respectively. The predominant taxa were as follows: phyla, Firmicutes and Proteobacteria, genera, Escherichia-Shigella, Lactobacillus and Lachnospira. The abundances of Lachnospira and Escherichia-Shigella were correlated with the expression of OVA-induced IgE and TJPs. These findings indicated that ABX administration, which modifies microbiome diversity and bacterial abundance, can disrupt colonic integrity, downregulate TJ proteins, damage the intestinal barrier, enhance enterocyte permeability, and promote the release of inflammatory factors, adversely affecting asthma alleviation and long-term repair.
Collapse
Affiliation(s)
- Cheng-Ling Xu
- College of Basic Medical Sciences, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Cui Wang
- College of Basic Medical Sciences, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Gao-Bin Li
- College of Basic Medical Sciences, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Tong Zhao
- College of Basic Medical Sciences, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Rui-Ling Zhou
- Department of Dermatology, First Affiliated Hospital, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650504, P.R. China
| | - Jing Chen
- College of Basic Medical Sciences, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
23
|
Alvernaz SA, Wenzel ES, Nagelli U, Pezley LB, LaBomascus B, Gilbert JA, Maki PM, Tussing-Humphreys L, Peñalver Bernabé B. Inflammatory Dietary Potential Is Associated with Vitamin Depletion and Gut Microbial Dysbiosis in Early Pregnancy. Nutrients 2024; 16:935. [PMID: 38612969 PMCID: PMC11013194 DOI: 10.3390/nu16070935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 04/14/2024] Open
Abstract
Pregnancy alters many physiological systems, including the maternal gut microbiota. Diet is a key regulator of this system and can alter the host immune system to promote inflammation. Multiple perinatal disorders have been associated with inflammation, maternal metabolic alterations, and gut microbial dysbiosis, including gestational diabetes mellitus, pre-eclampsia, preterm birth, and mood disorders. However, the effects of high-inflammatory diets on the gut microbiota during pregnancy have yet to be fully explored. We aimed to address this gap using a system-based approach to characterize associations among dietary inflammatory potential, a measure of diet quality, and the gut microbiome during pregnancy. Forty-seven pregnant persons were recruited prior to 16 weeks of gestation. Participants completed a food frequency questionnaire (FFQ) and provided fecal samples. Dietary inflammatory potential was assessed using the Dietary Inflammatory Index (DII) from the FFQ data. Fecal samples were analyzed using 16S rRNA amplicon sequencing. Differential taxon abundances with respect to the DII score were identified, and the microbial metabolic potential was predicted using PICRUSt2. Inflammatory diets were associated with decreased vitamin and mineral intake and a dysbiotic gut microbiota structure and predicted metabolism. Gut microbial compositional differences revealed a decrease in short-chain fatty acid producers such as Faecalibacterium, and an increase in predicted vitamin B12 synthesis, methylglyoxal detoxification, galactose metabolism, and multidrug efflux systems in pregnant individuals with increased DII scores. Dietary inflammatory potential was associated with a reduction in the consumption of vitamins and minerals and predicted gut microbiota metabolic dysregulation.
Collapse
Affiliation(s)
- Suzanne A. Alvernaz
- Department of Biomedical Engineering, University of Illinois, Chicago, IL 60607, USA; (S.A.A.); (U.N.)
| | - Elizabeth S. Wenzel
- Department of Psychology, University of Illinois, Chicago, IL 60607, USA; (E.S.W.); (P.M.M.)
| | - Unnathi Nagelli
- Department of Biomedical Engineering, University of Illinois, Chicago, IL 60607, USA; (S.A.A.); (U.N.)
| | - Lacey B. Pezley
- Department of Kinesiology and Nutrition, University of Illinois, Chicago, IL 60612, USA; (L.B.P.); (B.L.); (L.T.-H.)
| | - Bazil LaBomascus
- Department of Kinesiology and Nutrition, University of Illinois, Chicago, IL 60612, USA; (L.B.P.); (B.L.); (L.T.-H.)
| | - Jack A. Gilbert
- Department of Pediatrics, University of California, San Diego, CA 92093, USA;
- Scripps Oceanographic Institute, University of California, San Diego, CA 92037, USA
| | - Pauline M. Maki
- Department of Psychology, University of Illinois, Chicago, IL 60607, USA; (E.S.W.); (P.M.M.)
- Department of Psychiatry, University of Illinois, Chicago, IL 60612, USA
- Department of Obstetrics and Gynecology, University of Illinois, Chicago, IL 60612, USA
| | - Lisa Tussing-Humphreys
- Department of Kinesiology and Nutrition, University of Illinois, Chicago, IL 60612, USA; (L.B.P.); (B.L.); (L.T.-H.)
| | - Beatriz Peñalver Bernabé
- Department of Biomedical Engineering, University of Illinois, Chicago, IL 60607, USA; (S.A.A.); (U.N.)
- Center for Bioinformatics and Quantitative Biology, University of Illinois, Chicago, IL 60612, USA
| |
Collapse
|
24
|
Hussain A, Rui B, Ullah H, Dai P, Ahmad K, Yuan J, Liu Y, Li M. Limosilactobacillus reuteri HCS02-001 Attenuates Hyperuricemia through Gut Microbiota-Dependent Regulation of Uric Acid Biosynthesis and Excretion. Microorganisms 2024; 12:637. [PMID: 38674582 PMCID: PMC11052267 DOI: 10.3390/microorganisms12040637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
Hyperuricemia is a prevalent metabolic disorder that arises from abnormal purine metabolism and reduced excretion of uric acid (UA). The gut microbiota plays a significant role in the biosynthesis and excretion of UA. Probiotics capable of purine degradation possess the potential to prevent hyperuricemia. Our study aimed to screen probiotics in areas with abundant dairy products and longevity populations in China, which could attenuate the level of UA and explore the underlying mechanism. In this study, twenty-three lactic acid bacteria isolated from healthy Chinese infant feces and traditional fermented foods such as hurood and lump milk were evaluated for the ability to tolerance acid, bile, artificial gastric juice, and artificial intestinal juice to determine the potential of the candidate strains as probiotics. Eight strains were identified as possessing superior tolerance to simulated intestinal conditions and were further analyzed by high-performance liquid chromatography (HPLC), revealing that Limosilactobacillus reuteri HCS02-001 (Lact-1) and Lacticaseibacillus paracasei HCS17-040 (Lact-2) possess the most potent ability to degrade purine nucleosides. The effect of Lact-1 and Lact-2 on hyperuricemia was evaluated by intervening with them in the potassium oxonate and adenine-induced hyperuricemia Balb/c mice model in vivo. Our results showed that the level of serum UA in hyperuricemic mice can be efficiently reduced via the oral administration of Lact-1 (p < 0.05). It significantly inhibited the levels of liver inflammatory cytokines and hepatic xanthine oxidase through a TLR4/MyD88/NF-κB pathway across the gut-liver axis. Furthermore, UA transporters ABCG2 and SLC2A9 were substantially upregulated by the intervention of this probiotic. Fecal ATP levels were significantly induced, while fecal xanthine dehydrogenase and allantoinase levels were increased following probiotics. RNA sequencing of HT-29 cells line treated with Lact-1 and its metabolites demonstrated significant regulation of pathways related to hyperuricemia. In summary, these findings demonstrate that Limosilactobacillus reuteri HCS02-001 possesses a capacity to ameliorate hyperuricemia by inhibiting UA biosynthesis via enhancing gastrointestinal barrier functions and promoting UA removal through the upregulation of urate transporters, thereby providing a basis for the probiotic formulation by targeting the gut microbiota.
Collapse
Affiliation(s)
- Akbar Hussain
- College of Basic Medical Science, Dalian Medical University, Dalian 116041, China; (A.H.); (B.R.); (H.U.); (J.Y.)
| | - Binqi Rui
- College of Basic Medical Science, Dalian Medical University, Dalian 116041, China; (A.H.); (B.R.); (H.U.); (J.Y.)
| | - Hayan Ullah
- College of Basic Medical Science, Dalian Medical University, Dalian 116041, China; (A.H.); (B.R.); (H.U.); (J.Y.)
| | - Panpan Dai
- College of Basic Medical Science, Dalian Medical University, Dalian 116041, China; (A.H.); (B.R.); (H.U.); (J.Y.)
| | - Kabir Ahmad
- Department of Physiology, Dalian Medical University, Dalian 116041, China;
| | - Jieli Yuan
- College of Basic Medical Science, Dalian Medical University, Dalian 116041, China; (A.H.); (B.R.); (H.U.); (J.Y.)
| | - Yinhui Liu
- College of Basic Medical Science, Dalian Medical University, Dalian 116041, China; (A.H.); (B.R.); (H.U.); (J.Y.)
| | - Ming Li
- College of Basic Medical Science, Dalian Medical University, Dalian 116041, China; (A.H.); (B.R.); (H.U.); (J.Y.)
| |
Collapse
|
25
|
Xu YX, Liu LD, Zhu JY, Zhu SS, Ye BQ, Yang JL, Huang JY, Huang ZH, You Y, Li WK, He JL, Xia M, Liu Y. Alistipes indistinctus-derived hippuric acid promotes intestinal urate excretion to alleviate hyperuricemia. Cell Host Microbe 2024; 32:366-381.e9. [PMID: 38412863 DOI: 10.1016/j.chom.2024.02.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/10/2024] [Accepted: 02/02/2024] [Indexed: 02/29/2024]
Abstract
Hyperuricemia induces inflammatory arthritis and accelerates the progression of renal and cardiovascular diseases. Gut microbiota has been linked to the development of hyperuricemia through unclear mechanisms. Here, we show that the abundance and centrality of Alistipes indistinctus are depleted in subjects with hyperuricemia. Integrative metagenomic and metabolomic analysis identified hippuric acid as the key microbial effector that mediates the uric-acid-lowering effect of A. indistinctus. Mechanistically, A. indistinctus-derived hippuric acid enhances the binding of peroxisome-proliferator-activated receptor γ (PPARγ) to the promoter of ATP-binding cassette subfamily G member 2 (ABCG2), which in turn boosts intestinal urate excretion. To facilitate this enhanced excretion, hippuric acid also promotes ABCG2 localization to the brush border membranes in a PDZ-domain-containing 1 (PDZK1)-dependent manner. These findings indicate that A. indistinctus and hippuric acid promote intestinal urate excretion and offer insights into microbiota-host crosstalk in the maintenance of uric acid homeostasis.
Collapse
Affiliation(s)
- Ying-Xi Xu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Lu-Di Liu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Jiang-Yuan Zhu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Shan-Shan Zhu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Bing-Qi Ye
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Jia-Lu Yang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Jing-Yi Huang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Zhi-Hao Huang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Yi You
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Wen-Kang Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Jia-Lin He
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Min Xia
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China
| | - Yan Liu
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, and Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, Guangdong, P.R. China.
| |
Collapse
|
26
|
Trubitsina NP, Matiiv AB, Rogoza TM, Zudilova AA, Bezgina MD, Zhouravleva GA, Bondarev SA. Role of the Gut Microbiome and Bacterial Amyloids in the Development of Synucleinopathies. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:523-542. [PMID: 38648770 DOI: 10.1134/s0006297924030118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/16/2024] [Accepted: 01/24/2024] [Indexed: 04/25/2024]
Abstract
Less than ten years ago, evidence began to accumulate about association between the changes in the composition of gut microbiota and development of human synucleinopathies, in particular sporadic form of Parkinson's disease. We collected data from more than one hundred and thirty experimental studies that reported similar results and summarized the frequencies of detection of different groups of bacteria in these studies. It is important to note that it is extremely rare that a unidirectional change in the population of one or another group of microorganisms (only an elevation or only a reduction) was detected in the patients with Parkinson's disease. However, we were able to identify several groups of bacteria that were overrepresented in the patients with Parkinson's disease in the analyzed studies. There are various hypotheses about the molecular mechanisms that explain such relationships. Usually, α-synuclein aggregation is associated with the development of inflammatory processes that occur in response to the changes in the microbiome. However, experimental evidence is accumulating on the influence of bacterial proteins, including amyloids (curli), as well as various metabolites, on the α-synuclein aggregation. In the review, we provided up-to-date information about such examples.
Collapse
Affiliation(s)
- Nina P Trubitsina
- Department of Genetics and Biotechnology, Saint Petersburg State University, Saint Petersburg, 199034, Russia
| | - Anton B Matiiv
- Department of Genetics and Biotechnology, Saint Petersburg State University, Saint Petersburg, 199034, Russia
| | - Tatyana M Rogoza
- Department of Genetics and Biotechnology, Saint Petersburg State University, Saint Petersburg, 199034, Russia
- St. Petersburg Branch of the Vavilov Institute of General Genetics, Saint Petersburg, 198504, Russia
| | - Anna A Zudilova
- Department of Genetics and Biotechnology, Saint Petersburg State University, Saint Petersburg, 199034, Russia
| | - Mariya D Bezgina
- Department of Genetics and Biotechnology, Saint Petersburg State University, Saint Petersburg, 199034, Russia
| | - Galina A Zhouravleva
- Department of Genetics and Biotechnology, Saint Petersburg State University, Saint Petersburg, 199034, Russia
- Laboratory of Amyloid Biology, Saint Petersburg State University, Saint Petersburg, 199034, Russia
| | - Stanislav A Bondarev
- Department of Genetics and Biotechnology, Saint Petersburg State University, Saint Petersburg, 199034, Russia.
- Laboratory of Amyloid Biology, Saint Petersburg State University, Saint Petersburg, 199034, Russia
| |
Collapse
|
27
|
Qi X, Ma Y, Guan K, Zhao L, Ma Y, Wang R. Whey Protein Peptide Pro-Glu-Trp Ameliorates Hyperuricemia by Enhancing Intestinal Uric Acid Excretion, Modulating the Gut Microbiota, and Protecting the Intestinal Barrier in Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:2573-2584. [PMID: 38240209 DOI: 10.1021/acs.jafc.3c00984] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
Hyperuricemia (HUA) is a metabolic disorder characterized by an increase in the concentrations of uric acid (UA) in the bloodstream, intricately linked to the onset and progression of numerous chronic diseases. The tripeptide Pro-Glu-Trp (PEW) was identified as a xanthine oxidase (XOD) inhibitory peptide derived from whey protein, which was previously shown to mitigate HUA by suppressing UA synthesis and enhancing renal UA excretion. However, the effects of PEW on the intestinal UA excretion pathway remain unclear. This study investigated the impact of PEW on alleviating HUA in rats from the perspective of intestinal UA transport, gut microbiota, and intestinal barrier. The results indicated that PEW inhibited the XOD activity in the serum, jejunum, and ileum, ameliorated intestinal morphology changes and oxidative stress, and upregulated the expression of ABCG2 and GLUT9 in the small intestine. PEW reversed gut microbiota dysbiosis by decreasing the abundance of harmful bacteria (e.g., Bacteroides, Alloprevotella, and Desulfovibrio) and increasing the abundance of beneficial microbes (e.g., Muribaculaceae, Lactobacillus, and Ruminococcus) and elevated the concentration of short-chain fatty acids. PEW upregulated the expression of occludin and ZO-1 and decreased serum IL-1β, IL-6, and TNF-α levels. Our findings suggested that PEW supplementation ameliorated HUA by enhancing intestinal UA excretion, modulating the gut microbiota, and restoring the intestinal barrier function.
Collapse
Affiliation(s)
- Xiaofen Qi
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
- Engineering Research Center of Agricultural Microbiology Technology, Ministry of Education & Heilongjiang Provincial Key Laboratory of Ecological Restoration and Resource Utilization for Cold Region & Key Laboratory of Microbiology, College of Heilongjiang Province & School of Life Sciences, Heilongjiang University, Harbin 150080, China
| | - Yanfeng Ma
- Mengniu Hi-tech Dairy (Beijing) Co., Ltd., Beijing 101107, China
| | - Kaifang Guan
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Le Zhao
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Ying Ma
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Rongchun Wang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| |
Collapse
|
28
|
Nguyen JB, Marshall CW, Cook CN. The buzz within: the role of the gut microbiome in honeybee social behavior. J Exp Biol 2024; 227:jeb246400. [PMID: 38344873 DOI: 10.1242/jeb.246400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Abstract
Gut symbionts influence the physiology and behavior of their host, but the extent to which these effects scale to social behaviors is an emerging area of research. The use of the western honeybee (Apis mellifera) as a model enables researchers to investigate the gut microbiome and behavior at several levels of social organization. Insight into gut microbial effects at the societal level is critical for our understanding of how involved microbial symbionts are in host biology. In this Commentary, we discuss recent findings in honeybee gut microbiome research and synthesize these with knowledge of the physiology and behavior of other model organisms to hypothesize how host-microbe interactions at the individual level could shape societal dynamics and evolution.
Collapse
Affiliation(s)
- J B Nguyen
- Department of Biological Sciences, Marquette University, Milwaukee, WI 53233, USA
| | - C W Marshall
- Department of Biological Sciences, Marquette University, Milwaukee, WI 53233, USA
| | - C N Cook
- Department of Biological Sciences, Marquette University, Milwaukee, WI 53233, USA
| |
Collapse
|
29
|
Bar-Yoseph H, Krekhno Z, Cirstea M, Holani R, Moon KM, Foster LJ, Wieck M, Piper HG, Finlay BB. The Effect of Nutrient Deprivation on Early Life Small Intestinal Mucosal Microbiome and Host Proteome. J Nutr 2024; 154:412-423. [PMID: 38110179 DOI: 10.1016/j.tjnut.2023.12.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/04/2023] [Accepted: 12/13/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Nutrition plays a vital role in shaping the intestinal microbiome. However, many hospitalized children undergo periods of fasting during medical treatment. Changes to the small intestinal microbiota in early life in the setting of enteral deprivation have not been well described. OBJECTIVE The aim of this study was to investigate the impact of enteral deprivation on the small intestinal mucosal microbiome and to identify factors that shape this interaction in infancy. METHODS Intestinal biopsies were collected from proximal (fed) and distal (unfed) small bowel at the time of ostomy closure in children with a small intestinal enterostomy. Mucosal and luminal microbiome comparisons were performed including β-diversity and differential abundance and correlations with clinical factors were analyzed. Host proteomics were compared between fed and unfed samples and correlated with microbiome parameters. Finally, microbial results were validated in another cohort of pediatric patients. RESULTS Samples from 13 children (median age 84 d) were collected. Mucosal microbiome communities in the fed and unfed segments were strikingly similar [paired UniFrac distance (β-diversity)], whereas luminal effluent differed significantly from fed samples (PERMANOVA, P = 0.003). Multivariate analysis revealed patient as the strongest predictor of the UniFrac distance. Environmental variables did not influence the intrapatient microbial dissimilarity. Host proteomics were similar intrapatient (paired fed-unfed Euclidian distance) and showed a correlation with the UniFrac distance (Spearman rho = 0.71, P < 0.001). Specific proteins and functional clusters were significantly different between paired samples, including lipid metabolism and intracellular trafficking, whereas no difference was seen in innate immune proteins. The microbiome results were validated in a different cohort with similar characteristics. CONCLUSION We found the host to be the most dominant factor in the structure of the early life small intestinal mucosal microbiome. Nutrient deprivation was associated with specific changes in the host proteome. Further research is needed to better understand this host-microbe-nutrition interaction.
Collapse
Affiliation(s)
- Haggai Bar-Yoseph
- Department of Gastroenterology, Rambam Health Care Campus, Haifa, Israel; Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Zakhar Krekhno
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mihai Cirstea
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ravi Holani
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kyung-Mee Moon
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Leonard J Foster
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Minna Wieck
- Department of Surgery, UC Davis Medical Center, Sacramento, CA, United States
| | - Hannah G Piper
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada.
| | - B Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
30
|
Alvernaz SA, Wenzel ES, Nagelli U, Pezley LB, LaBomascus B, Gilbert JA, Maki PM, Tussing-Humphreys L, Peñalver Bernabé B. Inflammatory dietary potential is associated with vitamin depletion and gut microbial dysbiosis in early pregnancy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.12.02.23299325. [PMID: 38076865 PMCID: PMC10705629 DOI: 10.1101/2023.12.02.23299325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Background Pregnancy alters many physiological systems, including the maternal gut microbiota. Diet is a key regulator of this system and can alter the host immune system to promote inflammation. Multiple perinatal disorders have been associated with inflammation, maternal metabolic alterations, and gut microbial dysbiosis, including gestational diabetes mellitus, preeclampsia, preterm birth, and mood disorders. However, the effects of high inflammatory diets on the gut microbiota during pregnancy have yet to be fully explored. Objective To use a systems-based approach to characterize associations among dietary inflammatory potential, a measure of diet quality, and the gut microbiome during pregnancy. Methods Forty-nine pregnant persons were recruited prior to 16 weeks of gestation. Participants completed a food frequency questionnaire (FFQ) and provided fecal samples. Dietary inflammatory potential was assessed using the Dietary Inflammatory Index (DII) from FFQ data. Fecal samples were analyzed using 16S rRNA amplicon sequencing. Differential taxon abundance with respect to DII score were identified, and microbial metabolic potential was predicted using PICRUSt2. Results Inflammatory diets were associated with decreased vitamin and mineral intake and dysbiotic gut microbiota structure and predicted metabolism. Gut microbial compositional differences revealed a decrease in short chain fatty acid producers such as Faecalibacterium, and an increase in predicted vitamin B12 synthesis, methylglyoxal detoxification, galactose metabolism and multi drug efflux systems in pregnant individuals with increased DII scores. Conclusions Dietary inflammatory potential was associated with a reduction in the consumption of vitamins & minerals and predicted gut microbiota metabolic dysregulation.
Collapse
Affiliation(s)
- Suzanne A. Alvernaz
- Department of Biomedical Engineering, University of Illinois, Chicago, IL, USA
| | | | - Unnathi Nagelli
- Department of Biomedical Engineering, University of Illinois, Chicago, IL, USA
| | - Lacey B. Pezley
- Department of Kinesiology and Nutrition, University of Illinois, Chicago, IL, USA
| | - Bazil LaBomascus
- Department of Kinesiology and Nutrition, University of Illinois, Chicago, IL, USA
| | - Jack A. Gilbert
- Department of Pediatrics, University of California, San Diego, CA, USA
- Scripps Oceanographic Institute, University of California, San Diego, CA, USA
| | - Pauline M. Maki
- Department of Psychology, University of Illinois, Chicago, IL, USA
- Department of Psychiatry, University of Illinois, Chicago, IL, USA
- Department of Obstetrics and Gynecology, University of Illinois, Chicago, IL, USA
| | | | - Beatriz Peñalver Bernabé
- Department of Biomedical Engineering, University of Illinois, Chicago, IL, USA
- Center for Bioinformatics and Quantitative Biology, University of Illinois, Chicago, IL, USA
| |
Collapse
|
31
|
Moore B, Jolly J, Izumiyama M, Kawai E, Ravasi T, Ryu T. Tissue-specific transcriptional response of post-larval clownfish to ocean warming. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 908:168221. [PMID: 37923256 DOI: 10.1016/j.scitotenv.2023.168221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 10/24/2023] [Accepted: 10/28/2023] [Indexed: 11/07/2023]
Abstract
Anthropogenically driven climate change is predicted to increase average sea surface temperatures, as well as the frequency and intensity of marine heatwaves in the future. This increasing temperature is predicted to have a range of negative physiological impacts on multiple life-stages of coral reef fish. Nevertheless, studies of early-life stages remain limited, and tissue-specific transcriptomic studies of post-larval coral reef fish are yet to be conducted. Here, in an aquaria-based study we investigate the tissue-specific (brain, liver, muscle, and digestive tract) transcriptomic response of post-larval (20 dph) Amphiprion ocellaris to temperatures associated with future climate change (+3 °C). Additionally, we utilized metatranscriptomic sequencing to investigate how the microbiome of the digestive tract changes at +3 °C. Our results show that the transcriptional response to elevated temperatures is highly tissue-specific, as the number of differentially expressed genes (DEGs) and gene functions varied amongst the brain (102), liver (1785), digestive tract (380), and muscle (447). All tissues displayed DEGs associated with thermal stress, as 23 heat-shock protein genes were upregulated in all tissues. Our results indicate that post-larval clownfish may experience liver fibrosis-like symptoms at +3 °C as genes associated with extracellular matrix structure, oxidative stress, inflammation, glucose transport, and metabolism were all upregulated. We also observe a shift in the digestive tract microbiome community structure, as Vibrio sp. replace Escherichia coli as the dominant bacteria. This shift is coupled with the dysregulation of various genes involved in immune response in the digestive tract. Overall, this study highlights post-larval clownfish will display tissue-specific transcriptomic responses to future increases in temperature, with many potentially harmful pathways activated at +3 °C.
Collapse
Affiliation(s)
- Billy Moore
- Marine Climate Change Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa 904-0495, Japan
| | - Jeffrey Jolly
- Marine Climate Change Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa 904-0495, Japan
| | - Michael Izumiyama
- Marine Climate Change Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa 904-0495, Japan
| | - Erina Kawai
- Marine Climate Change Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa 904-0495, Japan
| | - Timothy Ravasi
- Marine Climate Change Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa 904-0495, Japan
| | - Taewoo Ryu
- Marine Climate Change Unit, Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa 904-0495, Japan.
| |
Collapse
|
32
|
Zhang Q, Gong J, Xiang H, Hu R, Yang X, Lv J, Zhang W, Liu M, Deng X, Yuan X, He Z, Jiang Y, Tan B, He J, Wu S. Effects of main active components of rosemary on growth performance, meat quality and lipid metabolism in finishing pigs. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2023; 15:341-349. [PMID: 38053801 PMCID: PMC10694069 DOI: 10.1016/j.aninu.2023.05.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 05/14/2023] [Accepted: 05/27/2023] [Indexed: 12/07/2023]
Abstract
Rosemary extracts have been widely used as feed additives in recent years. This study aimed to investigate the effects of rosmarinic acid (RA) and ursolic acid (UA), the main active components of rosemary, on growth performance, meat quality and lipid metabolism in finishing pigs. A total of 72 finishing pigs (Landrace; initial age of 150 d) were randomly divided into 3 treatments with 8 replicates of 3 pigs each, and fed a basal diet or diet containing 500 mg/kg of RA or UA. The results showed that dietary supplementation of RA or UA had no significant effect on the growth performance and carcass traits of finishing pigs (P > 0.05). However, both RA and UA significantly increased the triglyceride (TG) level in soleus muscle (P < 0.001). Supplementation of RA increased the expression of genes related to lipogenesis and transport including fatty acid synthase (FAS) (P < 0.001), sterol regulatory element binding protein-1c (SREBP1c) (P < 0.001) and peroxisome proliferator-activated receptor γ (PPARγ) (P < 0.05), while UA increased the expression of fatty acid transport protein 1 (FATP1), a gene related to lipid uptake (P < 0.05). However, RA reduced the expression of adipogenesis-related gene acetyl-coenzyme A carboxylase α (ACCα) (P < 0.01). Characterization of cecal microbiota indicated that RA increased the microbial richness (chao 1, P < 0.001) and diversity (observed species, P < 0.01). Further analysis of the genera revealed that RA increased the relative abundance of Bacteroides and g-UCG-005 (P < 0.05), and UA enriched Prevotella (P < 0.001). Correlation analysis showed that g-UCG-005 was positively correlated with the expression of FAS, carnitine palmitoyl transferase 1B (CPT1B), SREBP1c and PPARγ (P < 0.01). In conclusion, dietary supplementation of RA or UA may increase fat deposition in muscle of finishing pigs by regulating lipid metabolism and gut microbiota.
Collapse
Affiliation(s)
- Qianjin Zhang
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Jiatai Gong
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Hongkun Xiang
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Ruizhi Hu
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Xizi Yang
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Jing Lv
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Wentao Zhang
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Ming Liu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Xiong Deng
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Xupeng Yuan
- College of Animal Science and Technology, Hunan Biological and Electromechanical Polytechnic, Changsha 410127, China
| | - Ziyu He
- Department of Food Science and Biotechnology, Faculty of Agriculture, Kagoshima University, Kagoshima 890 - 0065, Japan
| | - Yixuan Jiang
- Hunan Delore Group Co. Ltd., Changsha 410131, China
| | - Bie Tan
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Jianhua He
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| | - Shusong Wu
- Hunan Collaborative Innovation Center for Utilization of Botanical Functional Ingredients, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| |
Collapse
|
33
|
Qi X, Guan K, Liu C, Chen H, Ma Y, Wang R. Whey protein peptides PEW and LLW synergistically ameliorate hyperuricemia and modulate gut microbiota in potassium oxonate and hypoxanthine-induced hyperuricemic rats. J Dairy Sci 2023; 106:7367-7381. [PMID: 37562644 DOI: 10.3168/jds.2023-23369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/29/2023] [Indexed: 08/12/2023]
Abstract
Pro-Glu-Trp (PEW) and Leu-Leu-Trp (LLW) are peptides derived from whey protein digestive products; both peptides exhibit xanthine oxidase inhibitory activity in vitro. However, it remains unclear whether these peptides can alleviate hyperuricemia (HUA) in vivo. In this study, we investigated the roles of PEW and LLW, both individually and in combination, in alleviating HUA induced by potassium oxonate and hypoxanthine. Together, PEW and LLW exhibited synergistic effects in reducing the serum levels of uric acid (UA), creatinine, and blood urea nitrogen, as well as increasing the fractional excretion of UA. The combined treatment with PEW and LLW inhibited UA synthesis, promoted UA excretion, and restored renal oxidative stress and mitochondrial damage. Moreover, the combined treatment alleviated dysbiosis of the gut microbiota, characterized by increased helpful microbial abundance, decreased harmful bacterial abundance, and increased production of short-chain fatty acids. Taken together, these results indicate that the combination of PEW and LLW mitigate HUA and kidney injury by rebalancing UA synthesis and excretion, modulating gut microbiota composition, and improving oxidative stress.
Collapse
Affiliation(s)
- Xiaofen Qi
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, Heilongjiang, China
| | - Kaifang Guan
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, Heilongjiang, China
| | - Chunhong Liu
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, Heilongjiang, China
| | - Haoran Chen
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, Heilongjiang, China
| | - Ying Ma
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, Heilongjiang, China.
| | - Rongchun Wang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, Heilongjiang, China.
| |
Collapse
|
34
|
Hayes MG, Langille MGI, Gu H. Cross-study analyses of microbial abundance using generalized common factor methods. BMC Bioinformatics 2023; 24:380. [PMID: 37807043 PMCID: PMC10561484 DOI: 10.1186/s12859-023-05509-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023] Open
Abstract
BACKGROUND By creating networks of biochemical pathways, communities of micro-organisms are able to modulate the properties of their environment and even the metabolic processes within their hosts. Next-generation high-throughput sequencing has led to a new frontier in microbial ecology, promising the ability to leverage the microbiome to make crucial advancements in the environmental and biomedical sciences. However, this is challenging, as genomic data are high-dimensional, sparse, and noisy. Much of this noise reflects the exact conditions under which sequencing took place, and is so significant that it limits consensus-based validation of study results. RESULTS We propose an ensemble approach for cross-study exploratory analyses of microbial abundance data in which we first estimate the variance-covariance matrix of the underlying abundances from each dataset on the log scale assuming Poisson sampling, and subsequently model these covariances jointly so as to find a shared low-dimensional subspace of the feature space. CONCLUSIONS By viewing the projection of the latent true abundances onto this common structure, the variation is pared down to that which is shared among all datasets, and is likely to reflect more generalizable biological signal than can be inferred from individual datasets. We investigate several ways of achieving this, demonstrate that they work well on simulated and real metagenomic data in terms of signal retention and interpretability, and recommend a particular implementation.
Collapse
Affiliation(s)
- Molly G Hayes
- Department of Mathematics and Statistics, Dalhousie University, Halifax, NS, Canada
| | - Morgan G I Langille
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Hong Gu
- Department of Mathematics and Statistics, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
35
|
Halma MTJ, Tuszynski JA, Marik PE. Cancer Metabolism as a Therapeutic Target and Review of Interventions. Nutrients 2023; 15:4245. [PMID: 37836529 PMCID: PMC10574675 DOI: 10.3390/nu15194245] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/20/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Cancer is amenable to low-cost treatments, given that it has a significant metabolic component, which can be affected through diet and lifestyle change at minimal cost. The Warburg hypothesis states that cancer cells have an altered cell metabolism towards anaerobic glycolysis. Given this metabolic reprogramming in cancer cells, it is possible to target cancers metabolically by depriving them of glucose. In addition to dietary and lifestyle modifications which work on tumors metabolically, there are a panoply of nutritional supplements and repurposed drugs associated with cancer prevention and better treatment outcomes. These interventions and their evidentiary basis are covered in the latter half of this review to guide future cancer treatment.
Collapse
Affiliation(s)
- Matthew T. J. Halma
- Department of Physics and Astronomy, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- EbMC Squared CIC, Bath BA2 4BL, UK
| | - Jack A. Tuszynski
- Department of Physics, University of Alberta, 11335 Saskatchewan Dr NW, Edmonton, AB T6G 2M9, Canada
- Department of Data Science and Engineering, The Silesian University of Technology, 44-100 Gliwice, Poland
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, I-1029 Turin, Italy
| | - Paul E. Marik
- Frontline COVID-19 Critical Care Alliance, Washington, DC 20036, USA
| |
Collapse
|
36
|
Dong R, Peng K, Shi L, Niu Q, Rafique H, Liu Y, Yuan L, Zou L, Li L, Messia MC, Hu X. Oat bran prevents high-fat-diet induced muscular dysfunction, systemic inflammation and oxidative stress through reconstructing gut microbiome and circulating metabolome. Food Res Int 2023; 172:113127. [PMID: 37689892 DOI: 10.1016/j.foodres.2023.113127] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 09/11/2023]
Abstract
Western-type diet characterized by high fat emerges a promoter of skeletal muscle dysfunctions. Oat bran was typically considered a healthy food of premium quality for its abundant dietary fiber. The present study comprehensively explored the effects of a diet rich in oat bran on skeletal muscle disfunctions in high-fat diet (HFD) fed mice. Dietary-fiber-rich oat bran significantly ameliorated HFD-induced skeletal muscle function abnormalities, as evidenced by a phenotype improvement in mice grip strength and endurance treadmill running distance, accompanied with the regulation of muscle functions related gene expressions, namely Fis1, Cytc, Mhy2 and Mhy4. Oat bran suppressed the production of systemic inflammatory cytokines while promoted superoxide dismutase and glutathione. Furthermore, oat bran significantly impacted gut microbiota composition by promoting short chain fatty acids (SCFAs) producers and certain probiotic genera, along with the enhancement of SCFAs. Oat bran also significantly decreased the circulating levels of inflammation-related metabolites and played roles in MAPK signaling, thereafter influencing skeletal muscle functions. Collectively, benefits from integration of biomedical indicators, microbiomics, and metabolomics demonstrates the benefits of oat bran consumption on prevention of HFD-related muscular dysfunctions via alleviating HFD-induced inflammation, gut dysbiosis, and systemic metabolism, pinpointing a novel mechanism underlying the muscle-promoting property of oat bran.
Collapse
Affiliation(s)
- Rui Dong
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Kejie Peng
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Lin Shi
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| | - Qianwen Niu
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Hamad Rafique
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yuan Liu
- Physical Education School, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Li Yuan
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Liang Zou
- School of Food and Biological Engineering, Chengdu University, Chengdu, Sichuan 610106, China
| | - Lu Li
- Guilin Seamild Foods Co., Ltd, Guilin, Guangxi 541004, China
| | - Maria Cristina Messia
- Department of Agricultural, Environmental and Food Sciences, University of Molise, Campobasso, Italy
| | - Xinzhong Hu
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| |
Collapse
|
37
|
Wang R, Halimulati M, Huang X, Ma Y, Li L, Zhang Z. Sulforaphane-driven reprogramming of gut microbiome and metabolome ameliorates the progression of hyperuricemia. J Adv Res 2023; 52:19-28. [PMID: 36371056 PMCID: PMC10555773 DOI: 10.1016/j.jare.2022.11.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 10/25/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022] Open
Abstract
INTRODUCTION Currently, revealing how to prevent and control hyperuricemia has become an essential public health issue. Sulforaphane hasawiderangeofapplications in the management of hyperuricemia. OBJECTIVE The study objective was to verify the uric acid-lowering effects and the regulation of the gut-kidney axis mediated by sulforaphane and identify host-microbial co-metabolites in hyperuricemia. METHODS A hyperuricemia model was established by administering feedstuffs with 4% potassium oxonate and 20% yeast. Forty male Sprague-Dawley rats were randomly divided into the normal control, hyperuricemia, allopurinol, and sulforaphane groups. Animals were treated by oral gavage for six consecutive weeks, and then phenotypic parameters, metabolomic profiling, and metagenomicsequencing were performed. RESULTS Sulforaphane could lower uric acid by decreasing urate synthesis and increasing renal urate excretion in hyperuricemic rats (P<0.05). We identified succinic acid and oxoglutaric acid as critical host-gut microbiome co-metabolites. Moreover, sulforaphane improved the diversity of microbial ecosystems and functions, as well as metabolic control of the kidney. Notably, sulforaphane exerted its renoprotective effect through epigenetic modification of Nrf2 and interaction between gut microbiota and epigenetic modification in hyperuricemic rats. CONCLUSION We revealed that sulforaphane could ameliorate the progression of hyperuricemia by reprogramming the gut microbiome and metabolome. Our findings may provide a good means for efficiently preventing and treating hyperuricemia.
Collapse
Affiliation(s)
- Ruoyu Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Haidian District, Beijing 100191, People's Republic of China
| | - Mairepaiti Halimulati
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Haidian District, Beijing 100191, People's Republic of China
| | - Xiaojie Huang
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Haidian District, Beijing 100191, People's Republic of China
| | - Yuxin Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Haidian District, Beijing 100191, People's Republic of China
| | - Lutong Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Haidian District, Beijing 100191, People's Republic of China
| | - Zhaofeng Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Haidian District, Beijing 100191, People's Republic of China; Beijing's Key Laboratory of Food Safety Toxicology Research and Evaluation, Beijing 100191, People's Republic of China.
| |
Collapse
|
38
|
Bishehsari F, Drees M, Adnan D, Sharma D, Green S, Koshy J, Giron LB, Goldman A, Abdel-Mohsen M, Rasmussen HE, Miller GE, Keshavarzian A. Multi-omics approach to socioeconomic disparity in metabolic syndrome reveals roles of diet and microbiome. Proteomics 2023; 23:e2300023. [PMID: 37525324 DOI: 10.1002/pmic.202300023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/23/2023] [Accepted: 07/10/2023] [Indexed: 08/02/2023]
Abstract
The epidemy of metabolic syndrome (MetS) is typically preceded by adoption of a "risky" lifestyle (e.g., dietary habit) among populations. Evidence shows that those with low socioeconomic status (SES) are at an increased risk for MetS. To investigate this, we recruited 123 obese subjects (body mass index [BMI] ≥ 30) from Chicago. Multi-omic data were collected to interrogate fecal microbiota, systemic markers of inflammation and immune activation, plasma metabolites, and plasma glycans. Intestinal permeability was measured using the sugar permeability testing. Our results suggest a heterogenous metabolic dysregulation among obese populations who are at risk of MetS. Systemic inflammation, linked to poor diet, intestinal microbiome dysbiosis, and gut barrier dysfunction may explain the development of MetS in these individuals. Our analysis revealed 37 key features associated with increased numbers of MetS features. These features were used to construct a composite metabolic-inflammatory (MI) score that was able to predict progression of MetS among at-risk individuals. The MI score was correlated with several markers of poor diet quality as well as lower levels of gut microbial diversity and abnormalities in several species of bacteria. This study reveals novel targets to reduce the burden of MetS and suggests access to healthy food options as a practical intervention.
Collapse
Affiliation(s)
- Faraz Bishehsari
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| | - Michael Drees
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| | - Darbaz Adnan
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| | - Deepak Sharma
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| | - Stefan Green
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| | - Jane Koshy
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Leila B Giron
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Aaron Goldman
- The Wistar Institute, Philadelphia, Pennsylvania, USA
| | | | | | - Gregory E Miller
- Institute for Policy Research and Dept of Psychology, Northwestern Univ, Evanston, Illinois, USA
| | - Ali Keshavarzian
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
39
|
Lee J, Wellenstein K, Rahnavard A, Nelson AT, Holter MM, Cummings B, Yeliseyev V, Castoldi A, Clish CB, Bry L, Siegel D, Kahn BB. Beneficial metabolic effects of PAHSAs depend on the gut microbiota in diet-induced obese mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.28.558803. [PMID: 37808673 PMCID: PMC10557726 DOI: 10.1101/2023.09.28.558803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Dietary lipids play an essential role in regulating the function of the gut microbiota and gastrointestinal tract, and these luminal interactions contribute to mediating host metabolism. PAHSAs are a class of lipids with anti-diabetic and anti-inflammatory properties, but whether the gut microbiota contributes to their beneficial effects on host metabolism is unknown. Here, we report that treating high fat diet (HFD)-fed germ-free mice with PAHSAs does not improve insulin sensitivity. However, transfer of feces from PAHSA-treated, but not Vehicle-treated, chow-fed mice increases insulin-sensitivity in HFD-fed germ free mice. Thus, the gut microbiota is necessary for and can transmit the insulin-sensitizing effects of PAHSAs in HFD-fed germ-free mice. Functional analyses of the cecal metagenome and lipidome of PAHSA-treated mice identified multiple lipid species that associate with the gut commensal Bacteroides thetaiotaomicron ( Bt ) and with insulin sensitivity resulting from PAHSA treatment. Bt supplementation in HFD-fed female mice prevented weight gain, reduced adiposity, improved glucose tolerance, fortified the colonic mucus barrier and reduced systemic inflammation versus chow-fed controls, effects that were not observed in HFD-fed male mice. Furthermore, ovariectomy partially reversed the beneficial Bt effects on host metabolism, indicating a role for sex hormones in mediating probiotic effects. Altogether, these studies highlight the fact that lipids can modulate the gut microbiota resulting in improvement in host metabolism and that PAHSA-induced changes in the microbiota result in at least some of their insulin-sensitizing effects in female mice.
Collapse
|
40
|
Tisza M, Lloyd R, Hoffman K, Smith D, Rewers M, Cregeen SJ, Petrosino JF. Phage-bacteria dynamics during the first years of life revealed by trans-kingdom marker gene analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.28.559994. [PMID: 37808738 PMCID: PMC10557657 DOI: 10.1101/2023.09.28.559994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Humans are colonized with commensal bacteria soon after birth, and, while this colonization is affected by lifestyle and other factors, bacterial colonization proceeds through well-studied phases. However, less is known about phage communities in early human development due to small study sizes, inability to leverage large databases, and lack of appropriate bioinformatics tools. In this study, whole genome shotgun sequencing data from the TEDDY study, composed of 12,262 longitudinal samples from 887 children in 4 countries, is reanalyzed to assess phage and bacterial dynamics simultaneously. Reads from these samples were mapped to marker genes from both bacteria and a new database of tens of thousands of phage taxa from human microbiomes. We uncover that each child is colonized by hundreds of different phages during the early years, and phages are more transitory than bacteria. Participants' samples continually harbor new phage species over time whereas the diversification of bacterial species begins to saturate. Phage data improves the ability for machine learning models to discriminate samples by country. Finally, while phage populations were individual-specific, striking patterns arose from the larger dataset, showing clear trends of ecological succession amongst phages, which correlated well with putative host bacteria. Improved understanding of phage-bacterial relationships may reveal new means by which to shape and modulate the microbiome and its constituents to improve health and reduce disease, particularly in vulnerable populations where antibiotic use and/or other more drastic measures may not be advised.
Collapse
Affiliation(s)
- Michael Tisza
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Richard Lloyd
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Kristi Hoffman
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Daniel Smith
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Marian Rewers
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, USA
| | - Sara Javornik Cregeen
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Joseph F Petrosino
- The Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
41
|
Wan J, Song J, Lv Q, Zhang H, Xiang Q, Dai H, Zheng H, Lin X, Zhang W. Alterations in the Gut Microbiome of Young Children with Airway Allergic Disease Revealed by Next-Generation Sequencing. J Asthma Allergy 2023; 16:961-972. [PMID: 37700874 PMCID: PMC10494927 DOI: 10.2147/jaa.s422537] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/17/2023] [Indexed: 09/14/2023] Open
Abstract
Purpose Recent studies had shown that gut microbiota played a significant role in the development of the immune system and may affect the course of airway allergic disease. We conducted this study to determine unique gut microbial associated with allergic disease in children by shotgun gene sequencing. Methods We collected fecal samples from children with allergic asthma (n = 23) and allergic rhinitis (n = 18), and healthy control (n = 19). The gut microbiota of specimens was analyzed by high-throughput metagenomic shotgun gene sequencing. Results The intestinal microbiota of children with allergic asthma and allergic rhinitis was characterized by increased microbial richness and diversity. Simpson and Shannon were significantly elevated in children with allergic asthma. Principal coordinates analysis (PCoA) showed that the gut microbial communities cluster patterns of children with asthma or rhinitis were significantly different from those of healthy controls. However, no significant difference was found between asthma group and rhinitis group At the phylum level, higher relative abundance of Firmicutes was found in the allergic rhinitis group and allergic asthma group, while the level of Bacteroidetes was significantly lower. At the genus level, Corynebacterium, Streptococcus, Dorea, Actinomyces, Bifidobacterium, Blautia, and Rothia were significantly enriched in the allergic asthma group. Finally, a random forest classifier model selected 16 general signatures to discriminate the allergic asthma group from the healthy control group. Conclusion In conclusion, children in the allergic rhinitis group and allergic asthma group had altered gut microbiomes in comparison with the healthy control group. Compared to healthy children, the gut microbiome in children with allergic diseases has higher pro-inflammatory potential and increased production of pro-inflammatory molecules.
Collapse
Affiliation(s)
- Jinyi Wan
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China
- Department of Pediatric Internal Medicine, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, 317000, People’s Republic of China
| | - Jingjing Song
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China
| | - Qingqing Lv
- Department of Pediatric Internal Medicine, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, 317000, People’s Republic of China
| | - Hui Zhang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China
| | - Qiangwei Xiang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China
| | - Huan Dai
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China
| | - Hang Zheng
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China
| | - Xixi Lin
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China
| | - Weixi Zhang
- Department of Pediatric Allergy and Immunology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, 325027, People’s Republic of China
| |
Collapse
|
42
|
Zhang L, Liu X, Zhang C. Effect of PET microplastics on the growth, digestive enzymes, and intestinal flora of the sea cucumber Apostichopus japonicus. MARINE ENVIRONMENTAL RESEARCH 2023; 190:106125. [PMID: 37552920 DOI: 10.1016/j.marenvres.2023.106125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 07/16/2023] [Accepted: 08/03/2023] [Indexed: 08/10/2023]
Abstract
Marine microplastic (MP) pollution is becoming a serious problem and their potentially toxic effects on marine organisms have attracted much attention. Sea cucumber is very important for the safety and health of marine ecosystems. However, there have been relatively few studies on the effects of microplastic pollution on sea cucumbers at environmentally-related concentrations and under controlled conditions. Therefore, this study evaluated the effects of polyethylene terephthalate (PET) microplastics (particle sizes: 0.5-45 μm, 2-200 μm, and 20-300 μm; and three concentration levels for each particle size, approximately 103, 104, and 105 particles/kg) on the basic biological indicators, intestinal digestive enzymes, and intestinal flora of Apostichopus japonicus after a 28-day feeding experiment. This study showed that environmentally-related and high concentrations of microplastics had little effect on A. japonicus. This study provides valuable reference information about the effects of marine microplastic pollution on sea cucumbers.
Collapse
Affiliation(s)
- Libin Zhang
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China; Center for Ocean Mega-Sciences, Chinese Academy of Sciences, Qingdao, 266071, China; CAS Engineering Laboratory for Marine Ranching, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Shandong Province Key Laboratory of Experimental Marine Biology, Qingdao, 266071, China.
| | - Xiang Liu
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China; Center for Ocean Mega-Sciences, Chinese Academy of Sciences, Qingdao, 266071, China; CAS Engineering Laboratory for Marine Ranching, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Shandong Province Key Laboratory of Experimental Marine Biology, Qingdao, 266071, China; College of Environment and Safety Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China
| | - Chenxi Zhang
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, China; Center for Ocean Mega-Sciences, Chinese Academy of Sciences, Qingdao, 266071, China; CAS Engineering Laboratory for Marine Ranching, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Shandong Province Key Laboratory of Experimental Marine Biology, Qingdao, 266071, China; College of Environment and Safety Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China
| |
Collapse
|
43
|
Xie G, Hu Q, Cao X, Wu W, Dai P, Guo W, Wang O, Wei L, Ren R, Li Y. Clinical identification and microbiota analysis of Chlamydia psittaci- and Chlamydia abortus- pneumonia by metagenomic next-generation sequencing. Front Cell Infect Microbiol 2023; 13:1157540. [PMID: 37434780 PMCID: PMC10331293 DOI: 10.3389/fcimb.2023.1157540] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/29/2023] [Indexed: 07/13/2023] Open
Abstract
Introduction Recently, the incidence of chlamydial pneumonia caused by rare pathogens such as C. psittaci or C. abortus has shown a significant upward trend. The non-specific clinical manifestations and the limitations of traditional pathogen identification methods determine that chlamydial pneumonia is likely to be poorly diagnosed or even misdiagnosed, and may further result in delayed treatment or unnecessary antibiotic use. mNGS's non-preference and high sensitivity give us the opportunity to obtain more sensitive detection results than traditional methods for rare pathogens such as C. psittaci or C. abortus. Methods In the present study, we investigated both the pathogenic profile characteristics and the lower respiratory tract microbiota of pneumonia patients with different chlamydial infection patterns using mNGS. Results More co-infecting pathogens were found to be detectable in clinical samples from patients infected with C. psittaci compared to C. abortus, suggesting that patients infected with C. psittaci may have a higher risk of mixed infection, which in turn leads to more severe clinical symptoms and a longer disease course cycle. Further, we also used mNGS data to analyze for the first time the characteristic differences in the lower respiratory tract microbiota of patients with and without chlamydial pneumonia, the impact of the pattern of Chlamydia infection on the lower respiratory tract microbiota, and the clinical relevance of these characteristics. Significantly different profiles of lower respiratory tract microbiota and microecological diversity were found among different clinical subgroups, and in particular, mixed infections with C. psittaci and C. abortus resulted in lower lung microbiota diversity, suggesting that chlamydial infections shape the unique lung microbiota pathology, while mixed infections with different Chlamydia may have important effects on the composition and diversity of the lung microbiota. Discussion The present study provides possible evidences supporting the close correlation between chlamydial infection, altered microbial diversity in patients' lungs and clinical parameters associated with infection or inflammation in patients, which also provides a new research direction to better understand the pathogenic mechanisms of pulmonary infections caused by Chlamydia.
Collapse
Affiliation(s)
- Gongxun Xie
- Department of Pathology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Qing Hu
- Department of Pathology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Xuefang Cao
- Institute of Innovative Applications, MatriDx Biotechnology Co., Ltd, Hangzhou, Zhejiang, China
| | - Wenjie Wu
- Institute of Innovative Applications, MatriDx Biotechnology Co., Ltd, Hangzhou, Zhejiang, China
| | - Penghui Dai
- Department of Pathology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Wei Guo
- Department of Pathology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Ouxi Wang
- Institute of Innovative Applications, MatriDx Biotechnology Co., Ltd, Hangzhou, Zhejiang, China
| | - Liang Wei
- Institute of Innovative Applications, MatriDx Biotechnology Co., Ltd, Hangzhou, Zhejiang, China
| | - Ruotong Ren
- Institute of Innovative Applications, MatriDx Biotechnology Co., Ltd, Hangzhou, Zhejiang, China
- Foshan Branch, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yanchun Li
- Department of Pathology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
44
|
Rusch JA, Layden BT, Dugas LR. Signalling cognition: the gut microbiota and hypothalamic-pituitary-adrenal axis. Front Endocrinol (Lausanne) 2023; 14:1130689. [PMID: 37404311 PMCID: PMC10316519 DOI: 10.3389/fendo.2023.1130689] [Citation(s) in RCA: 92] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 05/25/2023] [Indexed: 07/06/2023] Open
Abstract
Cognitive function in humans depends on the complex and interplay between multiple body systems, including the hypothalamic-pituitary-adrenal (HPA) axis. The gut microbiota, which vastly outnumbers human cells and has a genetic potential that exceeds that of the human genome, plays a crucial role in this interplay. The microbiota-gut-brain (MGB) axis is a bidirectional signalling pathway that operates through neural, endocrine, immune, and metabolic pathways. One of the major neuroendocrine systems responding to stress is the HPA axis which produces glucocorticoids such as cortisol in humans and corticosterone in rodents. Appropriate concentrations of cortisol are essential for normal neurodevelopment and function, as well as cognitive processes such as learning and memory, and studies have shown that microbes modulate the HPA axis throughout life. Stress can significantly impact the MGB axis via the HPA axis and other pathways. Animal research has advanced our understanding of these mechanisms and pathways, leading to a paradigm shift in conceptual thinking about the influence of the microbiota on human health and disease. Preclinical and human trials are currently underway to determine how these animal models translate to humans. In this review article, we summarize the current knowledge of the relationship between the gut microbiota, HPA axis, and cognition, and provide an overview of the main findings and conclusions in this broad field.
Collapse
Affiliation(s)
- Jody A. Rusch
- Division of Chemical Pathology, Department of Pathology, University of Cape Town, Cape Town, South Africa
- C17 Chemical Pathology Laboratory, Groote Schuur Hospital, National Health Laboratory Service, Cape Town, South Africa
| | - Brian T. Layden
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, United States
| | - Lara R. Dugas
- Division of Epidemiology and Biostatistics, School of Public Health, University of Cape Town, Cape Town, South Africa
- Public Health Sciences, Parkinson School of Health Sciences and Public Health, Loyola University Chicago, Maywood, IL, United States
| |
Collapse
|
45
|
Liang L, Meng Z, Zhang F, Jianguo Z, Fang S, Hu Q, Tang X, Li Y. Lactobacillus gasseri LG08 and Leuconostoc mesenteroides LM58 exert preventive effect on the development of hyperuricemia by repairing antioxidant system and intestinal flora balance. Front Microbiol 2023; 14:1211831. [PMID: 37378287 PMCID: PMC10291327 DOI: 10.3389/fmicb.2023.1211831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Introduction Currently, hyperuricemia has shown a surprisingly rising trend, which attracts widespread attention due to potentially major health risks. Considering the inevitable side effects of long-term medicine, probiotics are emerging as potential therapeutics due to their ability to improve uric acid metabolism and superior safety. Methods In our study, two strains of probiotics, Lactobacillus gasseri LG08 (LG08) and Leuconostoc mesenteroides LM58 (LM58) isolated from kimchi were evaluated for the prebiotic properties in vitro and uric-lowering effects in vivo. Here, hyperuricemia animal model and 16S rRNA gene amplicons analysis were further studied to investigate whether these probiotics exert different effects in prevention and treatment. Results In vivo indicators and intestinal flora immunity revealed that both LG08 and LM58 significantly prevent the development and progression of hyperuricemia, repair the antioxidant system and maintain intestinal flora balance in healthy rats, especially LM58. After hyperuricemia was formed, although the effect of LG08 and LM58 could decrease the level of uric acid, the effect to reverse and repair antioxidant levels in the body was limited. Discussion In our study, these findings have important implications for hyperuricemia prevention and therapy, and provided more mechanistic insights into the effect of probiotics in hyperuricemia.
Collapse
Affiliation(s)
- Lizhen Liang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Zihui Meng
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| | - Fei Zhang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhu Jianguo
- Department of Research and Development, Wecare-Bio Probiotics Co., Ltd., Suzhou, China
| | - Shuguang Fang
- Department of Research and Development, Wecare-Bio Probiotics Co., Ltd., Suzhou, China
| | - Qingang Hu
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xuna Tang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yanan Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, China
| |
Collapse
|
46
|
Xie Z, Li Y, Xiong K, Tu Z, Waiho K, Yang C, Deng Y, Li S, K H Fang J, Hu M, Dupont S, Wang Y. Combined effect of salinity and hypoxia on digestive enzymes and intestinal microbiota in the oyster Crassostrea hongkongensis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 331:121921. [PMID: 37263564 DOI: 10.1016/j.envpol.2023.121921] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 05/26/2023] [Accepted: 05/27/2023] [Indexed: 06/03/2023]
Abstract
Anthropologic activities caused frequent eutrophication in coastal and estuarine waters, resulting in diel-cycling hypoxia. Given global climate change, extreme weather events often occur, thus salinity fluctuation frequently breaks out in these waters. This study aimed to evaluate the combined effects of salinity and hypoxia on intestinal microbiota and digestive enzymes of Crassostrea hongkongensis. Specifically, we sequenced 16 S rRNA of intestinal microbiota and measured the digestive enzymes trypsin (TRS), lipase (LPS) and amylase (AMY) in oysters exposed for 28 days to three salinities (10, 25 and 35) and two dissolved oxygen conditions, normoxia (6 mg/L) and hypoxia (6 mg/L for 12 h, 2 mg/L for 12 h). Oysters in normoxia and salinity of 25 were treated as control. After 28-day exposure, for microbial components, Fusobacteriota, Firmicutes, Bacteroidota, Proteobacteria and Actinobacteriota comprised the majority for all experimental groups. Compared with the control group, the diversity and structure of intestinal microbiota tended to change in all treated groups. The species richness in C. hongkongensis intestine also changed. It was the most significant that high salinity increased Proteobacteria proportion while low salinity and hypoxia increased Fusobacteriota but decreased Proteobacteria, respectively. Additionally, Actinobacteriota was sensitive and changed under environmental stressor (P < 0.01). The prediction results on intestinal microbiota showed that, all functions of oysters were up-regulated to distinct degrees under low/high salinity with hypoxia. According to the KEGG prediction, cellular processes were more active and energy metabolism upregulated, indicating the adaptation of C. hongkongensis to environmental change. Periodical hypoxia and low/high salinity had complex effect on the digestive enzymes, in which the activity of TRS and LPS decreased while AMY increased. High/low salinity and periodical hypoxia can change the secretion of digestive enzymes and influence intestinal microbial diversity and species richness of C. hongkongensis, deducing the chronic adverse effects on the digestive physiology in long-term exposure.
Collapse
Affiliation(s)
- Zhe Xie
- International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, Shanghai, 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Yuting Li
- International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, Shanghai, 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Kai Xiong
- International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, Shanghai, 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Zhihan Tu
- International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, Shanghai, 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Khor Waiho
- Higher Institution Centre of Excellence (HICoE), Institute of Tropical Aquaculture and Fisheries, Universiti Malaysia Terengganu, Terengganu, 21030, Malaysia
| | - Chuangye Yang
- Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Yuewen Deng
- Fisheries College, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Saishuai Li
- International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, Shanghai, 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - James K H Fang
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| | - Menghong Hu
- International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, Shanghai, 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Sam Dupont
- Department of Biological & Environmental Sciences, University of Gothenburg, 45178, Fiskebäckskil, Sweden; International Atomic Energy Agency, Environment Laboratories, 98000, Principality of Monaco, Monaco
| | - Youji Wang
- International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, Shanghai, 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China.
| |
Collapse
|
47
|
Luqman A. The orchestra of human bacteriome by hormones. Microb Pathog 2023; 180:106125. [PMID: 37119938 DOI: 10.1016/j.micpath.2023.106125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/07/2023] [Accepted: 04/24/2023] [Indexed: 05/01/2023]
Abstract
Human microbiome interact reciprocally with the host. Recent findings showed the capability of microorganisms to response towards host signaling molecules, such as hormones. Studies confirmed the complex response of bacteria in response to hormones exposure. These hormones impact many aspects on bacteria, such as the growth, metabolism, and virulence. The effects of each hormone seem to be species-specific. The most studied hormones are cathecolamines also known as stress hormones that consists of epinephrine, norepinephrine and dopamine. These hormones affect the growth of bacteria either inhibit or enhance by acting like a siderophore. Epinephrine and norepinephrine have also been reported to activate QseBC, a quorum sensing in Gram-negative bacteria and eventually enhances the virulence of pathogens. Other hormones were also reported to play a role in shaping human microbiome composition and affect their behavior. Considering the complex response of bacteria on hormones, it highlights the necessity to take the impact of hormones on bacteria into account in studying human health in relation to human microbiome.
Collapse
Affiliation(s)
- Arif Luqman
- Biology Department, Institut Teknologi Sepuluh Nopember, Surabaya, Indonesia.
| |
Collapse
|
48
|
Chen C, Liu L, Zhong Y, Wang M, Ai Y, Hou Y, Chen H, Lin X, Zhang Y, Ding M, Luo T, Li J, Li X, Xiao X. Gut microbiota-bile acids-glucagon like peptide-1 axis contributes the resistance to high fat diet-induced obesity in mice. J Nutr Biochem 2023; 117:109358. [PMID: 37085058 DOI: 10.1016/j.jnutbio.2023.109358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 04/03/2023] [Accepted: 04/14/2023] [Indexed: 04/23/2023]
Abstract
In human and rodents, some individuals may remain lean even when they are challenged with high calorie intake. The underlying mechanism for resistance to diet-induced obesity was poorly understood. Here, we used C57BL/6J mice to establish animal models of high-fat diet (HFD) induced obesity sensitive (DIO) mice and obesity resistant (DIR) mice. We then investigated the role of gut microbiota, bile acids (BAs) and brown adipose tissue (BAT) thermogenesis in the development of DIR. Reduced fat accumulation, increased glucose tolerance and energy expenditure through BAT activation were observed in DIR mice. The plasma BAs of DIR mice especially the unconjugated BAs were significantly decreased, while intestine tauro-conjugated bile acids (T-CA, T-β-MCA, T-ω-MCA and T-UDCA) were significantly increased in DIR mice. The composition of the gut flora also changed drastically, and negative correlation was found between metabolic profiles (plasma TG, TC, LDL and body weight) and the abundance of Ruminiclostridium in DIR mice, while genus Anaerotruncus abundance in DOR mice was found to be positively correlated. After fecal microbiota transplants, HFD fed recipient mice exhibited a trend toward reduced adiposity and improved glucose tolerance, while showing increased serum tauro-conjugated BAs levels. STC-1 cell experiments confirmed tauro-conjugated BA (T-β-MCA) activated FXR/TGR5 pathway and induced the production of GLP-1, inhibiting genes that regulate the ceramide synthesis. Our results indicated that the DIR mice exhibited higher energy expenditure by activating BAT thermogenesis, which may be related altered gut microbiota-bile acids-glucagon like peptide-1 axis.
Collapse
Affiliation(s)
- Chunxiu Chen
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Lingli Liu
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Ying Zhong
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Miaoran Wang
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Yanbiao Ai
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Yi Hou
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Experimental Teaching & Management Center, Chongqing Medical University, Chongqing 401331, China
| | - Hong Chen
- Key Laboratory of Laboratory Medical Diagnosis, Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xiaojing Lin
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yunqi Zhang
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Min Ding
- Key Laboratory of Laboratory Medical Diagnosis, Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Ting Luo
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jibin Li
- Department of Nutrition and Food Hygiene, School of Public Health, Chongqing Medical University, Chongqing 400016, China.
| | - Xinyu Li
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Pharmacy, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Xiaoqiu Xiao
- Department of Endocrinology, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
49
|
Cao Y, Nguyen LH, Tica S, Otegbeye E, Zong X, Roelstraete B, Chan AT, Warner BB, Stephansson O, Ludvigsson JF. Evaluation of Birth by Cesarean Delivery and Development of Early-Onset Colorectal Cancer. JAMA Netw Open 2023; 6:e2310316. [PMID: 37103933 PMCID: PMC10140807 DOI: 10.1001/jamanetworkopen.2023.10316] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/13/2023] [Indexed: 04/28/2023] Open
Abstract
IMPORTANCE The incidence of early-onset colorectal cancer (CRC), diagnosed younger than 50 years of age, has increased worldwide. Gut dysbiosis throughout the life course is hypothesized as a leading mechanism, yet epidemiologic data are limited. OBJECTIVE To prospectively examine the association between birth by cesarean delivery and early-onset CRC among offspring. DESIGN, SETTING, AND PARTICIPANTS In this population-based, nationwide case-control study in Sweden, adults diagnosed with CRC between 18 and 49 years of age from 1991 to 2017 were identified through the Epidemiology Strengthened by Histopathology Reports in Sweden (ESPRESSO) cohort. Up to 5 general population control individuals without CRC were matched with each case on age, sex, calendar year, and county of residence. Pathology-confirmed end points were linked with the Swedish Medical Birth Register and other national registers. Analyses were conducted from March 2022 through March 2023. EXPOSURE Birth by cesarean delivery. MAIN OUTCOMES AND MEASURES The primary outcome was development of early-onset CRC in the overall population and by sex. RESULTS We identified 564 case patients with incident early-onset CRC (mean [SD] age, 32.9 [6.2] years; 284 [50.4%] male) and 2180 matched controls (mean [SD] age, 32.7 [6.3] years; 1104 [50.6%] male). Compared with vaginal delivery, birth by cesarean delivery was not associated with early-onset CRC in the overall population (adjusted odds ratio [aOR], 1.28; 95% CI, 0.91-1.79) after multivariable adjustment for matching and maternal and pregnancy-related factors. A positive association was found for females (aOR, 1.62; 95% CI, 1.01-2.60), but there was no association for males (aOR, 1.05; 95% CI, 0.64-1.72). CONCLUSIONS AND RELEVANCE In this nationwide, population-based case-control study, birth by cesarean delivery was not associated with early-onset CRC compared with birth by vaginal delivery in the overall population in Sweden. However, females born by cesarean delivery had greater odds of early-onset CRC compared with individuals born through vaginal delivery. This finding suggests that early-life gut dysbiosis may contribute to early-onset CRC in females.
Collapse
Affiliation(s)
- Yin Cao
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St Louis, Missouri
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Long H. Nguyen
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Stefani Tica
- Division of Pediatric Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri
| | - Ebunoluwa Otegbeye
- Department of Surgery, Washington University School of Medicine, St Louis, Missouri
| | - Xiaoyu Zong
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St Louis, Missouri
| | - Bjorn Roelstraete
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Andrew T. Chan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Barbara B. Warner
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri
| | - Olof Stephansson
- Clinical Epidemiology Division, Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden
- Division of Women’s Health, Department of Obstetrics, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas F. Ludvigsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Paediatrics, Örebro University Hospital, Örebro, Sweden
- Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, New York
| |
Collapse
|
50
|
Feng R, Zhu Q, Li Q, Zhai Y, Wang J, Qin C, Liang D, Zhang R, Tian H, Liu H, Chen Y, Fu Y, Wang X, Ding X. Microbiota-ear-brain interaction is associated with generalized anxiety disorder through activation of inflammatory cytokine responses. Front Immunol 2023; 14:1117726. [PMID: 36969214 PMCID: PMC10033601 DOI: 10.3389/fimmu.2023.1117726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/03/2023] [Indexed: 03/12/2023] Open
Abstract
IntroductionGeneralized anxiety disorder (GAD) is one of the most enduring anxiety disorders, being associated with increased systemic inflammation. However, the trigger and mechanisms underlying the activation of inflammatory cytokine responses in GAD remain poorly understood.Materials and methodsWe characterized the ear canal microbiome in GAD patients through 16S rRNA gene sequencing and metagenomic sequencing and identified the serum inflammatory markers in GAD patients. Spearman correlations were applied to test the relationship between the microbiota changes and systemic inflammation.ResultsOur findings showed the higher microbial diversity, accompanied with the significantly increased abundance of Proteobacteria, and decreased abundance of Firmicutes in the ear canal of GAD participants compared to that of the age- and sex-matched healthy controls (HC). Metagenomic sequencing showed that Pseudomonas aeruginosa were significantly increased at species-level in GAD patients. Furthermore, we observed the relative abundance of Pseudomonas aeruginosa was positively associated with elevated systemic inflammatory markers and the severity of disease, suggesting that these ear canal microbiota alterations might be correlated with GAD by activating the inflammatory response.ConclusionsThese findings indicate that microbiota-ear-brain interaction via upregulating inflammatory reaction involve in the development of GAD, as well as suggest that ear canal bacterial communities may be a target for therapeutic intervention.
Collapse
Affiliation(s)
- Renyi Feng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
| | - Qingyong Zhu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
| | - Qingchen Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
| | - Yanping Zhai
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
| | - Jiuqi Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
| | - Chi Qin
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
| | - Dongxiao Liang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
| | - Rui Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
| | - Haiyan Tian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
| | - Han Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
| | - Yongkang Chen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
| | - Yu Fu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
| | - Xuejing Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
- *Correspondence: Xuebing Ding, ; Xuejing Wang,
| | - Xuebing Ding
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Parkinson and Movement Disorder, Zhengzhou University, Zhengzhou, Henan, China
- *Correspondence: Xuebing Ding, ; Xuejing Wang,
| |
Collapse
|